You are on page 1of 780

Kidney

Transplantation
Principles and Practice
EIGHTH EDITION

Stuart J. Knechtle, MD, FACS


William R. Kenan, Jr. Professor of Surgery
Executive Director, Duke Transplant Center
Duke University School of Medicine
Durham, North Carolina, USA

Lorna P. Marson, MBBS, MD, FRCSEng, FRCSEd, FRCP(Ed)


Professor of Transplant Surgery
Clinical Sciences (Surgery)
University of Edinburgh
Edinburgh, United Kingdom

Sir Peter J. Morris, MD, PhD, FRS, FRCS


Emeritus Nuffield Professor of Surgery
University of Oxford;
Honorary Professor
University of London;
Nuffield Department of Surgical Sciences
University of Oxford
Oxford, United Kingdom
1600 John F. Kennedy Blvd.
Ste 1800
Philadelphia, PA 19103-2899

KIDNEY TRANSPLANTATION: PRINCIPLES AND PRACTICE,


EIGHTH EDITION ISBN: 978-0-323-53186-3
Copyright © 2020 by Elsevier, Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about the
Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance
Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

Notice

Practitioners and researchers must always rely on their own experience and knowledge in evaluating and
using any information, methods, compounds or experiments described herein. Because of rapid advances
in the medical sciences, in particular, independent verification of diagnoses and drug dosages should be
made. To the fullest extent of the law, no responsibility is assumed by Elsevier, authors, editors or con-
tributors for any injury and/or damage to persons or property as a matter of products liability, negligence
or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in
the material herein.

Previous editions copyrighted 2014, 2008, 2001, 1994, 1988, 1984 and 1979.

Library of Congress Control Number: 2019948029

Content Strategist: Russell Gabbedy/Jessica L. McCool


Content Development Specialist: Meghan Andress
Publishing Services Manager: Deepthi Unni
Project Manager: Beula Christopher
Design Direction: Christian J. Bilbow

Printed in China

Last digit is the print number: 9 8 7 6 5 4 3 2 1


Video Contents

CHAPTER 2 CHAPTER 17
2.1 Introduction 17.1 Calcineurin inhibitor mechanism of action
animation
2.2 Formation of an immune synapse MATTHEW L. HOLZNER, VIKRAM WADHERA, AMIT BASU,
DAN DAVIS SANDER FLORMAN, and RON SHAPIRO

2.3 3-D rotational image of immune synapses CHAPTER 36


FIONA J. CULLEY
36.1 Real-time ultrasound guided pancreas allograft
2.4 An NK cell killing its target biopsy
DAN DAVIS TALAL M. AL-QAOUD, DIXON B. KAUFMAN, PETER J. FRIEND, and
JON S. ­ODORICO
CHAPTER 5
CHAPTER 42
5.1 Laparoscopic: peritoneal dialysis - catheter
insertion 42.1 The transplant library on OvidSP
ADAM D. BARLOW, JAMES P. HUNTER, and MICHAEL L. NICHOLSON LISET H. M. PENGEL

CHAPTER 6 42.2 The transplant library on Evidentia


LISET H. M. PENGEL
6.1 Brain death examination
LAURA S. JOHNSON, NICHOLAS BYRON PITTS, and
RAM M. SUBRAMANIAN

CHAPTER 8
8.1 Laparoendoscopic single site (LESS) donor
nephrectomy: technique and outcomes
ROLF N. BARTH

vii
Preface to the First Edition
Renal transplantation is now an accepted treatment of The recognition that allogeneic tissues would be rejected
patients in end-stage renal failure. A successful trans- was further established in later years by Drs. Gibson and
plant restores not merely life but an acceptable quality of Medawar, who treated burn patients with homografts in
life to such patients. The number of patients in endstage Glasgow during the Second World War. Indeed, it was the
renal failure in the Western World who might be treated crash of a bomber behind the Medawars’ house in Oxford
by hemodialysis and transplantation is considerable and during the early years of the war that first stimulated his
comprises some 30-50 new patients/million of popula- interest in transplantation, especially of skin.
tion. Unfortunately in most, if not all, countries the sup- In his address at the opening of the new Oxford Trans-
ply of kidneys for transplantation is insufficient to meet the plant Unit in 1977, Sir Peter Medawar recounted this event.
demand. Furthermore, hemodialysis facilities are usually
inadequate to make up this deficit so that many patients Early in the war, an R.A.F. Whitley bomber crashed into a house
are still dying of renal disease who could be restored to a in North Oxford with much serious injury and loss of life. Among
useful and productive life. Nevertheless, few of us would the injured was a young man with a third degree burn extend-
have imagined even 10 years ago that transplantation of ing over about 60% of his body. People burned as severely as this
the kidney would have become such a relatively common never raised a medical problem before: they always died; but the
procedure as is the case today, and indeed well over 30,000 blood transfusion services and the control of infection made possi-
kidney transplantations have been performed throughout ble by the topical use of sulphonamide drugs now made it possible
the world. for them to stay alive. Dr. John F. Barnes, a colleague of mine in
Transplantation of the kidney for the treatment of renal Professor H. W. Florey’s School of Pathology, asked me to see this
failure has been an attractive concept for many years. As patient in the hope that being an experimental biologist I might
long ago as 1945, three young surgeons at the Peter Bent have some ideas for treatment. With more than half his body sur-
Brigham Hospital in Boston, Charles Hufnagel, Ernest face quite raw, this poor young man was a deeply shocking sight;
Landsteiner and David Hume, joined the vessels of a I thought of and tried out a number of ingenious methods, none
cadaver kidney to the brachial vessels of a young woman of which worked, for ekeing out his own skin for grafting, trying
who was comatose from acute renal failure due to septice- to make one piece of skin do the work of ten or more. The obvious
mia. The kidney functioned for several days before it was solution was to use skin grafts from a relative or voluntary donor,
removed, and the woman regained consciousness. Shortly but this was not possible then and it is not possible now.
afterwards, the woman’s own kidneys began to function I believe I saw it as my metier to find out why it was not pos-
and she made a full recovery. The advent of the artificial sible to graft skin from one human being to another, and what
kidney at that time meant that this approach to the treat- could be done about it. I accordingly began research on the sub-
ment of acute renal failure was no longer necessary, but ject with the Burns Unit of the Glasgow Royal Infirmary, and
attention was soon given to the possibility of transplanting subsequently in the Zoology Department in Oxford. If anybody
kidneys to patients with end-stage renal failure who were had then told me that one day, in Oxford, kidneys would be
requiring dialysis on the newly developed artificial kidney transplanted from one human being to another, not as a perilous
to stay alive. surgical venture, but as something more in the common run of
Although the first experimental kidney transplants in ani- things, I should have dismissed it as science fiction; yet it is just
mals were reported first in Vienna by Dr. Emerich Ulmann this that has come about, thanks to the enterprise of Professor
in 1902 and then in 1905 by Dr. Alexis Carrel in the United Morris and his colleagues.
States, the problem of rejection was not mentioned by
either author. Later in 1910, Carrel did discuss the possible Nevertheless in 1951, David Hume in Boston embarked
differences between an autograft and a homograft. The vas- on a series of cadaver kidney transplants in which the kid-
cular techniques developed by Carrel for the anastomosis of ney was placed in the thigh of the recipient. All but one
the renal vessels to the recipient vessels are still used today. of these kidneys were rejected within a matter of days
But in 1923, Dr. Carl Williamson of the Mayo Clinic clearly or weeks, the one exception being a patient in whom the
defined the difference between an autografted and homo- kidney functioned for nearly 6 months and enabled the
grafted kidney and even published histological pictures of a patient to leave the hospital! This event provided hope for
rejecting kidney. Furthermore, he predicted the future use the future as no immunosuppressive therapy had been
of tissue matching in renal transplantation. used in this patient. At this time, the problems of rejection
of kidney allografts in the dog were being clearly defined
It is unfortunate that the lower animals, such as the dog, do not by Dr. Morton Simonsen in Copenhagen and Dr. William
possess a blood grouping like that of man. In the future it may be Dempster in London, but in 1953, a major boost to trans-
possible to work out a satisfactory way of determining the reac- plantation research was provided by the demonstration, by
tion of the recipient’s blood serum or tissues to those of the donor Drs. Rupert Billingham, Lesley Brent and Peter Medawar,
and the reverse; perhaps in this way we can obtain more light on that tolerance to an allogeneic skin graft in an adult animal
this as yet relatively dark side of biology. could be produced by injecting the fetus with donor strain
viii
Preface to the First Edition ix

tissue, thus confirming experimentally the clonal selection of renal transplantation improved. Another major area
hypothesis of Burnet and Fenner in the recognition of self of endeavor in renal transplantation at that time was
and non-self. The induction of specific unresponsiveness of directed at the study of methods of matching donor and
a host to a tissue allograft has remained the ultimate goal of recipient for histocompatibility antigens with the aim of
transplant immunologists ever since. lessening the immune response to the graft and so per-
Then in 1954, the first kidney transplant between iden- haps allowing a decrease in the immunosuppressive drug
tical twins was carried out successfully at the Peter Bent therapy. Although this aim has only been achieved to
Brigham Hospital which led to a number of further success- any great extent in siblings who are HLA identical, tis-
ful identical twin transplants in Boston and elsewhere in sue typing has made a significant contribution to renal
the world over the next few years. transplantation, perhaps best illustrated by the recogni-
There still remained the apparently almost insoluble prob- tion in the late sixties that the performance of a trans-
lem of rejection of any kidney other than an identical-twin plant in the presence of donor-specific presensitization in
kidney. The first attempts to suppress the immune response the recipient leads to hyperacute or accelerated rejection
to a kidney allograft employed total body irradiation of the of the graft in most instances. Nevertheless, the more
recipient and were carried out by Dr. Merril’s group in Bos- recent description of the Ia-like system in man (HLA-DR)
ton, two groups in Paris under the direction of Drs. Kuss may have an important impact on tissue typing in renal
and Hamburger, respectively, and by Professor Shackman’s transplantation. The present decade also has seen an
group in London. Rejection of a graft could be suppressed enormous effort directed at immunological monitoring in
by irradiation, but the complications of the irradiation renal transplantation and at attempts to induce experi-
were such that this was really an unacceptable approach, mental specific immunosuppression. We have solved
although an occasional relatively long-term acceptance of most of the technical problems of renal transplantation;
a graft provided encouragement for the future. we have been left with the problem of rejection and the
Then came the discovery by Drs. Schwartz and Dameshek complications arising from the drug therapy given to pre-
in 1959 that 6-mercaptopurine could suppress the immune vent rejection.
response of rabbits to human serum albumin. Shortly after- Although the contributions in this book cover all aspects of
wards, they showed that the survival of skin allografts in renal transplantation, certain subjects, as for example immu-
rabbits was significantly prolonged by the same drug. nological monitoring before transplantation, transplanta-
This event ushered in the present era of renal transplan- tion in children and cancer after renal transplantation, have
tation, for very quickly Roy Calne in London and Charles received considerable emphasis as they do represent devel-
Zukoski working with David Hume in Virginia showed that oping areas of great interest, and I must take responsibility
this same drug markedly prolonged the survival of kidney for this emphasis. For in the seventies we have seen many of
allografts in dogs. And indeed,6-mercaptopurine was first the principles and practice of renal transplantation become
used in a patient in Boston in 1960. Elion and Hitchings established and the areas of future investigation become more
of the Burroughs Wellcome Research Laboratories in New clearly defined. With an ever-increasing demand for renal
York State then developed azathioprine, which quickly transplantation, more and more people in many different dis-
replaced 6-mercaptopurine in clinical practice as it was less ciplines, doctors (surgeons, physicians, pathologists, virolo-
toxic. With the addition of steroids, the standard immuno- gists, immunologists), nurses, scientists and ancillary staff
suppressive therapy of today was introduced to the practice are becoming involved in renal transplantation either in the
of renal transplantation in the early sixties. clinic or in the laboratory. It is to these people I hope this book
Not that this meant the solution of the problems of will be of value.
renal transplantation for this combination of drugs was
dangerous and mortality was high in those early years. Sir Peter J. Morris
But there was a significant number of long-term suc- Oxford, UK
cessful transplants, and as experience grew, the results November 1978
Preface to the Eighth Edition

Kidney transplant patients and practitioners benefit from proteins and includes considerable new data on the clinical
updated knowledge of current and improved practice guide- use of costimulation blockade.
lines and novel techniques, in addition to being familiar Some areas of renal transplantation remain challenges
with well-established principles. For these reasons we have for the field and this certainly would include the sensitized
sought out leading international experts to write the chap- patient, antibody-mediated rejection, and management
ters of this 8th edition of the Textbook of Kidney Transplan- of chronic allograft failure. These topics are addressed in
tation. What has not changed over the past 41 years since detail in associated chapters. Quite a number of chapters
the first edition is Professor Morris’s dedication to the text- have been completely rewritten by new authors compared
book’s quality and his personal attention to the details that with the 7th edition, and we believe that these new chapters
are included. He has been the lead editor since the first edi- offer refreshing perspectives on their respective topics. We
tion, indeed the sole editor of the first five editions. Professor acknowledge that our field continues to be guided by new
Marson and Knechtle are delighted that he has chosen to basic and clinical research that in many cases is beyond the
include us as editors, as this text remains the most widely scope of this text, despite our desire to treat subjects compre-
circulated authoritative book on the subject of kidney hensively. We have sought to include what is most pertinent
transplantation, used internationally to help develop prac- to current clinical practice in the field. Our hope is that the
tice guidelines and train specialists. We are furthermore coming decades will continue to build on the remarkable
grateful to the authors who have produced the content record of progress in kidney transplantation that we have
of this 8th edition, including its up-to-date outcomes data witnessed since the first successful kidney transplant in
and analysis of the evidence supporting current practice in 1954 by the late Joseph Murray at Peter Bent Brigham Hos-
the field. Finally, we thank the leadership of Elsevier for its pital in Boston. Ours is an exciting field that offers improved
excellent communication with the authors and editors and and extended life to many persons with severely impaired
for their technical assistance with all aspects of the produc- renal function. It is also our hope that improved immuno-
tion of this complex project. suppressive therapy will further prolong graft survival and
In this 8th edition, we have chosen to combine the chap- reduce the side effects of infection and malignancy, ulti-
ters on azathioprine and mycophenolate based on the relat- mately extending patient survival yet further. Improved
edness of these compounds as inhibitors of cell proliferation. preservation techniques offer the prospect of increasing the
We have added two new chapters, one addressing kidney use of kidneys that were previously considered to be of inad-
allocation because policy varies in the international com- equate quality, and thereby increase the supply of donor
munity, reflecting the ethical and societal values of differ- kidneys. Improved antiviral agents have made possible the
ent countries and populations. Secondly, we have added a use of HIV-positive and hepatitis C-positive donor kidneys.
chapter on biomarkers of kidney injury and rejection. The These innovations are described in this updated text, which
latter is in recognition of the need for better monitoring we expect will inspire further good work.
tools for kidney injury and rejection to guide therapy and
patient management. Given the large number of candidate Stuart J. Knechtle
assays for injury and rejection and their relatively nascent Durham, NC, USA
status with respect to clinical use, we suspect that this Lorna P. Marson
will be a rapidly developing field in coming years and will Edinburgh, UK
help guide improvement of long-term kidney transplant Sir Peter J. Morris
outcomes. The chapter in the previous edition on belata- Oxford, UK
cept has merged with the chapter on antibody and fusion

x
Contributors

Gaurav Agarwal, MD Jeremy R. Chapman, AC, FRACP, FRCP


Assistant Professor of Medicine Clinical Director
University of Alabama at Birmingham Division of Medicine and Cancer
Birmingham, Alabama, USA Westmead Hospital, Westmead
Sydney, New South Wales, Australia
Talal M. Al-Qaoud, MD, FRCSC
Assistant Professor of Surgery Menna R. Clatworthy, BSc, MBBCh, PhD, FRCP
Department of Surgery, Division of Transplantation NIHR Research Professor and Reader in Immunity and
University of Maryland Inflammation
Baltimore, Maryland, USA Molecular Immunity Unit, Department of Medicine
University of Cambridge
Barbara D. Alexander, MD, MHS Cambridge, United Kingdom
Department of Medicine, Division of Infectious Diseases
Duke University Bradley Henry Collins, MD
Durham, North Carolina, USA Associate Professor
Department of Surgery
Richard D.M. Allen, MBBS, FRACS Duke University Medical Center
Professor Emeritus Durham, North Carolina, USA
University of Sydney
Sydney, Australia Robert B. Colvin, MD
Benjamin Castleman Distinguished Professor of Pathology
Frederike Ambagtsheer, PhD, LL.M. Pathology
Doctor Harvard Medical School and Massachusetts General
Internal Medicine, Transplantation and Nephrology Hospital
Erasmus MC Boston, Massachusetts, USA
Rotterdam, Netherlands
Lynn D. Cornell, MD
Rolf N. Barth, MD Consultant, Division of Anatomic Pathology
Professor Associate Professor of Laboratory Medicine and Pathology
Department of Surgery Mayo Clinic
University of Maryland School of Medicine Rochester, Minnesota, USA
Baltimore, Maryland, USA
Sylvia F. Costa, MD
Amit Basu, MD, FACS, FRCS Adjunct Assistant Professor
Attending Surgeon Medicine, Division of Infectious Diseases
Surgery Duke University
Jamaica Hospital Medical Center Durham, North Carolina, USA
Jamaica, New York, USA
Alice Crane, MD, PhD
Tomas Castro-Dopico, MBiochem, PhD Doctor
Research Associate Urology
Molecular Immunity Unit, Department of Medicine Cleveland Clinic
University of Cambridge Cleveland, Ohio, USA
Cambridge, United Kingdom

Eileen T. Chambers, MD
Associate Professor
Pediatrics and Surgery
Duke University
Durham, North Carolina, USA

xi
xii Contributors

Andrew Davenport, MD, FRCP Susan V. Fuggle, DPhil, MSc, BSc


Professor of Dialysis and ICU Nephrology Professor of Transplant Immunology
UCL Department for Nephrology Nuffield Department of Surgical Sciences
Royal Free Hospital University of Oxford;
University College London Consultant Clinical Scientist
London, United Kingdom Transplant Immunology and Immunogenetics
Laboratory, Oxford Transplant Centre
Matthew J. Ellis, MD Oxford University Hospitals NHS Foundation Trust
Medical Director Kidney Transplant Oxford, United Kingdom
Nephrology
Duke University Rouba Garro, MD
Durham, North Carolina, USA Assistant Professor
Pediatrics
Brian Ezekian, MD Emory University
General Surgery Resident Atlanta, Georgia, USA
Department of Surgery
Duke University Medical Center Robert S. Gaston, MD
Durham, North Carolina, USA Director
Comprehensive Transplant Institute;
Casey Victoria Farin, MD Professor of Medicine and Surgery
Clinical Fellow Robert G. Luke Endowed Chair in Transplant Nephrology
Multiple Sclerosis and Neuroimmunology University of Alabama at Birmingham
Department of Neurology Birmingham, Alabama, USA
Duke University
Durham, North Carolina, USA Edward K. Geissler, PhD
University Hospital Regensburg
Alton B. Farris III, MD Department of Surgery
Associate Professor, and Director Division of Experimental Surgery
Laboratory of Nephropathology and Electron Microscopy Regensburg, Germany
Department of Pathology
Emory University Sommer Elizabeth Gentry, PhD
Atlanta, Georgia, USA Professor
Mathematics
Jay A. Fishman, MD United States Naval Academy
Professor of Medicine Annapolis, Maryland, USA;
Harvard Medical School; Research Associate
Physician Surgery
Division of Infectious Disease and Massachusetts General Johns Hopkins University School of Medicine
Hospital Transplant Center Baltimore, Maryland, USA
Boston, Massachusetts, USA
James A. Gilbert, BM, BS, MA(Ed), FRCS
Sander Florman, MD Consultant Transplant and Vascular Access Surgeon
Professor of Surgery, Director Renal and Transplant
Recanati/Miller Transplantation Institute Oxford University Hospitals
Icahn School of Medicine at Mount Sinai Oxford, United Kingdom
New York City, New York, USA
David Hamilton, MB, ChB, PhD, FRCS
John L.R. Forsythe, MD, FRCS Eng, FRCS Ed, FEBS, Retired Transplant Surgeon
FRCPEd Independent Scholar
Honorary Professor The World of Learning
Transplant Surgery St. Andrews, United Kingdom
University of Edinburgh
Edinburgh, United Kingdom Reem E. Hamoda, MPH
Public Health Program Associate
Peter J. Friend, MD Department of Surgery, Division of Transplantation
Professor of Transplantation Emory University School of Medicine
Nuffield Department of Surgical Sciences Atlanta, Georgia, USA
University of Oxford
Oxford, United Kingdom
Contributors xiii

Benson M. Hoffman, MA, PhD Stuart J. Knechtle, MD, FACS


Associate Professor William R. Kenan, Jr. Professor of Surgery
Department of Psychiatry and Behavioral Sciences Executive Director, Duke Transplant Center
Duke University Medical Center Duke University School of Medicine
Durham, North Carolina, USA Durham, North Carolina, USA

Matthew L. Holzner, MD Simon R. Knight, MA, MB, MChir, FRCS


Postdoctoral Fellow Senior Clinical Research Fellow
Recanati/Miller Transplantation Institute Centre for Evidence in Transplantation
Icahn School of Medicine at Mount Sinai Oxford Transplant Centre
New York City, New York, USA Nuffield Department of Surgical Sciences
University of Oxford
Joanna Hooten, BS, MD Oxford, United Kingdom
Staff Dermatologist
Chelsea Dermatology Kate Kronish, MD
Chelsea, Michigan, USA Assistant Professor
Department of Anesthesia and
James P. Hunter, BSc (Hons), MBChB, MD, FRCS Perioperative Care
Senior Clinical Research Fellow and Consultant Transplant University of California San Francisco
Surgeon San Francisco, California, USA
University Hospitals Coventry and Warwickshire
Coventry, United Kingdom; John C. LaMattina, MD
University of Oxford Associate Professor
Oxford, United Kingdom Surgery
University of Maryland School of Medicine
Alan G. Jardine, BSc, MD, FRCP Baltimore, Maryland, USA
Professor of Renal Medicine and Head of the
Undergraduate Medical School Jennifer S. Lees, BA (Hons), MA (Cantab), MBChB, MRCP
Institute of Cardiovascular and Medical Sciences Clinical Research Fellow/Specialist
University of Glasgow Trainee in Nephrology
Glasgow, United Kingdom University of Glasgow/NHS Greater
Glasgow and Clyde
Laura S. Johnson, MD Glasgow, United Kingdom
Assistant Professor
Department of Surgery Henri Leuvenink, PhD
Georgetown University School of Medicine UMCG
Washington, District of Columbia, USA Surgery
University Medical Center Groningen
Arman A. Kahokehr, MB, PhD, FRACS Grorolfningen, Netherlands
Fellow in Reconstructive Urology
Duke University Medical Center Jayme E. Locke, MD, MPH, FACS, FAST
Durham, North Carolina, USA Associate Professor of Surgery
Director
Dixon B. Kaufman, MD, PhD University of Alabama at Birmingham Comprehensive
Ray D. Owen Professor of Surgery Transplant Institute
Department of Surgery, Division of Transplantation Birmingham, Alabama, USA
University of Wisconsin-Madison School of Medicine and
Public Health Michael R. Lucey, MB, BCh, MD
Madison, Wisconsin, USA Professor
Medicine, Gastroenterology and Hepatology
Karen L. Keung, MBBS, FRACP University of Wisconsin School of Medicine and Public
Nephrologist Health
Centre for Transplant and Renal Research Madison, Wisconsin, USA
Westmead Institute of Medical Research
Westmead, New South Wales, Australia Matthew William Luedke, MD
Assistant Professor
Allan D. Kirk, MD, PhD Department of Neurology
Professor and Chairman Duke University;
Surgery Department of Medicine, Division of Neurology
Duke University Veterans Affairs Medical Center
Durham, North Carolina, USA Durham, North Carolina, USA
xiv Contributors

Anne Louise Marano, BS/BA, MD Brian J. Nankivell, MB, BS, MSc, PhD, MD, MRCP(UK),
Assistant Professor FRACP
Dermatology Doctor, Renal Medicine
Duke Univeristy Westmead Hospital
Durham, North Carolina, USA Westmead, New South Wales, Australia

Lorna P. Marson, MBBS, MD, FRCSEng, FRCSEd, Claus U. Niemann, MD


FRCP(Ed) Professor
Professor of Transplant Surgery Department of Anesthesia and Perioperative Care;
Clinical Sciences (Surgery) Professor
University of Edinburgh Department of Surgery
Edinburgh, United Kingdom University of California San Francisco
San Francisco, California, USA
Chantal Mathieu, MD, PhD
Professor of Medicine John O’Callaghan, BSc, MBBS, DPhil
Faculty of Medicine Katholieke Specialty Registrar
University of Leuven (KU Leuven); Transplantation Surgery
Head, Endocrinology Oxford University Hospitals
University Hospitals Leuven; Oxford, United Kingdom
Laboratory of Clinical and Experimental
Endocrinology Philip John O’Connell, MD, PhD
University of Leuven (KU Leuven) Director of Transplantation
Leuven, Belgium Renal Unit
University of Sydney at Westmead Hospital
Madhav C. Menon, MBBS, MD, FACP Westmead, New South Wales, Australia
Assistant Professor
Medicine Nephrology and Recanati-Miller Transplant Jon S. Odorico, MD
Institute Professor of Surgery
Icahn School of Medicine at Mount Sinai Department of Surgery, Division of Transplantation
New York City, New York, USA University of Wisconsin-Madison School of Medicine and
Public Health,
Sir Peter J. Morris, MD, PhD, FRS, FRCS Madison, Wisconsin, USA
Emeritus Nuffield Professor of Surgery
University of Oxford; Andrea Olmos, MD
Honorary Professor Assistant Professor
University of London; Department of Anesthesia and Perioperative Care
Nuffield Department of Surgical Sciences University of California San Francisco
University of Oxford San Francisco, California, USA
Oxford, United Kingdom
Gabriel Oniscu, MD, FRCS
Elmi Muller, MBChB, PhD Consultant Transplant Surgeon
Professor Transplant Unit
Surgery Royal Infirmary of Edinburgh;
Groote Schuur Hospital Honorary Clinical Senior Lecturer
University of Cape Town Clinical Surgery
Cape Town, Western Cape, South Africa University of Edinburgh
Edinburgh, United Kingdom
Barbara Murphy, MD
Murray M. Rosenberg Professor of Medicine Rachel E. Patzer, PhD, MPH
Chair of the Department of Medicine Associate Professor
Mount Sinai Health System Department of Surgery
New York City, New York, USA Emory University School of Medicine;
Rollins School of Public Health
Sarah A. Myers, MD Department of Epidemiology
Professor Emory University
Dermatology Atlanta, Georgia, USA
Duke University
Durham, North Carolina, USA
Contributors xv

Liset H.M. Pengel, PhD Caroline K. Saulino, PsyD


Co-director Medical Instructor
Peter Morris Centre for Evidence in Transplantation Department of Psychiatry and Behavioral Sciences
Nuffield Department of Surgical Sciences Duke University
University of Oxford Durham, North Carolina, USA
Oxford, United Kingdom
Carrie Schinstock, MD
Andrew C. Peterson, MD, FACS Doctor
Professor of Surgery Nephrology and Hypertension
Duke University Medical Center; Mayo Clinic
Urology Residency Program Director Rochester, Minnesota, USA
Duke University
Durham, North Carolina, USA Paul M. Schroder, MD, PhD
Research Fellow
Jacques Pirenne, MD, MSc, PhD Department of Surgery
Professor of Surgery Duke University School of Medicine
Faculty of Medicine Durham, North Carolina, USA
University of Leuven (KU Leuven);
Head Dorry L. Segev, MD, PhD
Abdominal Transplant Surgery Professor of Surgery and Epidemiology
University Hospitals Leuven; Surgery
Director Johns Hopkins University;
Abdominal Transplant Surgery Laboratory Associate Vice Chair
Department of Microbiology and Immunology Surgery
University of Leuven (KU Leuven) Johns Hopkins Hospital
Leuven, Belgium Baltimore, Maryland, USA

Rutger J. Ploeg, MD, PhD, FRCS Ron Shapiro, MD


Professor of Transplant Biology and Consultant Surgeon Professor of Surgery, Surgical Director
Nuffield Department of Surgical Sciences Kidney/Pancreas Transplantation
University of Oxford Recanati/Miller Transplantation Institute
Oxford, United Kingdom; Icahn School of Medicine at Mount Sinai
Professor of Transplant Biology New York City, New York, USA
LUMC Transplant Centre
University of Leiden Daniel A. Shoskes, MD, MSc, FRCSC
Leiden, Netherlands; Professor of Urology
Professor of Transplant Surgery Urology
Surgery Cleveland Clinic
Medical Faculty Cleveland, Ohio, USA
University of Groningen
Groningen, Netherlands Patrick J. Smith, PhD, MPH
Associate Professor
Brenda Maria Rosales, BA, BMS (Honours), MPH Department of Psychiatry and Behavioral Sciences
Sydney School of Public Health Department of Medicine, Division of Pulmonary, Allergy,
The University of Sydney and Critical Care Medicine
Sydney, New South Wales, Australia Department of Population Health Sciences, Center for
Health Measurement
Nasia Safdar, MD Duke University Medical Center
Professor Durham, North Carolina, USA
Medicine, Infectious Diseases
University of Wisconsin School of Medicine and Public Ben Sprangers, MD, PhD, MBA, MPH
Health Associate Professor of Medicine
Madison, Wisconsin, USA Faculty of Medicine
University of Leuven (KU Leuven);
Adnan Said, MD, MS Department of Nephrology
Associate Professor University Hospitals Leuven;
Medicine, Gastroenterology and Hepatology Laboratory of Molecular Immunology (Rega Institute)
University of Wisconsin School of Medicine and Public Microbiology and Immunology
Health University of Leuven (KU Leuven)
Madison, Wisconsin, USA Leuven, Belgium
xvi Contributors

Mark Stegall, MD Christopher J.E. Watson, MA, MD, BChir


Transplant Center Professor of Transplantation
Mayo Clinic Department of Surgery
Rochester, Minnesota, USA University of Cambridge
Cambridge, United Kingdom
Ram M. Subramanian, MD
Associate Professor Angela Claire Webster, MBBS, MM (Clin Epid), PhD
Medicine and Surgery Professor of Clinical Epidemiology
Emory University School of Public Health
Atlanta, Georgia, USA University of Sydney
Sydney, New South Wales, Australia;
Craig J. Taylor, PhD, FRCPath Senior Staff Specialist
Consultant Clinical Scientist [Retired] Centre for Transplant and Renal Research
Histocompatibility and Immunogenetics (Tissue Typing) Westmead Hospital
Laboratory Westmead, New South Wales, Australia
Cambridge University Hospitals
Cambridge, Cambridgeshire, United Kingdom Willem Weimar, MD, PhD
Professor, Doctor
John F. Thompson, MD Internal Medicine
Professor of Melanoma and Surgical Oncology Erasmus MC
The University of Sydney Rotterdam, Netherlands
Melanoma Institute Australia;
Professor of Surgery (Melanoma and Surgical Oncology) Pamela D. Winterberg, MD
Sydney Medical School Assistant Professor
The University of Sydney Pediatric Nephrology
Sydney, New South Wales, Australia Emory University School of Medicine;
Children’s Healthcare of Atlanta
Vikram Wadhera, MD Atlanta, Georgia, USA
Assistant Professor of Surgery
Recanati/Miller Transplantation Institute Kathryn J. Wood, MD
Icahn School of Medicine at Mount Sinai Transplantation Research Immunology Group
New York City, New York, USA Nuffield Department of Surgical Sciences
University of Oxford
Mark Waer, MD, PhD Oxford, United Kingdom
Emeritus Professor of Medicine
Faculty of Medicine Diana A. Wu, MBChB
University of Leuven (KU Leuven); Research Fellow
Laboratory of Experimental Transplantation, Transplant Surgery
Microbiology and Immunology University of Edinburgh
University of Leuven (KU Leuven) Edinburgh, United Kingdom
Leuven, Belgium
1 Kidney Transplantation:
A History
DAVID HAMILTON

CHAPTER OUTLINE Early Experiments A Time of Optimism


Human Kidney Transplants Tissue Typing
The Middle Years The 1970s Plateau
Post World War II Waiting for Xenografts
Immunosuppression and the Modern Era Conclusion
Chemical Immunosuppression

The modern period of transplantation began in the late for a while. Neither Ullmann nor von Decastello continued
1950s, but two earlier periods of interest in clinical and with this work, although von Decastello was noted for his
experimental transplantation were the early 1950s and work on blood groups, and Ullmann published extensively
the first two decades of the 20th century. Hamilton1 on bowel and biliary surgery.
provides a bibliography of the history of organ trans- In Lyon, the department headed by Mathieu Jabou-
plantation. Table 1.1 summarizes landmarks in kidney lay (1860–1913) had a major influence (Fig. 1.2). In his
transplantation. research laboratories, his assistants Carrel, Briau, and Vil-
lard worked on improved methods of vascular suturing,
leading to Carrel’s famous article credited with establish-
Early Experiments ing the modern method of suturing.4 Carrel left to work in
the United States, and in the next 10 years he published
Interest in transplantation developed in the early part of the extensively on organ grafting, successfully carrying out
20th century because experimental and clinical surgical autografts of kidneys in cats and dogs and, showing that
skills were rapidly advancing, and many of the pioneering allografts, contrary to accepted opinion, eventually failed
surgeons took an interest in vascular surgical techniques as after functioning briefly, established the existence of “rejec-
part of their broad familiarity with the advance of all aspects tion” as it was later termed. He made attempts at tissue
of surgery. Payr’s demonstration of the first workable, matching and demonstrated cold-preservation of tissues.
although cumbersome, stent method of vascular suturing led He was awarded a Nobel Prize for this work in 1912.5
to widespread interest in organ transplantation in Europe.
Many centers were involved, notably Vienna, Bucharest,
and Lyon. The first successful experimental organ trans- Human Kidney Transplants
plant was reported by Ullmann in 1902. Emerich Ullmann
(1861–1937) (Fig. 1.1) had studied under Edward Albert Jaboulay, Carrel’s teacher, had carried out the first recorded
before obtaining a position at the Vienna Medical School, human kidney transplant in 1906,6 although Ullmann later
which was then at its height. Ullmann’s article shows that claimed an earlier attempt in 1902.7 Jaboulay was later to
he managed to autotransplant a dog kidney from its normal be better known for his work on thyroid and urologic sur-
position to the vessels of the neck, which resulted in some gery, but, doubtless encouraged by the success of Carrel and
urine flow. The animal was presented to a Vienna medical others in his laboratory, he carried out two xenograft kid-
society on March 1, 1902, and caused considerable com- ney transplants using a pig and goat as donors, transplant-
ment.2 At this time, Ullmann was chief surgeon to the Spital ing the organ to the arm or thigh of patients with chronic
der Baumhertigen Schwestern, and his experimental work renal failure. Each kidney worked for only 1 hour. This
was done in the Vienna Physiology Institute under Hofrath choice of an animal donor was acceptable at that time in
Exner. Exner’s son Alfred had already tried such a trans- view of the many claims in the surgical literature for suc-
plant without success. In the same year, another Vienna cess with xenograft skin, cornea, or bone.
physician, Alfred von Decastello, physician assistant at the More is known of the second and third attempts at
Second Medical Clinic, carried out dog-to-dog kidney trans- human kidney transplantation. Ernst Unger (1875–
plants at the Institute of Experimental Pathology.3 1938) (Fig. 1.3) had a thorough training in experimental
Ullmann and von Decastello had used Payr’s method, work and set up his own clinic in 1905 in Berlin, being
and later in 1902 Ullmann demonstrated a dog-to-goat joined there by distinguished colleagues. He continued
kidney transplant that, to his surprise, passed a little urine with experimental work and by 1909 reported successful
1
2 Kidney Transplantation: Principles and Practice

TABLE 1.1 Landmarks in Kidney Transplantation


1902 First successful experimental kidney
transplant2
1906 First human kidney transplant—xenograft6
1933 First human kidney transplant—allograft54
1950 Revival of experimental kidney transplanta-
tion4,16,57
1950–1953 Human kidney allografts without immunosuppres-
sion, in Paris18,19,56,59 and Boston21
1953 First use of live related donor, Paris20
1954 First transplant between identical twins, Boston22
1958 First description of leukocyte antigen MAC62
1959–1962 Radiation used for immunosuppression, in Bos-
ton24 and Paris25,56
1960 Effectiveness of 6-mercaptopurine (6-MP) in dog
kidney transplants29,42
1960 Prolonged graft survival in patient given 6-MP Fig. 1.2 Mathieu Jaboulay (1860–1913) and his surgical team at Lyon
after irradiation34 in 1903. Until his death in a rail accident, Jaboulay made numerous
1962 First use of tissue matching to select a donor and surgical contributions and encouraged Alexis Carrel’s work on vascu-
recipient44,47,49,56 lar anastomosis. In 1906 Jaboulay reported the first attempt at human
1966 Recognition that positive crossmatching leads to kidney transplantation.
hyperacute rejection29,50,56
1967 Creation of Eurotransplant46
1967 Development of kidney preservation
1973 Description of the transfusion effect57
1978 First clinical use of cyclosporine55
1978 Application of matching for HLA-DR in renal
transplantation29
1987 First of new wave of immunosuppressive agents
appears (tacrolimus)
1997 Transgenic pigs strategy63
2010 Laparoscopic kidney insertion64

Fig. 1.3 A contemporary cartoon of Ernst Unger (1875–1938) at work


at the Rudolf Virchow Hospital, Berlin. (Courtesy the Rudolf Virchow
Hospital.)

transplantation of the kidneys en masse from a fox terrier


to a boxer dog. The urine output continued for 14 days,
and the animal was presented to two medical societies. By
1910, Unger had performed more than 100 experimen-
tal kidney transplants. On December 10, 1909, Unger
attempted a transplant using a stillborn child’s kidney
grafted to a baboon. No urine was produced. The animal
died shortly after the operation, but postmortem exami-
nation showed that the vascular anastomosis had been
successful. This success and the new knowledge that mon-
keys and humans were serologically similar led Unger to
Fig. 1.1 Emerich Ullmann (1861–1937) carried out the first experimen-
tal kidney transplants in dogs in 1902. (Courtesy the Vienna University,
attempt, later in the same month, a monkey-to-human
Institute for the History of Medicine.) transplant.8 The patient was a young girl dying of renal
1 • Kidney Transplantation: A History 3

failure, and the kidney from an ape was sutured to the


thigh vessels. No urine was produced. Unger’s report con-
cluded that there was a biochemical barrier to transplan-
tation, a view mistakenly advocated by the basic science of
the day; his main contributions thereafter were in esopha-
geal surgery. (For a biography of Unger, see Winkler.9)
These early experiments established that kidney trans-
plants were technically possible. Methods of study of renal
function were primitive then; without routine measurement
of blood urea and without any radiologic methods, subtle
studies of transplant function were impossible. This impos-
sibility plus the uncertainty of the mechanism of allograft
rejection led to a diminished interest in organ transplantation
after about 10 years of activity. By the start of World War Fig. 1.4 Yu Yu Voronoy (1895–1961) had experience with dog allografts
I, interest in organ transplantation had almost ceased and before carrying out the first human kidney allograft in 1933 at Kherson
in the Ukraine. His experimental animal model is shown here.
was not resumed in the European departments of surgery
after the war. Carrel had switched his attention to studies
of tissue culture. Interest elsewhere also was low; in Brit- at immunosuppression. In transplantation circles, such as
ain and the United States, scarce research funds were being they were, there was not even the confidence to counter the
applied to fundamental biochemistry and physiology, rather vivid claims of Voronoff to rejuvenate human patients via
than applied projects of clinical relevance. Transplantation monkey gland grafts, and the endless reports of successful
immunology faded away after the bright start in the capable human skin homografts were not examined critically.
surgical hands of Carrel, Murphy’s sound grasp of immuno- The main event of this period was an isolated and little-
suppression, and Landsteiner’s awareness of the serologic known event—the first human kidney allograft. It was
detection of human antigens. Carrel, Murphy, and Land- performed in the Ukraine by the Soviet surgeon Yu Yu
steiner all worked at the Rockefeller Institute in New York. Voronoy.11 Voronoy was an experienced investigator, and
In 1914, in a remarkable lecture to the International he eventually performed six such transplants up to 1949.
Surgical Society, Carrel did anticipate the future develop- Voronoy (1895–1961) trained in surgery at Kiev under
ment of transplantation. His colleague at the Rockefeller Professor V.N. Shamov and obtained experience there
Institute, J. B. Murphy, had found that radiation or benzol with serologic methods of blood transfusion, then in their
treatment would increase the “take” of tumor grafts in rats, developmental stage. He used these methods to detect com-
and Carrel realized the potential of these findings: plement-fixing antibodies after testis slice transplants, and
later he had some success with the same methods applied
It is too soon to draw any definite conclusions from these experi- to kidney grafts (Fig. 1.4). In 1933 Voronoy transplanted a
ments. Nevertheless it is certain that a very important point has human kidney of blood group B to a patient of blood group O
been acquired with Dr. Murphy’s discovery that the power of with acute renal failure as a result of mercuric chloride poi-
the organism to eliminate foreign tissue was due to organs such soning. The donor kidney was obtained from a patient dying
as the spleen or bone marrow, and that when the action of these as a result of a head injury and was transplanted to the thigh
organs is less active a foreign tissue can develop rapidly after vessels under local anesthetic; the warm time for the kidney
it has been grafted. It is not possible to foresee whether or not was about 6 hours. There was a major mismatch for blood
the present experiments of Dr. Murphy will lead directly to the groups, and despite a modest exchange transfusion, the
practical solution of the problem in which we are interested. The kidney never worked. The patient died 2 days later; at post-
surgical side of the transplantation of organs is now completed, mortem, the donor vessels were patent. By 1949, Voronoy
as we are now able to perform transplantations of organs with reported six such transplants, although no substantial func-
perfect ease and with excellent results from an anatomical tion had occurred in any. (For a biography of Voronoy, see
standpoint. But as yet the methods cannot be applied to human Hamilton and Reid12 and Matevossian and colleagues.13)
surgery, for the reason that homoplastic transplantations are
almost always unsuccessful from the standpoint of the function-
ing of the organs. All our efforts must now be directed toward Post World War II
the biological methods which will prevent the reaction of the
organism against foreign tissue and allow the adapting of homo- The sounder basis of transplantation immunology, which
plastic grafts to their hosts.10 followed Medawar’s pioneer studies during World War II,
led to a new interest in human transplantation. In 1946
a human allograft kidney transplant to arm vessels under
The Middle Years local anesthetic was attempted by Hufnagel, Hume, and
Landsteiner at the Peter Bent Brigham Hospital in Boston.
Until the revival of interest in transplantation in the 1950s, The brief period of function of the kidney may have helped the
the 1930s and 1940s were a stagnant period in clinical sci- patient’s recovery from acute renal failure; it marked the
ence. The great European surgical centers had declined; in beginning of that hospital’s major interest in transplanta-
North America, only at the Mayo Clinic was there a cau- tion and dialysis.14
tious program of experimental transplantation without In the early 1950s, interest in experimental and clinical
building on Carrel’s work, notably failing to make attempts kidney transplantation increased. With a growing certainty
4 Kidney Transplantation: Principles and Practice

to encourage future careful empirical surgical adventures,


despite advice from scientists to wait for elegant immuno-
logic solutions. Although small doses of adrenocortico-
tropic hormone or cortisone were used, it was thought that
the endogenous immunosuppression of uremia was respon-
sible for these results, rather than the drug regimen. Many
of Hume’s tentative conclusions from this short series were
confirmed later, notably that prior blood transfusion might
be beneficial, that blood group matching of graft and donor
might be necessary, and that host bilateral nephrectomy
was necessary for control of posttransplant blood pres-
sure.21 The first observation of recurrent disease in a graft
was made, and accelerated arteriosclerosis in the graft ves-
sels was noted at postmortem. Other cases were reported
from Chicago, Toronto, and Cleveland in the early 1950s,
but because no sustained function was achieved, interest
in clinical and experimental renal allograft transplantation
waned, despite increasing knowledge of basic immunologic
mechanisms in the laboratory.
The technical lessons learned from the human allograft
attempts of the early 1950s allowed confidence in the surgi-
cal methods, and in Boston, on December 23, 1954, the first
transplant of a kidney from one identical twin to another
Fig. 1.5 David M. Hume (1917–1973) pioneered human kidney trans- with renal failure was performed. From then on, many
plantation at the Peter Bent Brigham Hospital, Boston, and the Medical
College of Virginia. He died in an air crash at the age of 55. such transplantations were performed successfully in Bos-
ton.22 Although sometimes seen now merely as a technical
triumph, valuable new findings emerged from this series.
that immunologic mechanisms were involved, the destruc- Some workers had predicted that, in the short term, the
tion of kidney allografts could be reinvestigated. Simonsen, activity of the inactive bladder could not be restored, and
then an intern in Ålborg in Denmark, persuaded his surgi- that in the long term, human kidney grafts would decline
cal seniors to teach him some vascular surgery; using dog in vitality as a result of denervation or ureteric reflux. Other
kidney transplants, he reported on the mechanism of kid- workers were convinced that a single kidney graft could not
ney rejection.15 Dempster in London also reexamined this restore biochemical normality to an adult, and that in any
question.16 Both workers found, like Küss in Paris, that the case the existing changes caused by chronic renal failure
pelvic position of the kidney was preferable to a superficial were irreversible. All of these gloomy predictions were neu-
site, and both concluded that an immunologic mechanism tralized by the success of the twin kidney transplants, and
was responsible for failure. Dempster found that radiation, the greatest triumph came when one such recipient became
but not cortisone, delayed rejection. Both workers consid- pregnant and had a normal infant, delivered cautiously by
ered that a humoral mechanism of rejection was likely. cesarean section, with the anxious transplanters in atten-
In the early 1950s, two groups simultaneously started dance. Many of the twin recipients are still alive today,
human kidney transplantation. In Paris, with encourage- although the good results were tempered by failures caused
ment from the nephrologist Jean Hamburger, the surgeons by the prompt return of glomerulonephritis in some trans-
Küss (five cases),17 Servelle (one case),18 and Dubost (one planted kidneys. This complication was later much reduced
case)19 reported on kidney allografts without immunosup- by immunosuppression. Other lessons learned were that the
pression in human patients, placing the graft in the now- hazard of multiple donor renal arteries provided a need for
familiar pelvic position. The Paris series included a case pretransplant angiography of the kidneys in living donors,
reported by Hamburger of the first live-related kidney trans- although it still was not thought necessary to perfuse or
plant, the donor being the mother of a boy whose solitary cool the donor organ. Lastly, there was the first airing of the
kidney had been damaged in a fall from a height. The kid- legal aspects of organ donation, particularly the problem of
ney functioned immediately, but was rejected abruptly on consent in young, highly motivated related donors. (For an
the 22nd day.20 In the United States, the Chicago surgeon account of this period, see Murray and colleagues.23)
Lawler had been the first to attempt such an intraabdomi-
nal kidney allograft in 1950; it was met with the intense
public interest and professional skepticism that were to
Immunosuppression and
characterize innovative transplantation thereafter. the Modern Era
A series of nine cases, closely studied, was recorded
from Boston, using the thigh position of the graft, and for In 1948, the first patients crippled with rheumatoid arthri-
the first time hemodialysis had been used in preparing tis were given the Merck Company’s Cortone (cortisone) at
the patients, employing Merrill’s skill with the early Kolff/ the Mayo Clinic, and intense worldwide interest in the phar-
Brigham machine. David Hume (Fig. 1.5) reported on this macologic actions of adrenal cortical hormones followed.
Boston experience in 1953. Modest unexpected survival of Careful studies by Medawar’s group in the early 1950s
the kidney was obtained in some of these cases and served suggested a modest immunosuppressive effect of cortisone,
1 • Kidney Transplantation: A History 5

but when Medawar shortly afterward showed profound,


specific, and long-lasting graft acceptance via the induction
of tolerance, the weak steroid effect was understandably
sidelined and thought to be of no clinical interest. Induc-
tion of tolerance in adult animals (rather than newborns)
was accomplished by lethal irradiation and bone marrow
infusion, and with this strong lead from the laboratory, it
was natural that the first attempts at human immunosup-
pression for organ transplants were with preliminary total-
body irradiation and allograft bone marrow rescue. These
procedures were carried out in Paris, Boston, and elsewhere
in the late 1950s.
This regimen was too difficult to control, and graft-ver-
sus-host disease was inevitable. It was found unexpectedly
that sublethal irradiation alone in human patients was
quite immunosuppressive, however, and this approach was Fig. 1.6 R. Küss (right) and M. Legrain (center) in 1960 with their first
used until 1962, the year of the first general availability of long-term kidney transplant survivor. The patient and her brother-
azathioprine (Imuran). In Boston, 12 patients were treated in-law donor (center right) are shown with the staff of the unit at the
in this way, but with only one long-term survival in a man Hôpital Foch. Immunosuppression with irradiation and mercaptopu-
rine was used. (Courtesy Prof. M. Legrain.)
receiving his transplant from his nonidentical twin.24 In
Paris, similar success was obtained with sibling grafts.25,26
These isolated kidney survivals after a single dose of radia- human kidney function was obtained, but in Paris, Küss and
tion gave further hope and showed again that the immu- associates34 reported one prolonged survival of a kidney from
nology of humans, dogs, and mice is different. These cases a nonrelated donor when 6-MP was used with intermittent
also showed that if a human organ could survive the initial prednisone in a recipient who also had received irradiation as
crucial rejection period, it could be protected or adapted to the main immunosuppressive agent (Fig. 1.6). This case was
the host in some way, possibly shielded by new endothe- the first success for chemical immunosuppression.
lium, by enhancement, or, as suggested later, by microchi- This change in approach, giving lifelong, risky medica-
meric tolerance induced by mobile cells in the graft. tion with toxic drugs, although an obvious development in
retrospect, was accepted with reluctance because it meant
leaving aside, at least in the short term, the hopes from the
Chemical Immunosuppression work of the transplantation immunologists for the elegant,
specific, one-shot, nontoxic tolerance regimen. Many work-
In 1958, at the New England Medical Center, attempts were ers thought that entry into this new paradigm was only a
made at human bone marrow transplantation for aplastic temporary diversion.
anemia and leukemia. To enable the marrow grafts to suc- In 1961 azathioprine became available for human use;
ceed, irradiation of the recipient was used. Results were poor, the dosage was difficult to judge at first. The first two Bos-
and mortality was high. Schwartz and Dameshek27 looked ton cases using the drug did not show prolonged survival
for alternatives to irradiation and reasoned that an antican- of the grafts, but in April 1962 the first extended successes
cer drug, such as 6-mercaptopurine (6-MP) or methotrexate, with human kidney allografts were obtained.35 Shortly
might be of use for immunosuppression in their patients. (For afterward, at the bedside rather than in the laboratory, it
an account of this period, see Schwartz.28) Their important was discovered that steroids, notably prednisolone, when
paper in 1959, showing a poor immune response to foreign given with azathioprine had a powerful synergistic effect.
protein in rabbits treated with 6-MP,27 was noticed by Roy The regular use of both together became a standard regi-
Calne, then a surgeon in training at the Royal Free Hospital, men after reports by Starzl and colleagues36 and Goodwin
London, and David Hume, new Chairman of Surgery at the and coworkers,37 and this combined therapy continued to
Medical College of Virginia. Calne had been disappointed at be the routine immunosuppressive method despite many
the failure of irradiation to prolong kidney allograft survival other suggested alternatives, until azathioprine was dis-
in dogs and, like others looking for an alternative, he found placed by cyclosporine much later. Use of the combined
that 6-MP was successful.29 Zukoski and colleagues30 in immunosuppression and the increasing use of live related
Richmond found the same effect. donors (rather than occasional twin or free or cadaver kid-
In 1960 Calne visited Boston for a period of research with neys), along with the remarkably good results reported
Murray, and Hitchings and Elion of Burroughs Wellcome, in 1963 from Denver36 and Richmond,38 greatly encour-
then at Tuckahoe, provided him with new derivatives of aged the practice of transplantation. (For an account of this
6-MP.31 Of these, BW57-322 (later known as azathioprine period, see Starzl.39)
[Imuran]) proved to be more successful in dog kidney trans-
plants and less toxic than 6-MP.32
From 1960 to 1961, 6-MP was used in many human A Time of Optimism
kidney transplants. In London at the Royal Free Hospital,
three cases were managed in this way, but without success, The mid-1960s was a period of great optimism. The rapid
although one patient receiving a live related transplant died improvement in results seemed to indicate that routine suc-
of tuberculosis rather than rejection.33 In Boston, no lasting cess was at hand. Looking to the future, calculations were
6 Kidney Transplantation: Principles and Practice

Medawar produced a powerful immunosuppressive anti-


lymphocyte serum, and production of versions suitable
for human use started.42 Initial results were favorable, but
the whole antilymphocyte serum had an unspectacular
role thereafter, added to from 1975 onward by the use of
monoclonal antibody versions. Hopes for another biologi-
cal solution to transplantation were raised in 1969 when
French and Batchelor43 found an enhancing serum effect in
the new experimental model of rat kidney transplantation
made possible by the development of microsurgical meth-
ods, but it proved impossible to mimic the effect in humans.

Tissue Typing
Fig. 1.7 Jean Dausset first described an antigen MAC, later known as
HLA-2, defined by numerous antisera from multitransfused patients, The greatest hopes resided in the evolution of tissue-typing
and which later was shown to be part of the major histocompatibility methods, which entered routine use in 1962 (Fig. 1.7).44,45
complex in humans (HLA). The increasing identification of the antigens of the human
leukocyte antigen (HLA) system seemed to promise excellent
made that suggested that enough donor organs would be clinical results in the future from close matching made pos-
available in the future if all large hospitals cooperated, and sible when choosing from a large pool of patients. Sharing of
such donations did start to come from outside the transplan- kidneys in Europe started in 1967 at van Rood’s suggestion,46
tation pioneer hospitals. Transplantation societies were set and in North America, Amos and Terasaki set up similar
up, and specialist journals were started. The improvements sharing schemes on both coasts of the United States. Others
in regular dialysis treatment meant an increasing pool of followed throughout the world, and these organizations not
patients in good health suitable for transplantation, and only improved the service but also soon gathered excellent
this allowed for better and planned preparation for trans- data on kidney transplant survival. The need to transport
plantation. With a return to dialysis being possible, heroic kidneys within these schemes encouraged construction of
efforts to save a rejected kidney were no longer necessary. perfusion pumps designed to increase the survival of organs
Management of patients improved in many aspects, and and the distance they could be transported.47 Much work on
the expected steroid long-term effects were met and man- perfusion fluids was done until the intracellular type of fluid
aged (primarily by the demonstration that low-dose steroids devised by Collins et al. in 1969 allowed a simple flush and
were as effective as high-dose steroids). The need for cool- chill to suffice for prolonged storage.48 Although the hopes for
ing of donor organs was belatedly recognized, many tests of typing were not fully realized, such schemes had other ben-
viability were announced, and transport of organs between efits in obtaining kidneys when urgently required for patients
centers began. Bone disease and exotic infections were with rarer blood groups, for children, or for highly sensitized
encountered and treated, but the kidney units were affected patients. Such patients had been recognized by the new lym-
by a hepatitis B epidemic in the mid-1960s, which affected phocytotoxicity testing using a crossmatch between donor
morale and status. The narrow age limit for transplantation cells and recipient serum. First noted by Terasaki and associ-
was widened, and in Richmond the first experience with ates49 and described in more detail by Kissmeyer-Nielsen and
kidney grafts in children was obtained. colleagues50 in 1966 and Williams and colleagues,51 such
Recipients of kidney transplants reentered the normal pretransplant testing explained cases of sudden failure and
business of life and became politicians, professors, pilots, led to a marked diminution in hyperacute rejection.
and fathers and mothers of normal children. Other good
news in the United States came when the federal govern-
ment accepted the costs of regular dialysis and transplan- The 1970s Plateau
tation in 1968. There were always unexpected findings,
usually reported from the pioneer units with the longest The 1970s was a period of consolidation, of improvements
survivors. Cautiously, second kidney transplants were in data collection such as the valuable European Dialysis
performed at Richmond when a first had failed; these did and Transplant Association surveys, and increased sophisti-
well, and the matter became routine. Chronic rejection cation in HLA typing methods and organ-sharing schemes.
and malignancy first were reported in kidney transplant Cadaver organ procurement generally increased as a result
recipients from Denver. As a result of the optimism, experi- of wider involvement of the public and medical profession,
mental heart transplantation started, the first human livers although the number of patients waiting for transplanta-
were grafted, and there was a revival of interest in xeno- tion persistently exceeded the organs available, and dona-
transplantation. Although the attempts of Reemtsma and tion declined transiently during times of public concern
coworkers,40 Hume,41 and Starzl39 at transplantation with over transplantation issues. Governments took initiatives
chimpanzee or baboon kidneys ultimately failed, rejec- to increase donations; in the United Kingdom, the Kidney
tion did not occur immediately, and the cases were studied Donor Card was introduced in 1971, becoming a multi-
closely and described. donor card 10 years later. In hospital practice, methods
In the search for better immunosuppression, there was of resuscitation and intensive care improved, and the con-
great excitement when laboratory studies by Woodruff and cept of brain death was established to prevent prolonged,
1 • Kidney Transplantation: A History 7

pointless ventilation, although its immediate application to natural antibody from recipient plasma were tried to deal
transplantation provoked controversy. Despite many new with the hyperacute phase of xenograft organ rejection.
claims for successful methods of immunosuppression, such Although the traditional hopes for xenografting of human
as trials of splenectomy, thymectomy, thoracic duct drain- patients had assumed that “concordant” species such as
age, and a new look at cyclophosphamide, no agent except the monkey would be used, a new strategy using genetic
antithymocyte globulin became established in routine use. engineering methods first used a line of transgenic pigs, a
Although patient survival after kidney transplantation distant species discordant with humans, with a modified
continued to increase, the 1970s did not show the expected endothelium that reduced the complement-mediated imme-
increase in cadaver graft survival. Some groups reported diate reaction.59 Hopes continue that these early develop-
decreased survival figures; this paradox was solved partly ments will evolve into a sophisticated successful routine.60
by the demonstration that blood transfusion during regular Meanwhile, the kidney transplanters can only watch, with
dialysis, which had been discouraged because of the risk of detached interest, the emergence of stem cell use in cellular
sensitization, was beneficial to the outcome of kidney trans- transplantation.
plantation,52 an observation made some years earlier by These new hopes for xenografts raised old fears among
Morris and coworkers.53 the public and legislators, notably regarding disease trans-
The 1970s ended with two innovations that revived mission. Although this had been a familiar problem in
hopes of reaching the goal of routine, safe, and successful human-to-human transplantation and had been met regu-
kidney transplantation. Ting and Morris54 reported the suc- larly and dealt with, governments required reassurances
cessful clinical application of HLA-DR matching, and Calne about xenotransplantation with the added threat of retro-
and associates55 revived memories of the excitement of the virus transmission.
early days of the use of azathioprine by introducing into
clinical practice the first serious rival to it in 20 years, cyclo-
sporine, which had been discovered to be a powerful immu- Conclusion
nosuppressive agent by Borel.56 Cyclosporine replaced the
earlier drug regimens and was the dominant agent in use Kidney transplantation was the first of the organ transplant
until the 1990s. Transplantation had grown to a suffi- procedures to develop because cadaveric donor kidneys
ciently large clinical service that it was worth the attention revived with time, the availability of live donors increased,
of the pharmaceutical companies, and in the 1990s steady and the crucial backup of dialysis was implemented. When
production of new agents occurred—tacrolimus, mycophe- radical new ideas are to be tested, pioneers still turn to kid-
nolate mofetil, rapamycin, FTY720, brequinar, and others. ney transplantation. Kidney transplantation is where it all
Any drug with promise was marketed aggressively, and started, with good reason, and it will always be a test bed
sponsored trials became a routine part of clinical life. for major innovation, including laparoscopic and robotic
The improved results of transplantation meant that the surgery.
shortage and procurement of organs became a more domi- Nowhere is the excitement of the early days reflected bet-
nant issue. Living donors were encouraged, to which were ter than in the recollections of 35 of the pioneers of trans-
added occasional altruistic donations, with use later of “kid- plantation gathered together by Terasaki.61
ney chains” to pass on locally incompatible organs. There
was a return to using possibly damaged “marginal” kid- References
neys and organs removed rapidly after cardiac death (DCD). 1. Hamilton D. Organ transplantation: a history. Pittsburgh: Pittsburgh
Comparisons of transplantation practice throughout the University Press; 2012.
world showed remarkable differences in attitudes to use of 2. Ullmann E. Experimentelle nierentransplantation. Wien Klin Woche­n­
schr 1902;15:281. [For a biography of Ullmann, see Lesky E. Die erste
live related donors and cadaver organs, depending on reli- Nierentransplantation: Emerich Ullmann (1861–1937). Munch Med
gion and cultural traditions. Kidney transplantation had Wochenschr 1974;116:1081.]
started as a difficult surgical and scientific challenge con- 3. von Decastello A. Experimentelle nierentransplantation. Wien Klin
fined to a few academic centers in the developed world, but Wochenschr 1902;15:317.
its success had led to the technique becoming a routine ser- 4. Carrel A. La technique operatoire des anastomoses vasculaires et la
transplantation des viscères. Lyon Med 1902;98:859.
vice in all parts of the world.57 In some nations not sharing 5. Hamilton D. The first transplant surgeon: the flawed genius of nobel
Western attitudes, the donor shortage meant the appear- prize winner, Alexis Carrel. Singapore: World Scientific; 2017.
ance of undesirable commercial developments in renal 6. Jaboulay M. Greffe de reins au pli du coude par soudure arte. Bull Lyon
transplantation, such as the purchase of kidneys from living Med 1906;107:575. [For a biography of Jaboulay, see Biogr Med Paris
1936;10:257.]
unrelated donors (discussed in more detail in Chapter 41).58 7. Ullmann E. Tissue and organ transplantation. Ann Surg 1914;60:195.
8. Unger E. Nierentransplantation. Berl Klin Wochenschr 1909;1:1057.
9. Winkler FA. Ernst Unger: a pioneer in modern surgery. J Hist Med
Waiting for Xenografts Allied Sci 1982;37:269.
10. Carrel A. The transplantation of organs. New York Times 1914.
11. Voronoy YY. Sobre el bloqueo del aparato reticulo-endothelial. Siglo
As the demand for kidney transplants continued to exceed Med 1936;97:296.
supply, other initiatives appeared and included study of 12. Hamilton D, Reid WA. Yu Yu Voronoy and the first human kidney
nations and areas with high donation rates (e.g., Spain). As allograft. Surg Gynecol Obstet 1984;159:289.
all attempts to increase donor supply fell short of the ever- 13. Matevossian E, Kern H, Hüser N, et al. Surgeon Yurii Voronoy (1895–
1961) — a pioneer in the history of clinical transplantation. Transpl
rising target, the radical alternative of the use of animal Int 2009;22:1132.
organs was examined afresh. Profound immunosuppres- 14. Moore FD. Give and take: the development of tissue transplantation.
sion alone was ineffective and, at first, methods of removing Philadelphia: WB Saunders; 1964.
8 Kidney Transplantation: Principles and Practice

15. Simonsen M. Biological incompatibility in kidney transplantation 39. Starzl TE. Personal reflections in transplantation. Surg Clin North Am
in dogs: serological investigations. Acta Pathol Microbiol Scand 1978;58:879.
1953;32:1. 40. Reemtsma K, McCracken BH, Schlegel JU, et al. Renal heterotrans-
16. Dempster WJ. The homotransplantation of kidneys in dogs. Br J Surg plantation in man. Ann Surg 1964;160:384.
1953;40:447. 41. Hume DM. Discussion. Ann Surg 1964;160:409.
17. Küss R, Teinturier J, Milliez P. Quelques essais de greffe du rein chez 42. Wolstenholme GEW, O’Connor M, editors. Antilymphocytic serum.
l’homme. Mem Acad Chir 1951;77:755. London: J&A Churchill; 1967.
18. Servelle M, Soulié P, Rougeulle J, et al. Greffe d’une reine de supplicie 43. French ME, Batchelor JR. Immunological enhancement of rat kidney
à une malade avec rein unique congénital, atteinte de nephrite chro- grafts. Lancet 1969;2:1103.
nique hypertensive azotémique. Bull Soc Med Hop Paris 1951;67:99. 44. Dausset J. The challenge of the early days of human histocompatibil-
19. Dubost C, Oeconomos N, Vaysse J, et al. Resultats d’une tentative de ity. Immunogenetics 1980;10:1.
greffe rénale. Bull Soc Med Hop Paris 1951;67:1372. 45. Hamburger J, Vaysse J, Crosnier J, et al. Renal homotransplantation in
20. Michon L, Hamburger J, Oeconomos N, et al. Une tentative de trans- man after radiation of the recipient. Am J Med 1962;32:854.
plantation rénale chez l’homme. Presse Med 1953;61:1419. 46. van Rood JJ. Histocompatibility testing. Copenhagen: Munkgaard;
21. Hume DM, Merrill JP, Miller BF, et al. Experiences with renal homo- 1967.
transplantation in the human: report of nine cases. J Clin Invest 47. Belzer FO, Ashby BS, Dunphy JS. 24-Hour and 72-hour preservation
1955;34:327. of canine kidneys. Lancet 1967;2:536.
22. Murray JE, Merrill JP, Harrison JH. Kidney transplantation between 48. Collins GM, Bravo-Shugarman M, Terasaki PI. Kidney preservation
seven pairs of identical twins. Ann Surg 1958;148:343. for transportation: initial perfusion and 30 hours’ ice storage. Lancet
23. Murray JE, Tilney NL, Wilson RE. Renal transplantation: a twenty-five 1969;2:1219.
year experience. Ann Surg 1976;184:565. 49. Terasaki PI, Marchioro TL, Starzl TE. In: Amos DB, van Rood JJ, edi-
24. Murray JE, Merrill JP, Dammin GJ, et al. Study of transplantation tors. Histocompatibility testing. Washington, DC: National Academy
immunity after total body irradiation: clinical and experimental inves- of Sciences; 1965:83.
tigation. Surgery 1960;48:272. 50. Kissmeyer-Nielsen F, Olsen S, Peterson VP, et al. Hyperacute rejection
25. Hamburger J, Vaysse J, Crosnier J, et al. Transplantation of a kidney of kidney allografts. Lancet 1966;2:662.
between non-monozygotic twins after irradiation of the receiver: good 51. Williams GM, Hume DM, Hudson RP, et al. Hyperacute renal-
function at the fourth month. Presse Med 1959;67:1771. homograft rejection in man. N Engl J Med 1968;279:611.
26. Küss R, Legraine M, Mathe G, et al. Prémices d’une homotransplanta- 52. Opelz G, Sengar DPS, Mickey MR, et al. Effect of blood transfusions on
tion rénale de soeur à frère non jumeaux. Presse Med 1960;68:755. subsequent kidney transplants. Transplant Proc 1973;5:253.
27. Schwartz R, Dameshek W. Drug-induced immunological tolerance. 53. Morris PJ, Ting A, Stocker J. Leucocyte antigens in renal transplanta-
Nature 1959;183:1682. tion, I: the paradox of blood transfusions in renal transplantation. Med
28. Schwartz RS. Perspectives on immunosuppression. In: Hitchings GH, J Aust 1968;2:1088.
editor. Design and achievements in chemotherapy. Durham, NC: 54. Ting A, Morris PJ. Matching for B-cell antigens of the HLA-
Burroughs Wellcome; 1976. p. 39–41. DR (D-related) series in cadaver renal transplantation. Lancet
29. Calne RY. The rejection of renal homografts: inhibition in dogs by 1978;1:575.
6-mercaptopurine. Lancet 1960;1:417. 55. Calne RY, White DJG, Thiru S, et al. Cyclosporin A in patients receiv-
30. Zukoski CF, Lee HM, Hume DM. The effect of 6-mercaptopurine on ing renal allografts from cadaver donors. Lancet 1978;2:1323.
renal homograft survival in the dog. Surg Forum 1960;11:47. 56. Borel JF. Comparative study of in vitro and in vivo drug effects on cell
31. Calne RY. The development of immunosuppressive therapy. Trans- mediated cytotoxicity. Immunology 1976;31:631.
plant Proc 1981;13:44. 57. Burdick JF, DeMeester J, Koyama I. Understanding organ procure-
32. Calne RY, Alexandre GPJ, Murray JE. The development of immuno- ment and the transplant bureaucracy. In: Ginns LC, Cosimi AB, Mor-
suppressive therapy. Ann NY Acad Sci 1962;99:743. ris PJ, editors. Transplantation. Boston: Blackwell; 1999. p. 875–94.
33. Hopewell J, Calne RY, Beswick I. Three clinical cases of renal trans- 58. Morris PJ. Problems facing the society today. Transplant Proc
plantation. BMJ 1964;1:411. 1987;19:16.
34. Küss R, Legraine M, Mathe G, et al. Homologous human kidney trans- 59. van den Bogaerde J, White DJG. Xenogeneic transplantation. Br Med
plantation. Postgrad Med J 1962;38:528. Bull 1997;53:904.
35. Murray JE, Merrill JP, Harrison JH, et al. Prolonged survival of human 60. D’Apice A, Cowan PJ. Gene-modified pigs. Xenotransplantation
kidney homografts by immunosuppressive drug therapy. N Engl J Med 2008;15:87.
1963;268:1315. 61. Terasaki PI. History of transplantation: thirty-five recollections. Los
36. Starzl TE, Marchioro TL, Waddell WR. The reversal of rejection in Angeles: UCLA Tissue Typing Laboratory; 1991.
human renal homografts with subsequent development of homograft 62. Dausset J. Iso-leuco-anticorps. Acta Haematol (Basel) 1958;20:156.
tolerance. Surg Gynecol Obstet 1963;117:385. 63. Oriol R, Ye Y, Koren E, Cooper DK. Carbohydrate antigens of pig tis-
37. Goodwin WE, Mims MM, Kaufman JJ. Human renal transplant, III: sues reacting with human natural antibodies as potential targets for
technical problems encountered in six cases of kidney homotrans- hyperacute vascular rejection in pig-to-man organ xenotransplanta-
plantation. Trans Am Assoc Genitourin Surg 1962;54:116. tion. Transplantation 1993;56:1433–42.
38. Hume DM, Magee JH, Kauffman HM, et al. Renal homotransplanta- 64. Rosales A, Salvador JT, Urdaneta G, Patiño D, Montlleó M, Esquena S,
tion in man in modified recipients. Ann Surg 1963;158:608. et al. Laparoscopic kidney transplantation. Eur Urol 2010;57:164–7.
2 The Immunology of
Transplantation
TOMAS CASTRO-DOPICO and MENNA R. CLATWORTHY

CHAPTER OUTLINE Introduction T Cell-Mediated Rejection


Activation of the Immune System Migration of Activated Cells Into the Graft
Peritransplant Mechanisms of Cytotoxicity
Damage Signals and Their Receptors Complement and TCMR
Soluble Innate Immunity – Complement B Cell Activation and Antibody-Mediated
Innate Immunity – Cellular Components Rejection
Neutrophils B Cell Activation
Macrophages B Cell Function
NK Cells and Innate Lymphoid Cells Alloantibodies
Cells at the Interface Between Innate and Antigen Presentation to CD4 T Cells
Adaptive Immunity Formation and Maintenance of Secondary
Stimulation of Adaptive Alloimmunity Lymphoid Tissue
ABO Blood Group Antigens Production of Proinflammatory Cytokines
HLA Molecules B Cells as Regulators of the Immune
Major Histocompatibility Antigens Response
Minor Histocompatibility Antigens Antibody-Effector Function in ABMR
Antigen Presentation Direct Stimulation of Endothelial MHC
Dendritic Cells Complement Activation
Direct, Indirect, and Semidirect Antigen FcγR Activation
Presentation Transplant Tolerance
T Cell Activation Factors Influencing Rejection Beyond the
Signal 1—TCR Activation Graft—The Microbiome
Signal 2—Costimulation Conclusion
Signal 3—Cytokines

Introduction underpinned by a greater understanding of the basic biol-


ogy of these important systems. Of note, attempts to under-
Solid organ transplant requires the removal of an organ stand the immune response to an allograft have historically
from one individual, the donor, and its placement in the relied on rodent and nonhuman primate models. Although
recipient. Whether the donor is living or deceased, this pro- useful, such studies do not always accurately reflect the
cess inevitably requires a temporary cessation of circulation alloimmune response in humans, and there is an increasing
and hence oxygenation, with attendant cellular dysfunction emphasis on the need for experimental medicine studies in
and damage. Thus when the blood supply is restored to the transplantation to enable advances in genomic, transcrip-
allograft in the recipient, and the recipient’s immune sys- tomic, and proteomic technologies to be harnessed toward
tem can access the transplant, there are broadly two main this goal.1 In this chapter, we will provide a description of
stimuli that may be recognized: damage-associated signals the various arms of the immune system and consider how
that activate the innate immune system, and differences in they contribute to the immune response to transplanted
cell surface molecules (such as human leukocyte antigens organs (Fig. 2.1).
[HLA] or blood group antigens) between donor and recipi-
ent that can activate the adaptive immune system. In the
past 50 years, the increased understanding of cellular adap- Activation of the Immune System
tive immunity has transformed our ability to suppress this Peritransplant
arm of the immune system, such that T cell-mediated rejec-
tion (TCMR) is now uncommon, occurring in less than 20% During the process of organ retrieval and reimplantation, there
of kidney transplant recipients, for example. However, the is an inevitable period of ischemia. The cessation of oxygen
control of innate and humoral adaptive immunity remains supply renders the cells unable to generate sufficient energy
challenging, and efforts to achieve this will need to be to continue homeostatic processes that maintain cellular
9
10 Kidney Transplantation: Principles and Practice

integrity, leading to damage or even death of some cells. This


cellular damage or death is associated with the release of mol-
Allograft rejection timeline
ecules that can be detected by both the innate and adaptive
immune system. During organ reperfusion, it is the innate
1 year 20+ years immune system that is principally activated. This ancient
system includes a soluble arm—the complement system and
a variety of opsonins that have evolved to facilitate pathogen
Pathology IRI TCMR aABMR cABMR
recognition, for example, C-reactive protein (CRP), comple-
ment activation products (C3b), natural immunoglobulin (Ig)
Innate M antibody, and a cellular arm, composed of phagocytes and
Complement innate lymphoid cells, including natural killer (NK) cells.
Immune response

Phagocytes
NK cells
DAMAGE SIGNALS AND THEIR RECEPTORS
Adaptive
CD4 T cells
The innate immune system has evolved to recognize mole-
CD8 T cells cules expressed by pathogens, known as pathogen-associated
B cells molecular patterns (PAMPs), including specific carbohydrates,
(non-antibody) lipopolysaccharide (LPS), flagellin, lipoteichoic acid, and double-
IgG stranded ribonucleic acid (RNA). This is achieved by an array of
Fig. 2.1 Timeline of allograft rejection. During transplantation and receptors, so-called pattern recognition receptors (PRRs), some
beyond, allografts are subjected to a variety of stresses that cause graft of which are surface bound and survey the extracellular envi-
damage and destruction. Indicated in the schematic shown here is a ronment, and some of which are located within the cell, in the
timeline of the pathologic insults potentially experienced by a trans- cytoplasm or endosomal compartments (Fig. 2.2). PRRs include
planted organ and the types of immune cells that contribute to each
pathology, stratified based on innate and adaptive immunity. aABMR,
cell-associated receptors, such as toll-like receptors (TLRs),2
Acute antibody-mediated rejection; cABMR, chronic antibody-mediated retinoic acid inducible gene-1–like receptors,3 and nucleotide-
rejection; IgG, immunoglobulin G; IRI, isch­emia reperfusion injury; NK, binding oligomerization domain (NOD)-like receptors,4 and sol-
natural killer; TCMR, T cell-mediated rejection. uble molecules, including CRP, ficolins, and mannan-binding
PAMP
DAMP DAMPs
(HMGB1...) (ATP...)

Extracellular

Cytoplasm PHAGOCYTE
Receptors
TLR1/2/4/5/6
(P2RX7...)
Endosome Cytosolic DNA sensors
RIG-I-like receptors
NOD-like receptors

TLR3/7-8/9
Inflammasome

NLRP3
IL-1β, TNF, CXCL8... ASC
CASP1

Active
Pro-IL-1β caspase IL-1β

TNF
CXCL8...
Fig. 2.2 Mechanisms of immune sensing and inflammatory cytokine production during sterile inflammation. Schematic of the different classes
of pattern recognition receptors and their potential to response to danger-associated molecular patterns (DAMPs) released during sterile inflammation
and ongoing allograft destruction. For example, high mobility group box 1 (HMGB1) binds to toll-like receptors (TLRs) to induce expression of inflamma-
tory mediators, while extracellular adenosine triphosphate (ATP) can engage cell surface receptors, such as P2RX7, leading to inflammasome assembly
(e.g., the nucleotide-binding domain leucine-rich repeat contain­ing protein 3 (NLRP3) inflammasome), caspase activation, and mature interleukin (IL)-1β
and IL-18 production. These mechanisms are particularly important in innate immune cells, such as macrophages and neutrophils.
2 • The Immunology of Transplantation 11

lectin (MBL).5 Matzinger first proposed that the immune system mannose-binding pathway is activated by carbohydrates pres-
may have the capacity to respond to damage signals, even in ent on pathogens or by damaged endothelium. The net result
the absence of microbes—the danger hypothesis—and may of activating any of the three pathways is the formation of a C3
have even evolved in response to these stimuli.6 It is now clear convertase (either C4bC2a or C3bBb), which cleaves C3. The
that many cell-damage or death-associated signals (termed dan- resulting C3b cleaves C5 and activates a final common path-
ger-associated molecular patterns [DAMPs]) are recognized by the way resulting in MAC formation. Complement activation also
same PRRs that mediate responses to PAMPs.7 These DAMPs leads to the formation of anaphylatoxins (C3a and C5a), which
include extracellular adenosine triphosphate (ATP),8 hyaluro- activate neutrophils and mast cells, promoting inflammation.
nan,9 uric acid, heat-shock proteins (HSPs), and high-mobility In addition, C3b can opsonize pathogens for uptake by comple-
group box 1 (HMGB1).10 These molecules are normally hidden ment receptors CR1 and CR3 on phagocytes and can activate B
from the immune system or are derived from degradation prod- cells; the latter may promote B cell activation in transplantation.
ucts of extracellular matrix components generated during isch- Because the alternative pathway is continuously acti-
emia reperfusion injury (IRI) and inflammation.11 Similarly, vated, effective regulation is critical to prevent inappropriate
falling intracellular potassium and oxidative stress can act as activation. Regulatory proteins may be circulating or mem-
intracellular danger signals.12–14 brane-bound. Circulating inhibitors include C1 esterase
inhibitor and factors H and I. Membrane-bound regulatory
proteins include membrane cofactor protein (MCP), CD55
SOLUBLE INNATE IMMUNITY—COMPLEMENT
(decay accelerating factor [DAF]), and CD59 (protectin).
The complement system is a series of protein kinases that Defects or mutations in complement regulatory proteins
are sequentially activated and culminate in the formation can result in severe renal pathology, for example, atypi-
of the membrane attack complex (MAC).15,16 The MAC cal hemolytic uremic syndrome (HUS), which can recur in
comprises complement components C5 to C9, which are the transplanted allograft.17,18 The C3 glomerulopathies,
inserted into the cell membrane (pathogen or host), disrupt- including dense deposit disease and type I and III mesan-
ing integrity and causing cell lysis (Fig. 2.3). In addition, giocapillary glomerulonephritides, are also underpinned
many proximal complement components may augment the by complement mutations.19,20 Small case series suggest a
immune response to the allograft. recurrence rate of around 60% in the transplanted organ.21
The complement system may be activated by three path- The C5 inhibitor eculizumab may well have efficacy in both
ways: the classical pathway, the alternative pathway, and the primary and recurrent forms of these diseases.22
MBL pathway. IgM or IgG immune complexes activate the The endothelial cell damage associated with ischemia-
classical pathway, and hence this pathway may become acti- reperfusion injury during transplantation leads to MBL
vated during antibody-mediated rejection (see section on B Cell and alternative complement pathway activation.23 Histo-
Activation). The alternative pathway is constitutively active logic evidence of complement activation (C3d deposition)
and must be controlled by a series of regulatory proteins. The is present in animal models and in human kidneys with

PATHWAY Classical MBL Alternative


Endothelial damage Endothelial damage
ACTIVATION Alloantibody
(ischemia) (ischemia)

C1 MBL-MASP1-MASP2

C3 Factor D

C4, C2 C4bC2a C3bBb Factor B

C3a C3b CR1-4 B cell activation

C5 C5b MAC ABMR

Neutrophil
C3aR/C5aR C5a recruitment and IRI
APC maturation degranulation
TCMR T cell activation
Fig. 2.3 The complement cascade. Activation of the complement cascade can contribute to tissue destruction directly via formation of the mem-
brane attack complex (MAC) and through complement receptor engagement and subsequent immune cell activation. This includes the maturation of
antigen-presenting cells and neutrophil activation. Three pathways are possible for complement activation and each may contribute to tissue destruc-
tion in rejection. The classical pathway is mediated by antibodies and may be driven by donor-specific IgG and IgM immune complexes during ABMR,
whereas activation of the alternative and MBL pathways results from damage to allograft endothelial cells as a result of IRI. All pathways converge on
the cleavage of C3 and the generation of the C5-cleaving fragment C3b and the anaphylatoxin C3a. Subsequently, C5a and C5b act as an anaphylatoxin
that binds to C5aR and a component of the MAC, respectively, leading to cell death.
12 Kidney Transplantation: Principles and Practice

acute tubular necrosis (ATN).24 Factor B-deficiency and a complex multistep process requiring a series of interactions
factor B-blocking antibody are protective in a murine model between the surface of the leukocyte and the endothelial
of IRI,24,25 suggesting alternative pathway involvement. cell or its extracellular matrix.41,42 The proteins involved
Biopsies in murine and human kidneys with ATN also dem- fall into three groups: the selectins, and members of the
onstrate MBL deposition,26 likely triggered by endogenous integrin and Ig superfamilies. Initial interaction and rolling
ligands expressed by dying cells, and MBL-deficient mice of neutrophils along the endothelium allow the leukocyte
are protected from IRI.27 Transplantation of a kidney from to sample the endothelial environment, while maintaining
a C3-deficient mouse into a C3-sufficient recipient results in its ability to detach and travel elsewhere. This step is largely
significant attenuation of IRI, in contrast to the reciprocal controlled by the selectins, although α4 integrins may also
transplant, suggesting that local C3 production in the kid- play a role. Endothelial cells express interleukin (IL)-8 and
ney rather than circulating C3 is the major player in IRI.28 platelet-activating factor, which induces strong neutrophil
In human kidneys, cold ischemia may alter the methylation adhesion. This interaction leads to signaling to the neutro-
state of the C3 promoter, resulting in increased local expres- phil, slowing and arresting the rolling process. Shedding
sion of C3 after reperfusion,29 which is associated with a of L-selectin by leukocytes allows their detachment and
diminished graft survival.30 Silencing of the gene encoding extravasation.43 The latter stages of leukocyte transmigra-
C3 using small interfering RNA (siRNA) has been shown to tion are regulated mainly by the β2 integrins and adhesion
reduce C3 expression, histologic and biochemical param- proteins of the immunoglobulin superfamily.
eters of kidney injury, and mortality in an animal model The expression of adhesion proteins involved in these
of IRI.31 The terminal pathway products C5a and C5b–C9 interactions is upregulated by proinflammatory cytokines.
appear to be critical in mediating cellular injury.32–34 A Ischemic damage alone results in increased expression of
C5-blocking antibody and C5a receptor antagonist have both several cytokines that upregulate the expression of selec-
been shown to abrogate IRI35 and gene silencing of the C5a tins.44,45 Other adhesion proteins, such as intercellular
receptor also protects mice from IRI.36 Gene silencing may adhesion molecule (ICAM)-1 and vascular cell adhesion
provide a promising tool in renal transplantation, because molecule (VCAM)-1 of the immunoglobulin superfamily
siRNA-to-complement components might be applied to the and E-selectin (endothelial-specific selectin), are upregu-
allograft during cold storage, before implantation.37 Simi- lated by cytokines induced by donor brain death46 and
larly, other strategies to inhibit local complement activation implantation.
may have utility in limiting allograft IRI.38,39 Ongoing clini- After exit from the vasculature, neutrophil PRR engage-
cal trial in renal transplantation to prevent IRI and delayed ment by DAMPs can induce the production of reactive oxy-
graft function include the use of C1 esterase inhibitors gen species, hydrolytic enzymes, and cytokines,47,48 with
(https://clinicaltrials.gov/ct2/show/NCT02134314) and graft neutrophilia linked to alloreactive T cell responses and
the C5 inhibitor eculizumab (https://clinicaltrials.gov/ct2/ disease activity in mouse models.49–51 Neutrophils can also
show/NCT02145182). However, although there was a undergo a form of programmed cell death known as NETo-
reduced rate of delayed graft function in a small pediatric sis, whereby activated neutrophils form so-called extracel-
trial (n = 57) using eculizumab in kidney transplantation, lular traps (NETs).52 These have been observed in human
there was an unexpectedly high rate of graft loss because of lung transplant recipients and in mouse models of allograft
thrombosis in eculizumab-treated subjects,40 necessitating IRI, although how they contribute to inflammation remains
caution in its future use in this context. controversial.53 Perhaps less appreciated is the potential
role of neutrophils in the resolution of inflammation in allo-
immunity: efferocytosis of apoptotic neutrophils leads to the
INNATE IMMUNITY—CELLULAR COMPONENTS production of antiinflammatory mediators, such as IL-10,
Cellular innate immunity comprises a variety of hema- while proresolving factors, including lipoxins and resolvins,
topoietic myeloid and lymphoid cells, often poised within are important in wound healing and may suppress ongoing
tissues for the rapid nonspecific detection of invading rejection.54
microorganisms and transformed cells. However, innate Macrophages
immunity also encompasses various nonhematopoietic
cells, such as the gastrointestinal, respiratory, and urogen- Tissue-resident macrophages represent the major innate
ital epithelium, which, in addition to forming a physical leukocyte population in most tissues.55 Through their
barrier, also express PRRs and orchestrate local immunity widespread expression of PRRs, these sentinel cells are
(Fig. 2.4). specialized in antigen phagocytosis and cytokine produc-
tion, being key drivers of inflammation in numerous set-
tings. During inflammatory conditions, the macrophage
Neutrophils pool is further reinforced by recruited monocytes from the
Although often viewed as nonspecific effector cells, granu- bloodstream, with several macrophage- and monocyte-
locytes, such as neutrophils and eosinophils, are likely to derived cytokines capable of contributing to tissue dam-
play a significant role in transplant pathology through their age.56 For example, tumor necrosis factor (TNF)α can
potent effector functions and rapid recruitment to sites of drive cellular necroptosis and the concomitant release
inflammation during IRI and rejection. It is also increas- of intracellular contents and DAMPs,57,58 in addition to
ingly appreciated that there may be tissue-resident popula- augmenting angiogenesis, matrix metalloprotease (MMP)
tions within a variety of organs. production, immune cell activation, and germinal center
Neutrophils are the dominant circulating phagocyte in formation, all with particular importance in the context
humans, and their recruitment into the graft involves a of transplantation.59 Furthermore, via production of IL-1β
2 • The Immunology of Transplantation 13

Epithelium Mononuclear phagocytes

Tissue Tissue Blood


dendritic cells macrophages monocyte

Physical barrier
Antimicrobial peptides Phagocytosis
Cytokine production Cytokine production
Mucus Antigen presentation

Granulocytes Lymphocytes

Blood Tissue granulocytes NK cells Helper ILCs


neutrophils (e.g., eosinophils) (CD56bright and CD56lo) (ILC1–3)

Phagocytosis Cytotoxicity
Microbicidal molecules Cytokine production
Cytokines/chemokines Antigen presentation
Histamine and eicosanoids
Fig. 2.4 The innate immune system. A schematic of the major components of the innate immune system and a brief summary of their respective
functions in inflammation and homeostasis. Epithelial cells form the physical barriers of mucosal gastrointestinal, respiratory, and urogenital tracts,
and play a critical role in host-microbe interactions at these environmental interfaces. Mononuclear phagocytes represent tissue-resident dendritic
cells and macrophages in addition to those recruited to tissues during inflammation. In most circumstances, these cells play essential roles in tissue
homeostasis, but can also prime alloreactive adaptive immune responses, are major sources of inflammatory mediators, and participate directly in tis-
sue destruction. Granulocytes include neutrophils, normally circulating in the blood but rapidly recruited to sites of inflammation, and tissue-resident
cells, such as eosinophils. The workhorses of innate immunity, they are increasingly appreciated for their roles in influencing adaptive immunity within
tissues and lymphoid organs. Innate lymphocytes encompass the well-characterized NK cells, which participate in receptor-mediated cell lysis and IFNγ
production, and the recently discovered class of helper innate lymphoid cells. Helper ILCs are enriched at mucosal surfaces and are potent sources of T
cell-associated cytokines, and also serve as APCs through expression of MHC class II molecules.

and IL-8, macrophages play a strategic role in the recruit- is provided by DAMP receptors, for example the ATP
ment of neutrophils to inflamed sites through the induc- receptor, P2X7R.
tion of adhesion molecules on endothelial cells and direct IL-1 plays a pivotal role in initiating and amplifying
chemotactic activity, respectively.60 sterile inflammation, as evidenced by experiments show-
Endogenous ligands with the capacity to engage mac- ing that mice deficient in the IL-1 receptor (IL-1R) or in the
rophage PRRs are generated during transplantation, adaptor protein MyD88 (which is required for IL-1R signal-
either through IRI or as a consequence of ongoing rejec- ing), demonstrate minimal neutrophilic inflammation after
tion.61 Detection of DAMPs leads to the association of challenge with necrotic cells.62 IL-1β has multiple actions,
nucleotide-binding domain leucine-rich repeat contain- including stimulation of nonhematopoietic cells to produce
ing protein (NLRP)3 with apoptosis-associated speck-like the neutrophil chemoattractants chemokine (C-X-C motif)
protein (ASC), and recruitment of procaspase-1, forming a ligand (CXCL)2 (also known as macrophage inflammatory
complex known as the NLRP3 inflammasome48,62,63 (Fig. protein [MIP]-2) and CXCL1 (also known as keratinocyte
2.3). Inflammasome activation results in the cleavage of chemoattractant [KC]).65 DAMPs may also act directly
procaspase-1 to caspase-1, which subsequently cleaves as chemotactic agents for neutrophils.47,66 IL-1β also
IL-1β and IL-18 from their precursors. Of note, in vitro increases the expression of cell adhesion molecules (e.g.,
data suggests that in macrophages, inflammasome acti- ICAM-1 [CD54]) on endothelial cells.67 ICAM-1 interacts
vation is a two-step process. First, macrophages must be with integrins (CD11 and CD18) on neutrophils and mono-
“primed” by TLR stimuli, resulting in NFγB-dependent cytes to promote endothelial adherence and subsequent
pro-IL-1β production and upregulation of NLRP3 expres- entry into tissues.
sion. A number of DAMPs are thought to signal via TLRs; There is a significant body of evidence that suggests
for example, HMGB1 activates TLR4.64 The second signal that sterile inflammation contributes to the severity of IRI;
14 Kidney Transplantation: Principles and Practice

neutralization of the DAMP HMGB1 with a monoclonal NK effector activity,91,92 although more recently CD28-
antibody attenuates renal injury after IRI, whereas recom- independent rejection in mouse models of transplanta-
binant HMGB1 exacerbates it.68 Some DAMPs stimulate tion has been shown to be NK dependent and sensitive
TLRs, and mice deficient in TLR-2 and TLR-4 are protected to blockade of NKG2D.93 The activating receptor NKG2D
from IRI with a reduction in neutrophil and macrophage is engaged by MHC class I polypeptide-related sequence
infiltration.69,70 Furthermore, NLRP3, ASC, and caspase-1 (MIC) A (MICA) and MICB, that are induced in allografts
deficient mice are protected from renal ischemic injury.71–73 during acute and chronic rejection.94 The binding of these
Pharmacologic inhibition of caspase-1 has been shown ligands to NKG2D activates NK cells to enhance effector
to reduce renal IRI74 and may therefore be a viable thera- functions, whereas the engagement of killer immunoglob-
peutic strategy in transplantation. In rodent models, treat- ulin-like receptors (KIRs) by KIR ligands such as HLA-C
ment with monoclonal antibodies directed against ICAM-1, (KIR2DL1 and KIR2DL2) and HLA Bw4 (KIR3DL1) gener-
CD11a, or CD11b also protect against IRI.75–77 In a human ally inhibit function. Genetic studies of donor and recipi-
phase I trial, ICAM blockade using a murine antibody BIRR1 ent HLA-C type (grouped as C1 and C2 depending on
was associated with a reduction in delayed graft function polymorphisms at position 77 and 80 and which seem to
in renal transplant recipients.78 However, a randomized exhibit differential NK cell inhibition) suggest that long-
controlled trial of anti-ICAM-1 antibody in renal transplan- term outcomes may be influenced by donor or recipient
tation failed to demonstrate any significant improvement interaction with KIRs.86,87 This has also been observed
in delayed graft function.79 Blockade of another adhesion when KIR HLA mismatches are analyzed in HLA-compat-
molecule, P-selectin, also attenuates leukocyte recruitment ible transplantation.95
and IRI in rodent models80,81 and in humans.82 Inhibitory receptor function underlies the phenomenon
Innate cells may also drive an adaptive alloimmune of responses to “missing self,”96,97 which contributes to
response. In TCMR, macrophages may act as antigen-pre- tumor immunity,98 the killing of stem cells,99 and hybrid
senting cells (APCs), and the IRI-associated inflammation resistance in experimental models of transplantation.88
may induce upregulation of major histocompatibility com- Beyond NK cells, another class of innate lymphocyte are
plex (MHC) class II (MHC-II) on resident cells, augmenting the recently described “helper” innate lymphoid cells (ILCs).
their antigen-presenting functions. In antibody-mediated The subject of intense research in the last decade, ILCs are
rejection (ABMR), IgG and complement are deposited in characterized by their similarity to helper T (Th) cell sub-
peritubular capillaries, facilitating monocyte, macrophage, sets, with the notable absence of somatically recombined
and neutrophil activation via their Fcγ receptors (FcγR) antigen-specific receptors or classical lineage markers.100
and complement receptors. Indeed, the presence of neutro- ILCs can subdivided into ILC1s, ILC2s, and ILC3s, which
phils within peritubular capillaries is one of the diagnostic mirror Th1, Th2, and Th17 subsets in terms of transcrip-
features of ABMR,83 and increased numbers of intraglo- tion factor dependency and effector cytokine profile. The
merular monocytes and macrophages have been observed phenotype and dynamics of donor and recipient helper ILCs
in C4d+ ABMR.84 Macrophages may also contribute to after transplantation remains poorly understood. How-
chronic ABMR; early macrophage infiltration is predic- ever, it is likely that the nature of the transplanted organ
tive of chronic allograft nephropathy and long-term graft dictates the relative contribution of ILCs to transplant phe-
survival.85 nomena: ILCs may be expected to have significant influence
on transplanted mucosal tissues, because they are particu-
NK Cells and Innate Lymphoid Cells larly enriched at these sites. For example, the production of
In the context of organ transplantation, it is increasingly homeostatic IL-22 and amphiregulin by ILC3s and ILC2s,
clear that NK cells play a significant role.86–88 NK cells respectively, may limit detrimental tissue destruction and
are a distinct class of cytotoxic lymphocyte characterized reinforce antimicrobial defense at the mucosal epithelium
by the production of perforin, granzymes, and IFNγ that in the gut and lung.101–103 Indeed, ILC3-derived IL-22 pro-
play a role as effector cells, lysing sensitive targets accord- duction has been implicated in reduced disease progression
ing to the presence or absence of specific target antigens. and intestinal tissue damage in murine models of graft-ver-
Two subsets of NK cells exist in humans, CD56bright cells sus-host disease.104,105 Conversely, the transition to ILC1-
and CD56dim cells, with CD56dim NK cells comprising like phenotypes is associated with increased inflammation
approximately 90% of blood and spleen NK cells. This sub- and may promote early graft dysfunction.106 Curiously, the
set expresses FcγRIIIA (CD16) and undergoes antibody- absence of ILC reconstitution in severe combined immu-
dependent cell-mediated cytotoxicity (ADCC), the targeted nodeficiency (SCID) patients after hematopoietic stem cell
release of cytotoxic molecules in response to FcγR ligation (HSC) transplantation, including NK cells, was not associ-
by IgG-opsonized cells. The relevance of ADCC to organ ated with any overt susceptibility to disease.107 Therefore
rejection will be discussed in more detail when discussing more investigation into the precise nature of ILCs within
mechanisms of ABMR. NK cells also express an array of allografts is needed.
other activating and inhibitory cell surface receptors that
dictate cellular activation depending on the microenvi- CELLS AT THE INTERFACE BETWEEN INNATE AND
ronment encountered by the cell. Although the impor- ADAPTIVE IMMUNITY
tance of NK cells in bone marrow transplantation has
been long established,89,90 their role in solid organ trans- Although sufficient for initial protection against most
plantation has taken longer to be recognized. Several lab- microorganisms and sterile insults, innate immunity plays a
oratories using different experimental models found that crucial role in shaping adaptive immune responses accord-
grafts survive indefinitely in the presence of demonstrable ing to the context in which antigen is encountered. Indeed,
2 • The Immunology of Transplantation 15

complex mechanisms have evolved to ensure optimal tar- gut.125,126 Similarly, others groups have shown that lung-
geting of different effector mechanisms against viruses, resident and systemic ILC2s and ILC3s are capable of driv-
bacteria, fungi, protozoa, and multicellular parasites, while ing T cell activation in a MHC-II-dependent manner.127,128
maintaining immunologic tolerance toward innocuous self Furthermore, human splenic ILCs support B cell antibody
and foreign antigens.108 production through the production of B cell activating mol-
A critical class of innate immune cell mediating this ecules, including a proliferation-inducing ligand (APRIL)
cross-talk between innate and adaptive immunity is the and B cell activating factor (BAFF), and maintenance of B
dendritic cell (DC). DCs pick up antigen within tissues and cell-helper neutrophils.129
migrate to local draining lymph nodes for MHC-mediated
presentation to antigen-specific T cells and the initiation Stimulation of Adaptive
of adaptive immunity.109 Furthermore, DCs integrate a
variety of secondary cues, such as PRR or cytokine stimu- Alloimmunity
lation, to dictate the fate of T cell activation. For example,
it is not surprising that mucosal-resident DCs are locally The antigen-specific or adaptive immune response to a
primed for the homeostatic induction of peripheral regu- graft occurs in two main stages. In the afferent arm, donor
latory T cells (Tregs) via production of TGFβ and retinoic antigens stimulate recipient lymphocytes, which become
acid,110–112 whereas those DCs elicited in the context of activated, proliferate, and differentiate while sending sig-
infection can skew T cell activation toward inflamma- nals for growth and differentiation to a variety of other cell
tory Th1, Th2, or Th17 subsets, depending on the nature types. In the efferent arm, effector leukocytes migrate into
of the pathogen.113,114 A similar set of considerations can the organ and donor-specific alloantibodies are synthe-
be applied to monocytes and tissue-resident macrophages. sized, both of which cause tissue damage. To initiate adap-
Although less efficient at antigen presentation than DCs, tive immunity, the graft must express antigens that are
several macrophage-derived cytokines can influence T cell recognized by the recipient as foreign, and these include
polarization and activation, including IL-1β, IL-23, IL-12, ABO antigens, HLA, and non-HLA “auto-antigens” that
and TNFα.115–117 This communication with T and B cells is are polymorphic.
bidirectional. T cell-derived IFNγ and IL-4 or IL-10 are clas-
sically associated with the differentiation of monocytes and ABO BLOOD GROUP ANTIGENS
macrophages to so-called M1 and M2 phenotypes, respec- When allocating an organ to a potential recipient the first
tively.118 M1 macrophages produce high levels of reactive consideration is to ensure that it is compatible for the ABO
oxygen species and proinflammatory cytokines and che- blood group antigens. ABO antigens are expressed by most
mokines, whereas M2 macrophages produce high levels of cell types in organ allografts and, were an ABO incompat-
IL-10 and tissue-remodeling factors. However, this remains ible transplant to be performed, the presence of naturally
an oversimplification of the complex macrophage pheno- occurring anti-A and anti-B antibodies in recipients will
types in vivo. Similarly, macrophages express high levels likely cause antibody-mediated hyperacute rejection and
of FcγRs, cell surface receptors that bind to the Fc portion rapid graft loss. Organs from blood group O donors may be
of IgG antibodies, and mediated potent cellular responses safely given to recipients of any blood groups (“universal
to opsonized microbes, immune complexes, or deposited donor”) and recipients who are blood group AB may safely
IgG.119 receive organs from donors of any blood group (“universal
In recent years, there has been increasing appreciation recipient”). In practice recipients of organs from deceased
for the role of certain subsets of granulocytes in the acti- donors receive ABO blood group identical organs to avoid
vation of adaptive immunity, particularly with regard to inequity of access to organs, although recipients of kidneys
B cell activation and maintenance. Neutrophils have been from living donors often receive an ABO compatible but
described to promote antibody production by splenic mar- non-ABO identical kidney.
ginal zone B cells via their production of B cell activating
cytokines and costimulatory molecules, such as IL-21 and HLA MOLECULES
CD40L, respectively.120 Furthermore, these cells express
FcγRs, with the potential for IgG-mediated feedback. There Histocompatibility antigens differ between members of
is also evidence that neutrophils can traffic to lymph nodes the same species and are therefore targets of the immune
(LNs) for presentation of antigen to T cells121 or licensing response in allogeneic transplantation. In all vertebrate
DCs for T cell activation by TNFα-mediated maturation.51 species, histocompatibility antigens can be divided into a
Eosinophils have also been demonstrated to be a major single, albeit multigenic, MHC and numerous minor his-
determinant of B cell maintenance within the bone marrow tocompatibility (miH) systems. Incompatibility between
and mucosal tissues via similar mechanisms.122–124 Given donor and recipient for either MHC or miH leads to an
their residency and recruitment to numerous tissues, it is immune response against the graft, more vigorous for MHC
likely that these cells can influence the induction or pro- than miH. Indeed rejection of MHC-compatible organ grafts
gression of alloimmunity. is often delayed, sometimes indefinitely, although in some
ILCs are critically dependent on, and influence the activ- mouse strain and organ combinations miH differences alone
ity of, neighboring immune cells, including those of the can result in acute rejection similar to that observed across
adaptive immune system. Indeed, seminal work by Son- full MHC mismatch.130 On the other hand, the outcomes of
nenberg and colleagues has demonstrated that ILC3s are allogeneic stem cell transplantation between HLA-identical
capable of MHC class II-mediated antigen presentation siblings can be significantly affected by miH mismatches
and the suppression of antigen-specific T cells within the causing graft-versus-host disease.131
16 Kidney Transplantation: Principles and Practice

Major Histocompatibility Antigens as foreign and engaged by the TCR in the so-called “direct
MHC class I proteins are cell surface glycoproteins com- alloimmune response.” The cellular immune response to
posed of two chains—the alpha chain, which is highly poly- MHC alloantigens is consequently unique in its diversity
morphic and encoded by a class I gene, and a nonvariable and therefore the number of T cells that can be recruited to
β2-microglobulin chain (molecular weight approximately an immune response.141,142 Clinically, we currently assess
12 kD). MHC class I proteins are expressed on most nucle- and attempt to optimally match transplant donors and
ated cells, albeit at variable levels, and they are generally recipients according to the number of HLA-A, -B, and -DR
responsible for activating cytotoxic CD8 T cells. MHC class mismatches, with a minimum of 0 mismatches (0-0-0) and
II proteins are encoded entirely within the MHC and are a maximum of 6 mismatches (2-2-2) considered in the algo-
composed of two membrane-anchored glycoproteins, an rithm. In general, a greater emphasis is placed on matching
alpha and a beta chain. MHC class II molecules present pep- at DR loci because of the capacity of MHC-II mismatches to
tides and activate CD4-expressing helper T cells. The tissue activate CD4 T cells.
distribution of MHC class II proteins is far more restricted
than that of class I, being expressed constitutively only by B Minor Histocompatibility Antigens
lymphocytes, DCs, and some endothelial cells (particularly Several genes within the class I and class II regions do
in humans). During an immune or inflammatory response, not encode classical MHC proteins. In addition to those
many other cell types may be induced to express MHC class involved in antigen processing, others encode nonclassical
II proteins.132–136 MHC proteins that are similar in structure to classical MHC
Both MHC class I and MHC class II molecules have the proteins but are nonpolymorphic. These may have antigen-
capacity to present peptides but the origin of these pep- presenting capacity for specialized antigens, such as lipids
tides differs between the two. In the case of MHC-I, they (e.g., mycolic acid and lipoarabinomannan from Mycobac-
are largely acquired from the intracellular environment, terium) or peptides of different sequence but with common
whereas MHC-II largely present peptides acquired from the characteristics (e.g., with N-formylated amino termini).
extracellular environment. Nevertheless, so called “cross- Others such as HLA-G play a role in immune regulation143
presentation” between these pathways may occur, particu- particularly at the feto-maternal barrier.144
larly in the context of specialized antigen presentation by The class III region of the MHC is large and contains
DCs.137,138 genes encoding proteins with a wide range of functions
A combination of MHC and peptide forms a compound including many with roles in the immune system, includ-
epitope that is engaged by the antigen-specific T cell recep- ing TNFα and TNFβ.145 Polymorphisms that determine the
tor (TCR). The peptide-binding groove is usually occupied production of such cytokines have also been linked to cer-
by many different peptides, derived from self-proteins (often tain immune responses, including transplant rejection.146
those from the MHC) which, during infection, are replaced Although the highest degree of genetic polymorphism
by those derived from pathogens.139 The TCR repertoire is within a species lies within the MHC, many other loci
subject to negative thymic selection so that autoreactive encode proteins with a lower degree of variability. It is clear
cells are purged and positive thymic selection for TCRs that from genetic studies that these proteins can act as trans-
engage with peptides presented by autologous MHC occurs. plantation antigens; they are miH antigens. Their structure
When a pathogen invades, MHC proteins become loaded and distribution were for many years elusive. Although T
with foreign peptides that are engaged by TCR in a self- cells could recognize and respond to cells from MHC-identi-
restricted immune response. cal individuals, it was almost impossible to raise antibodies
In humans, the HLA class I molecules are HLA-A, -B, against the antigens involved, making biochemical char-
and -C; MHC class II molecules are HLA-DR, -DP, and -DQ. acterization difficult. The knowledge that T cells recognize
Their role in presenting antigenic peptide to the TCR has small peptides, together with the application of molecular
led to the evolution of a high level of genetic diversity such genetic techniques, allowed the characterization of the pro-
that there are thousands of variants of both MHC class I and totypic miH antigen, the male antigen or H-Y.147,148 From
class II genes in the human population. This is likely to have such work, it is clear that miH antigens generally represent
evolved in response to their role as restriction elements in peptides from low-polymorphic proteins presented in the
the response to pathogen-derived peptides. Certain cohorts MHC groove, in the same way as a conventional antigen
of animals within species that have limited polymorphism derived from infectious agents. The so-called H-Y antigen
at MHC loci have been devastated by infections that are is actually derived from a group of such proteins encoded
cleared without difficulty in closely related species with poly- on the Y chromosome.147–150 The first observation explains
morphic MHC.140 This genetic diversity in the MHC loci is why it has been difficult to raise antibodies to miH antigens:
an important driver of alloimmune sensitization stimulated because the combination of autologous MHC with alloge-
by pregnancy, blood transfusion, and prior transplantation. neic peptide constitutes a relatively poor conformational
The immune mechanisms involved in these responses are determinant for antibody binding, despite being an ade-
not fundamentally different from those involved with any quate determinant for TCR engagement.
other antigen. The cellular immune response to alloantigen MiH antigens can play a prominent role in rejection in
is, however, fundamentally different at least in magnitude, a recipient who is given an MHC-compatible graft but in
because MHC molecules bind a diverse range of endogenous whom preexisting sensitization to miH antigens exists.
peptides, which are therefore normally presented at the cell This situation can be shown in the rat and mouse151,152
surface. Allogeneic MHC generate a correspondingly wide and probably explains the occurrence of rejection episodes
range of compound epitopes distinct from the repertoire gen- (which rarely result in graft loss) in renal transplants per-
erated by syngeneic MHC. These are therefore recognized formed between HLA-identical siblings. Multiple miH
2 • The Immunology of Transplantation 17

differences have been shown to represent an immunogenic


stimulus equivalent to that of the MHC in a nonsensitized
recipient of a cardiac allograft in the mouse151 but it is dif-
ficult to gather similar data in clinical transplantation. Tis-
sue-specific polymorphic protein antigens have also been
described, for example, in mouse skin153 and rat kidney.154
An endothelial-monocyte antigenic system has been shown
in humans, and it has been suggested that cells sensitized to Donor leukocytes
these antigens can cause graft damage. This area has been and shed antigens
reviewed by other authors.155–157

ANTIGEN PRESENTATION Allograft-draining


lymph node
As discussed, donor antigens are presented to T cells in the
context of MHC. Activation of CD4 T cells is of particular
importance, given their ability to promote both cellular and
humoral adaptive responses, and has been demonstrated
in a number of experimental transplant systems.132,158–161
CD4 T cell activation requires the presentation of antigen Allo-MHC/
peptide
in the context of MHC-II and is carried out by professional

DIRECT
APCs, namely DCs, B cells, and some macrophages. DCs in
particular, have received great attention in this context.
T cell
receptor
Dendritic Cells Donor DC/APC Recipient T cell
DCs are present in all organs, including those that are
routinely transplanted, and in secondary lymphoid org­ Self-MHC +
shed donor
ans.162–164 Much of the work on DC function has emerged
INDIRECT 1

antigen
from murine studies, with more limited data from human
tissues, because of the challenge of obtaining fresh clini-
cal samples for in-depth analysis. However, expression of T cell
receptor
CD11c and MHC-II are useful markers to identify classical Recipient DC/APC Recipient T cell
DCs (cDC) in human tissues. cDC can be further subdivided
into the cDC1 subset that are CD141 (THBD) and XCR1+, Shed
the cDC2 subset that express CD1c+, and a further CD1c/ allo-MHC/
SEMI-DIRECT

peptide
CD141− subset. All three subsets have recently been isolated exosomes
in the human kidney using flow cytometric analysis.165
After transplantation, these cells migrate out of the trans-
planted organ, into the bloodstream, and will subsequently T cell
Recipient DC/APC Recipient T cell
encounter the recipient lymphoid system, where they are able receptor
to interact with and stimulate the host immune response.166,167
Fig. 2.5 Mechanisms of antigen presentation by dendritic cells in
Tissue-resident DCs have an immature phenotype,168 but acti- alloimmunity. Donor tissue-resident DCs and shed antigens derived
vation with PAMPs or DAMPs results in their rapid maturation from transplanted allografts can activate recipient T cells for allore-
into potently immunogenic APCs169–172 with high expression active immunity. Direct antigen presentation is mediated by migrat-
of MHC class I and class II antigens together with a range of ing donor-derived DCs within local lymphoid tissue. Indirect antigen
presentation arises from recipient DCs acquiring shed antigens from
costimulatory molecules and cytokines (see section on Direct, damaged tissue. Semidirect antigen presentation results from shed
Indirect, and Semidirect Antigen Presentation). allo-MHC/peptide exosomes being acquired by recipient DCs for pre-
DCs with an immature or partly mature phenotype sentation to recipient T cells.
deliver tolerogenic rather than activating signals and play
a crucial role in the induction of Tregs, T cell anergy, and of peptide and MHC can be engaged by recipient TCR, in a
deletion.171,173 These effects are mediated by secretion process termed direct antigen presentation (Fig. 2.5). The
of cytokines such as IL-10 and TGFβ, which promote the T cell response to allogeneic MHC occurs at a remarkably
emergence of Treg cells and through the expression of “neg- high frequency—in the order of 1 in 100. This relates in
ative costimulatory molecules” such as programmed death- part to the wide range of endogenous peptides that occupy
ligand (PD-L)1/2, inducible T cell costimulatory (ICOS) the MHC groove, generating an equivalent number of com-
ligand, Ig-like transcript (ILT)3/4, and Fas ligand. pound epitopes. It is also dependent on the capacity of the
TCR to recognize multiple distinct ligands, that is to say
Direct, Indirect, and Semidirect Antigen polyspecificity is a feature of TCR engagement.141,177
Presentation Alloantigens can also be processed and presented con-
Allogeneic MHC on DCs derived from the graft can present ventionally by recipient antigen-presenting cells. This is
a wide range of endogenous peptides derived from donor termed indirect antigen presentation (see Fig. 2.5). The indi-
tissue, both from nonpolymorphic proteins174 but also rect pathway accounts for the fact that, in animal models,
from MHC proteins themselves.175,176 These combinations elimination of passenger leukocytes from the graft does not
18 Kidney Transplantation: Principles and Practice

abolish although it may alleviate rejection. Indeed, skin wide range of genes encoding cytokines and cell surface pro-
grafts from class II−/− mice transplanted onto normal mice teins. These signaling pathways are the targets of a number
were rejected in a CD4+ T cell dependent manner.178 These of immunosuppressive medications, including calcineurin
and other experiments demonstrated the potential role of inhibitors, and are described in detail elsewhere.201–206
self MHC class II restricted presentation of exogenous allo-
antigen to stimulate T cell responses through the indirect Signal 2—Costimulation
pathway.179–181 To generate sustained T cell activation, engagement of
In clinical transplantation, and in particular in the con- surface costimulatory molecules is required in addition to
text of chronic allograft dysfunction, evidence for the role signal 1 (see Fig. 2.6). These costimulatory molecules deter-
of indirect allorecognition has been presented in various mine and mediate short-term function and long-term fate
reports using peptides derived from polymorphic regions during priming, expansion, and death of T cells. There are
of MHC antigens182,183 or cytoplasmic membrane protein many families of costimulatory molecules including those
preparations.184,185 These studies need careful interpreta- of the immunoglobulin superfamily (e.g., B7), tumor necro-
tion, however, because such assays present particular diffi- sis factor family (e.g., CD154), G protein-coupled receptors
culties for reproducibility and standardization in an outbred (e.g., C3a and C5a receptors), and lectin receptors (e.g.,
population.186,187 DC-SIGN). As increasing numbers of molecules have been
It has been proposed that the direct allogeneic response described it has become evident that these interactions
dominates acute rejection and indirect allogeneic response are highly complex, involving paracrine and cell contact
allows for ongoing class II restricted responses after the loss dependent mechanisms.
of donor DCs.188–190 This is almost certainly an oversim- In the absence of costimulatory signals, T cells may
plification,191,192 because there are significant differences become anergic, that is they are unresponsive even if they
between the expression of MHC molecules on the endothe- subsequently receive an adequate second signal.207–211
lium of mouse versus human, with long-term tonic expres- Anergic cells can also inhibit the activation of neighboring
sion of MHC-II on human endothelium even in the absence T cells.212–215
of inflammation. Therefore mouse transplant experiments The molecular basis for the costimulatory or second sig-
may not reflect the response in humans. Nevertheless, these nal for naive T cell activation was defined as that between
models have implicated indirect allorecognition in provid- CD28 and the B7 family molecule now known as CD80.216
ing cognate help for B cell alloantibody formation,193,194 These costimulatory interactions were not only the first
and are supported by the observation of an increased risk to be described but also the first to be manipulated in the
for graft loss associated with nondonor-specific and donor- setting of clinical transplantation.217,218 The cell surface
specific antibody.195,196 protein CD28 is now known to be a member of a family of
In addition to direct and indirect antigen presentation, similar proteins.219–221 Activation of downstream signal-
there is evidence that intact proteins can be exchanged ing via CD28 results from ligation with B7 family proteins,
between cells in cell culture systems, that MHC proteins CD80 or CD86. These proteins are expressed by APCs such
transfer in vivo197 and that transferred MHC stimulates as DCs and engage CD28 during antigen presentation. Sig-
allogeneic responses in vitro.197–200 The importance of this naling through CD28 in the context of TCR ligation results
semidirect pathway of antigen presentation to transplant in an increase in glucose metabolism and high levels of
outcomes remains to be clearly established. cytokine and chemokine expression, particularly IL-2, a
cytokine that promotes T cell proliferation and survival.
CD28-deficient mice have impaired immune responses
T CELL ACTIVATION
but can reject skin grafts, albeit in a delayed fashion222;
T cell activation requires not only engagement of the TCR this is likely because of other costimulatory proteins that
by a peptide:MHC complex (signal 1), but also engagement can substitute the action of CD28.219,221,223 Blocking the
of cell surface costimulatory molecules present on APC CD28 pathway in normal animals inhibits the alloimmune
(signal 2) and T cell stimulation by cytokines (signal 3) response and results in prolonged graft survival or toler-
(Fig. 2.6). ance.224–226 The most widely used reagent for this pur-
pose has been cytotoxic T lymphocyte-associated protein 4
Signal 1—TCR Activation (CTLA-4)-Ig. This blocks B7 engagement of both CD28 and
The T cell receptor comprises an alpha and a beta chain that CTLA-4, the latter of which could be counterproductive
recognize and bind to peptide:MHC complexes. The TCR has because CTLA-4 acts primarily as a coinhibitory molecule,
no catalytic activity of its own, but forms a complex with counterbalancing the effects of CD28. This is evident in the
six CD3 subunits (γδɛ2ζ2) that contain cytoplasmic immu- severe phenotype of CTLA-4−/− mice, in which animals die
noreceptor tyrosine-based activation motifs (ITAMs).201 of lymphoproliferative disorder within a few weeks of birth.
Lymphocyte-specific protein tyrosine kinase (LCK) phos- Similar in structure to CD28, CTLA-4 inhibits the earliest
phorylates these ITAMs after TCR ligation resulting in events in T cell activation. CTLA-4 has a higher affinity for
association with the ζ-chain-associated protein kinase: zeta- CD80 and CD86 than does CD28227,228 and its engagement
chain-associated protein kinase 70 (ZAP70).202 ZAP70 with CD80 induces a lattice structure at the cell surface con-
recruitment results in phosphorylation of the adapter pro- sisting of alternating CTLA-4 and CD80 homodimers. These
teins SH2 domain-containing leukocyte protein of 76 kD properties of CTLA-4 may limit the ability of CD80 to inter-
(SLP76) and linker of activated T cells (LAT). LAT facilitates act with and cluster CD28 at the immune synapse, poten-
the formation of multiprotein complexes that drive multiple tially explaining the finding that low levels of CTLA-4 can
activation pathways203 leading to de novo expression of a be effective at inhibiting immune responses. CTLA-4 may
2 • The Immunology of Transplantation 19

Allograft-draining lymph node

Signal 1 Recipient
Dendritic cell
MHC-II TCR CD4+ T cell

Signal 2
CD80/
CD28
CD86
CD40 CD40L

Signal 3 IL-2
Th-polarizing cytokines

Granzymes

Perforin
pore
FasL

Fas CD8+ T cell


activation and
Allograft expansion
destruction
Fig. 2.6 Molecular mechanisms of T cell activation. Within local draining lymph nodes, DC-T cell interaction in the presence of alloantigen results
in recipient T cell activation through a variety of molecular mechanisms. Signal 1 is provided by antigen-loaded MHC class II molecules interacting with
an antigen-specific TCR. Signal 2 is mediated by costimulatory molecules on antigen-presenting cells, such as CD80/CD86 and CD40, which engage the
corresponding receptors on T cells, in this case CD28 and CD40L, respectively. Signal 3 is mediated by APC-derived cytokines, which skew T cells into
specific subsets, such as Th1, Th2, or Th17 cells. Activated T cells also secrete IL-2, resulting in autocrine signaling and T cell expansion and survival, and
paracrine activation of local T cells, such as CD8+ effector T cells. CD8+ T cell activation can then induce tissue destruction through FasL and perforin/
granzyme-mediated mechanisms.

also deliver a negative intracellular signal229 independent of molecule 1 (DNAM-1) and cytotoxic and regulatory T-cell
its effect on CD28 that halts cell cycle progression and IL-2 molecule (CRTAM) or coinhibitory CTLA-4, programmed
production230 or affects TCR engagement.231 The actions cell death protein 1 (PD-1), B- and T-lymphocyte attenua-
of CTLA-4 may be even more complex at a polyclonal level tor (BTLA), lymphocyte-activation gene 3 (LAG-3), T cell
because CTLA-4 engagement may promote Treg function Ig and mucin-domain containing-3 (TIM-3), T cell immu-
in vivo.232,233 A modified CTLA4-Ig fusion protein, belata- noreceptor with Ig and ITIM domains (TIGIT), and leuko-
cept, has undergone robust assessment in two large clinical cyte-associated Ig-like receptor 1 (LAIR-1). A second major
trials, BENEFIT and BENEFIT-EXT, and these demonstrate family of costimulatory molecules on the T cell surface
that it is as effective as calcineurin-inhibitors (CNIs) in pre- is the TNF superfamily including CD27, CD134 (OX40),
venting acute TCMR in humans, but avoids CNI-associated and CD137 (4-1BB), which interact with a range of TNF-
nephrotoxicity resulting in better allograft function.234–236 receptor family members.223,238,239 On the T cell surface the
Belatacept-treated subjects also had a significantly lower TNF receptor family member CD154 (CD40 ligand, gp39)
rate of development of de novo donor-specific antibody itself interacts with CD40 on B cells, DCs, and monocytes.
(DSA), and nonhuman primate studies suggest this may be Larsen and coworkers showed that blocking this interaction
because of inhibition of B cell-T follicular helper (Tfh) cell could prolong graft survival in a mouse cardiac transplant
interactions.237 model.240 Even more impressive, however, were data that
Since the role of CD28 engagement was defined other combined CD28 and CD40 blockade induced permanent
members of this family of molecules have been identified; survival of allogeneic skin grafts in mice with no long-term
bound by various ligands, they generate effects that can deterioration of graft integrity.241 Tolerance to graft anti-
broadly be termed costimulatory ICOS, DNAX accessory gens could not be shown in these mice, despite the excellent
20 Kidney Transplantation: Principles and Practice

survival of the transplant itself. In a large animal setting,


kidney graft rejection in monkeys can be prevented com- T Cell-Mediated Rejection
pletely with antibodies to CD154242; however, its clinical
application is limited by thromboembolic events associated TCMR, previously known as acute cellular rejection, is the
with its expression on platelets. Alternative approaches most common type of rejection observed in the current era.
using nondepleting antibodies to CD40 are therefore now TCMR occurs in around 20% of kidney transplant recipients
being explored.243 and most frequently occurs in the first 6 months posttrans-
An understanding that memory T cells are an important plant (see Fig. 2.1). TCMR is characterized by immune cell
barrier to successful engraftment in the presence of immu- infiltration into the graft and may involve epithelial cells,
nosuppression and to tolerance induction, combined with for example a tubulitis in kidney transplants, or in more
the fact that they may be relatively resistant to CD28 inhibi- severe cases, an arteritis. This infiltrate is composed of CD8
tion, has directed interest toward identifying molecular tar- T cells, CD4 T cells, monocytes, macrophages, and B cells.
gets in T memory cells. The expression of CD2 on T effector The sequence of events that culminate in TCMR include the
memory cells,244,245 of and lymphocyte function-associated migration of immune cells into the graft, which is depen-
antigen (LFA)-1 on T memory cells,246 and even expres- dent on the production of chemokines by graft-resident cells
sion of a specific potassium channel on T effector memory and on the interaction of adhesion molecules on endothe-
cells247 have been investigated. These approaches require lial cell and immune cells. Once in the graft, cellular effector
further assessment and are not without potential compet- functions are activated causing damage to the transplant,
ing risks.248,249 and the cytotoxic functions of CD8 T cells in particular play
a key role in this process.
Signal 3—Cytokines
The consequences of TCR engagement and costimula- MIGRATION OF ACTIVATED CELLS INTO
tion are proliferation and differentiation of the T cells THE GRAFT
into effector phenotypes and the concomitant production
of cytokines required to stimulate themselves, and other To enter a site of inflammation, leukocytes must migrate
immune cell types, including CD8 T cells, macrophages, across the vascular endothelium. This migration process
DCs, and B cells. IL-2 is a potent activator and proprolif- is controlled by chemokines, and by cell-cell interactions
erative cytokine for T cells. Its effects are dependent on between the leukocyte and the endothelium.42 Activated
binding to its cell surface receptor, which has three sub- and memory cells bear adhesion proteins, chemokine recep-
units, α (CD25), β, and γ. During T cell activation the α tors, and addressins, which allow homing to and migration
subunit becomes associated with the other subunits to into peripheral tissues.260,261 In transplantation, this acti-
form a high affinity receptor. Blockade of the IL-2 recep- vation is thought to predominantly take place in second-
tor by targeting the α-chain profoundly inhibits T cell ary lymphoid organs, because aly mice lacking lymphoid
proliferation. CD25 blockade with basiliximab or dacli- organs have reduced graft rejection262,263 as do splenecto-
zumab have proven efficacy as induction agents in renal mized mice deficient in LTα or LTβ (and therefore lacking in
transplantation.250,251 lymph nodes and Peyer’s patches).264
A number of other cytokines act to polarize CD4 T cells In small bowel transplantation, recipient-derived leuko-
toward different fates, where they promote different types of cytes migrate in large numbers into the mesenteric lymph
immune responses. Th1 polarized cells mainly produce IFNγ nodes and Peyer’s patches of the graft.265–268 This likely
and drive cell-mediated inflammation and immunoglobulin results from normal homing of immune cells to the large
class switching to IgG antibodies. Th2 cells produce IL-4, volume of lymphoid tissue within the graft. During chronic
IL-5, and IL-13, and are involved in IgE class switching and rejection, lymphoid neogenesis or ectopic accumulations
eosinophil recruitment. Th17 cells produce IL-17 and IL-22 of lymphoid cells may develop within transplants in mouse
and play an important role in the clearance of extracellular models and in humans, enabling local activation of immune
pathogens and in autoimmune pathology. cells within the organ.269 The movement of lymphocytes
The role of different cytokines in allograft rejection has out of secondary lymphoid organs requires sphenogosine-
been approached using neutralizing antibodies and mice 1-phosphate receptor, a G protein‐coupled protein, and
deficient in specific cytokines. However, interpretation this has been targeted using FTY720 (fingolimod). FTY720
of these experiments is complex, because the absence of acts as a high-affinity agonist for S1P1R, inducing internal-
the cytokines can influence immune system development, ization of the receptor. This renders the cells unresponsive
there is redundancy in the action of cytokines, and cyto- to the S1P1, depriving them of an obligatory signal required
kines may have opposing actions depending on the con- for egress from lymphoid organs. Lymphocytes are therefore
text. This is illustrated by studies of IL-2−/− or IFNγ−/− mice, unable to access the peripheral circulation and allograft.
which demonstrated that neither were required for rejec- Although FTY720 was used in a number of clinical trials in
tion.252,253 IL-15 can for example substitute many of the transplantation, its use was associated with macula edema
actions of IL-2, and IL-15 transcripts are found in grafts and bradycardia, and therefore it has not entered routine
placed in IL-2−/− mice. Subsequent studies demonstrated clinical use.270–272
that whereas neither IL-2 or IFNγ were required for rejec- The processes underpinning the movement of lympho-
tion, both were required for tolerance induction.254 This cytes into the graft are similar to those described for neu-
suggested nonredundant functions for both IL-2255,256 trophils in the section on Activation of the Immune System
and IFNγ257–259 in the function of regulatory T cells (see Peritransplant. Of note, antigen-activated lymphocytes
section on Transplant Tolerance). migrate into nonlymphoid tissues260,273,274 and may show
2 • The Immunology of Transplantation 21

tissue-selective homing and preference for sites where they causing the type of tissue damage associated with rejec-
are most likely to reencounter their specific antigen.275 This tion. Third, graft rejection may be delayed by CD8 T cell
process may be facilitated further by cognate recognition by depletion.130,160,300–302
the T cell of MHC class II/peptide complexes on the vascular The importance of the individual effector mechanisms of
endothelium.276 Blocking adhesion molecule interactions CD8 T cells in mediating rejection is debated. Mice deficient
has been attempted in experimental and clinical transplan- in perforin are still able to reject skin303 and organ grafts,304
tation.78,277–279 In general, cocktails of antibodies blocking even when the grafts are resistant to Fas-mediated and
multiple adhesion molecules are more potent than single TNFα-mediated killing,305 but grafts mismatched only at
antibodies280 but the results vary. Indeed, in one study a the MHC class I were found to be rejected more slowly in
combination of antibodies blocking both ICAM-1 and LFA-1 perforin knockout mice.304
led to accelerated rejection of rat cardiac allografts.281 Anti- A number of elegant experiments in animal trans-
sense oligonucleotides have also been used to prevent the plant models demonstrate that both antigen-specific and
expression of ICAM-1 and were effective in prolonging graft nonantigen-specific effector mechanisms because of col-
survival in experimental models.282 Small molecule inhibi- lateral damage of bystander cells may be involved in graft
tors also may effectively interrupt the interactions required destruction.306–310
for leukocyte adhesion and extravasation,283 and these
reagents may simultaneously be effective in blocking IRI COMPLEMENT AND TCMR
and rejection.284
Chemokines play a crucial role in leukocyte trafficking Murine models suggest that complement may play a role in
under both normal conditions and in the setting of ster- acute TCMR. In a fully MHC-mismatched model (C57BL/6
ile inflammation and rejection. There are more than 40 to B10.BR), kidneys from C3-deficient donors survived for
different chemokines belonging to two major structural long periods without immunosuppressive treatment, in con-
families that are recognized by a range of chemokine trast to wildtype C57BL/6 donor grafts that were rejected
receptors expressed on immune cells: CC or β chemokines within 2 weeks. Recipient C3 had only a minor effect in this
(e.g., MIP-1α/β, [CCL3/4] regulated on activation, nor- model.311 Similarly, in a cardiac allograft model, deficiency
mal T cell expressed and secreted [RANTES; also known of the complement regulator CD55 (DAF) in transplanted
as CCL5], and monocyte chemoattractant protein [MCP- hearts resulted in aggressive TCMR.312 The mechanism by
1; also known as CCL2]) which attract T cells, mono- which complement augments TCMR may be a result of the
cyte/macrophages, DCs, NK cells, and some polymorphs effects of C3a and C5a on antigen-presenting cells313 or a
and CXC or α chemokines (e.g., IL-8 [CXCL8] and IFNγ- result of a direct effect on T cells via ligation of C3aR and
inducible protein) which primarily attract neutrophils and C5aR.314
T cells.285–289
A number of experimental transplant models have dem-
onstrated the importance of chemokines in mediating B Cell Activation and Antibody-
immune cell infiltration in IRI and in acute and chronic
rejection.285,286,290–293 For example, CCR1-deficient mice
Mediated Rejection
accept MHC class II mismatched grafts without immuno-
B CELL ACTIVATION
suppression and MHC class I and II mismatched grafts with
only low-dose immunosuppression.294 CXCL10−/− recipi- B cells are immune cells of the lymphoid lineage that develop
ents show normal rejection kinetics of a wildtype graft, in the bone marrow (Fig. 2.7). They are characterized by
but CXCL10−/− grafts placed into wildtype recipients show the expression of antibody as their surface antigen receptor,
prolonged survival.295 These murine studies have sparked the B cell receptor (BCR), and other markers such as CD20
interest in therapeutic targeting of chemokine networks in and CD19. Antibodies comprise two heavy and two light
human transplantation, but as yet, no agents are currently chains, and are classified according to the heavy chain they
used in clinical practice.296 express; IgM antibodies have a μ heavy chain, IgG antibod-
ies a γ heavy chain, etc. When B cells emerge from bone
MECHANISMS OF CYTOTOXICITY marrow, they express an IgM antibody and pass through
a transitional phase (expressing high levels of CD24 and
CD8 cytotoxic T cells damage and destroy target cells CD38 on their surface) before becoming follicular B cells
by the production of lytic molecules such as perforin and that reside within secondary lymphoid organs (the lymph
granzyme, and through the induction of Fas-Fas-Ligand- node and spleen).
mediated apoptosis (Fig. 2.6). In cell culture systems, MHC- When B cells encounter an antigen that binds their sur-
mismatched lymphocytes proliferate and produce cytokines face BCR they may either develop into short-lived plasma-
in response to one another in the mixed lymphocyte reac- blasts that produce early, germ-line encoded antibody315 or
tion. This results in the differentiation of CD8 T cells into alternatively, become a germinal center B cell. Here, they
effectors that lyse target cells bearing the mismatched MHC divide and mutate their variable region genes (somatic
antigens.297,298 There is considerable evidence that CD8 T hypermutation) in an attempt to produce antibody with
cells are involved in TCMR. First, they make up a significant a higher affinity for antigen.316 They also undergo class
proportion of infiltrating leukocytes in rejection allografts, switching, so that the heavy chain present in antibody
in contrast to the low number observed in grafts of ani- changes from μ to one of the other isotypes. During many
mals treated with cyclosporine to prevent rejection.92,299 rounds of division and hypermutation, B cells with a high
Second, cloned populations of CD8 T cells are capable of affinity BCR are positively selected and differentiate into
22 Kidney Transplantation: Principles and Practice

B2 CELLS
SECONDARY LYMPHOID ORGAN
Memory
B cell

Follicular CSR
B cell SHM
Transitional Germinal
B cell center

High-affinity
Immature B cell Long-lived BM
plasmablast
B cell i. ii. iii. follicle plasmablast

Marginal zone
B cell
T follicular Short-lived
helper cell plasmablast

i. MHC-II presentation ii. Co-stimulation (CD40L) iii. IL-21 and IL-4

B1 CELLS REGULATORY B CELLS


PLEURAL/PERITONEAL CAVITY HUMAN

CD24+
CD27+
CD5+ IL-10
CD38+
CD5+ Natural IgM CD1d+
Fig. 2.7 Molecular mechanisms of B cell activation. B cells can be divided into two major populations: B1 and B2. B2 cells contribute to the follicular
B cell population within lymphoid organs and to marginal zone B cells in the spleen. After stimulation of the BCR, follicular B cells receive help signals
at the interface of the T cell zone through antigen presentation to T follicular helper cells and CD40- and cytokine-mediated signals. Activated follicular
B cells undergo rounds to somatic hypermutation (SHM) and class-switch recombination (CSR), resulting in the generation of short-lived plasmablasts,
high-affinity long-lived plasma cells, and memory B cells that contribute to the high-affinity class-switched antibody pool. B1 cells are enriched in the
pleural and peritoneal cavities and are a major source of natural polyreactive IgM. Regulatory B cells also exist with the capacity to secrete antiinflam-
matory molecules, such as IL-10, that are essential for the suppression of damaging allo- and autoimmunity.

either memory B cells or plasma cells.317 A subset of CD4 lymphoid organs (lymph node and spleen), and can there-
T cells, known as Tfh cells, are engaged by germinal center fore have broad effects on immune responses far beyond
(GC) B cells presenting antigen to them. This Tfh-B cell inter- their most obvious function of antibody generation.
action is essential for the progress of the GC reaction and
for the development of memory B cells (that express CD27) Alloantibodies
and plasma cells.318,319 Only a small proportion of plasma Antibodies are the only soluble components of the adaptive
cells arising from the GC become established as long-lived immune system and have wider tissue distribution than their
plasma cells in the bone marrow. They reside within a num- cellular immune counterparts. Alloantibodies, particularly
ber of limited niches, do not proliferate, but act as long-term those that recognize donor HLA, can mediate hyperacute
antibody factories, producing IgG.320 Plasma cells have also rejection, acute ABMR, and chronic ABMR. Early efforts to
been described in inflamed tissues in autoimmunity and assess whether recipient antibodies could potentially rec-
within allografts.321–324 ognize donor cells were based on the cytotoxic crossmatch
assay, incubating recipient serum with donor cells in the
presence of complement.325 Currently, HLA genotyping of
B CELL FUNCTION
the donor and recipient and the use of single HLA antigen
B lymphocytes are best known for their ability to produce beads (SAB) allow a more precise assessment of the titer of
antibodies—the soluble (humoral) mediators of an adaptive DSA, assisting in the assessment of pretransplant immuno-
immune response. In addition, B cells can act as important logic risk.326,327 Currently, around 30% of patients on the
antigen-presenting cells for CD4 T cells, thereby initiating kidney transplant waiting list are sensitized and have vary-
cellular immunity, and may produce cytokines that can ing levels of HLA antibodies that may preclude transplan-
both activate and regulate a range of other immune cells. tation or require an antibody-reduction strategy to allow
B cells also contribute to the development of secondary transplantation to proceed.328 A meta-analysis of published
2 • The Immunology of Transplantation 23

data suggests that even low titers of pretransplant DSA may also promote intranodal lymphangiogenesis, increas-
(detectable by SAB but with a negative flow cytometric ing antigen and DC distribution in the lymph nodes.355
crossmatch), doubles the risk of ABMR and increases the They also may be involved in the generation of tertiary lym-
risk of graft failure by 76%.329 In addition, the development phoid structures that emerge in inflamed organs affected by
of de novo DSA posttransplant in nonsensitized transplant autoimmunity, for example in Sjogren disease, rheumatoid
recipients is associated with an increased frequency of acute arthritis, or type 1 diabetes mellitus,356,357 that may be the
ABMR and worse graft survival.330 The pathogenicity of source of some autoantibodies.321,358
HLA DSA also varies according to its specificity, with MHC
class II DSA having a worse effect on the allograft than class Production of Proinflammatory Cytokines
I DSA.330,331 In addition, non-HLA antibodies such as those B cells have the capacity to produce a number of cyto-
binding MICA or angiotensin II type 1 receptors may also kines after stimulation and can thereby affect a variety of
have a deleterious effect on grafts.332–334 Sigdel and col- immune cells.359 IL6 is a cytokine required for T-dependent
leagues used high-density protein arrays to analyze serum antibody responses,360 but B cells can themselves produce
samples obtained from 172 renal transplant recipients and this cytokine,361 and this may contribute to autoimmune
identified 38 de novo non-HLA antibodies that significantly pathology, for example, via Th17 cell activation leading to
associated with the development of chronic allograft injury exacerbation of experimental autoimmune encephalomy-
on protocol biopsies.335 Other non-HLA binding antibodies elitis.362 Indeed, B cells from patients with multiple sclero-
that may negatively affect the allograft include antibodies sis also demonstrate increased IL-6 production compared
binding apoptotic cells.336,337 with healthy controls.362 B cell production of TNFα and
IFNγ can also promote Th1 T cells361,363 and macrophage
Antigen Presentation to CD4 T Cells activation.364
In order for CD4 T cells to orchestrate adaptive immune Recently so-called innate response activator (IRA) B cells
responses, they must be activated by antigen presented to have been described. These splenic IgM+ B cells can pro-
them in the context of an MHC class II molecule, together duce granulocyte macrophage-colony stimulating factor
with costimulatory signals. B cells are very effective APCs (GM-CSF) in response to LPS, a TLR4 ligand, facilitating the
and have several advantages over DCs, traditionally con- mobilization of neutrophils.365 This makes an important
sidered to be the principle APC. First, they may have a contribution to pathogen containment in mouse models
high-affinity, specific receptor for antigen (the BCR), and of gram-negative sepsis and in pleural responses to gram-
can therefore efficiently and rapidly acquire large amounts positive bacteria366 but may exacerbate atherosclerosis via
of antigen for presentation.338,339 Second, they can clon- Th1 cell activation.367 These B cells are thought to origi-
ally proliferate, and therefore may rapidly become the nate from B1 B cells, a subset enriched in the peritoneal and
numerically dominant APC. Murine models have shown pleural cavities.368,369
the importance of B cells for T cell activation in vivo,340,341
including in the context of autoimmunity342 and alloim- B Cells as Regulators of the Immune Response
munity.343 In human kidney transplant recipients, tran- There is increasing evidence that B cells can not only act
scriptomic analysis of kidney biopsies with TCMR supports as effectors of the immune response but can also play an
the notion that B cells contribute to worse outcomes in immunoregulatory role, particularly via the production of
TCMR. Sarwal and colleagues identified a B cell transcrip- IL-10370 but also by contact-dependent mechanisms (e.g.,
tomic signature (CD20, CD74, immunoglobulin heavy and via PD-L1 expression371) and perhaps by direct granzyme-
light chains) in allografts with steroid-resistant TCMR and B-dependent cytotoxicity.372
a poorer outcome.344 Furthermore, the 11 genes found to IL-10-producing B cells have been shown to be important
comprise a “common rejection module,” present in samples in limiting autoimmunity in mouse models. In mice, sev-
obtained from a variety of organs during rejection, include eral groups have identified IL-10-producing B cells within
the chemokines CXCL9 and CXCL10345 that are produced a number of B cell populations including B1, transitional,
by B cells after interactions with cytotoxic T cells.346 Two and marginal zone subsets.373–375 IL-10-producing B cells
B cell genes (CD72 and BTLA) are also among those most have also been identified in humans and comprise around
differentially expressed in biopsies with TCMR.347 Together, 5% of circulating B cells, although cells with the potential
these data emphasize the potential importance of B cells as to produce IL-10 may be found at higher frequencies. As in
APCs in transplantation, and in this context, it is notable mice, human IL-10-producing B cells are present within dif-
that B cell depletion has proven to be an effective treatment ferent subsets, including transitional B cells. Several mark-
for autoimmune diseases considered to be mediated by T ers, such as CD5, CD1d, Tim-1, CD9, and CD80, have also
cells, including rheumatoid arthritis, multiple sclerosis, and been reported to localize to these “regulatory cells.”376–381
type 1 diabetes mellitus.348–350 Unlike regulatory T cells that can be identified by expres-
sion of Foxp3, a subset-specific transcription factor has
Formation and Maintenance of Secondary not been identified in regulatory B cells. Some recent data
Lymphoid Tissue suggest that plasma cells may be the main source of IL-10
B cells produce lymphotoxins (LTs) and thereby initiate the in vivo.382
formation and shape the architecture of lymph nodes and The potential importance of B cells with a regulatory
spleen.351–353 Their ongoing production of LTα1β2 is also capacity has now been identified in a number of disease con-
required for the maintenance and integrity of subcapsular texts, including systemic lupus erythematosus (SLE),376,377
sinus macrophages that form a protective screen around the allergy,379,383 transplant tolerance,384,385 and protec-
perimeter of lymph nodes.354 B cell production of VEGF-A tion from allograft rejection.386 In particular, the balance
24 Kidney Transplantation: Principles and Practice

between the production of IL-10 and proinflammatory growth factor (FGF) receptor and increased FGF ligand
cytokines such as IL-6 and TNFα may be important in regu- binding. Furthermore, DSA deposition could induce expres-
lating immune responses. For example, a lower number of sion of endothelial P-selectin for adherence of monocytes.
CD24/CD38high transitional B cells that produce high lev- Reed and colleagues have produced an elegant body of work
els of IL-10 versus TNFα is associated with worse allograft demonstrating that HLA antibodies can have direct effects
outcome in renal transplant recipients.387 In contrast, on allograft endothelial cells via variable region binding.393
in patients with drug-free long-term graft function that
are deemed “operationally tolerant” there is a significant Complement Activation
increase in the number of total B cells, particularly mem- IgG immune complexes can activate complement via the
ory B cells and B cells expressing CD1d and CD5. In vitro, B classical pathway and this process is evident in ABMR by
cells purified from these subjects had a relative increase in the detection of C4d on peritubular capillaries.83 Further
FcγRIIB expression and an increase in B cell scaffold protein evidence that complement fixation may contribute to the
with ankyrin repeats 1 (BANK1), a negative regulator of pathogenicity of alloantibodies is provided by the studies
CD40 signaling.388 Other studies show a higher proportion investigating C1q or C3d binding. Loupy et al. investigated
of CD24/CD38high transitional B cells associated with long- 1016 antibody-compatible renal transplant recipients and
term transplant tolerance and B cells from tolerant subjects demonstrated that patients that developed C1q-binding
produce more IL-10 after in vitro stimulation.384,385 DSA after transplantation had the lowest 5-year rate of
graft survival (54%), compared with those with noncomple-
ment-DSA (93%).395 A more recent study suggests that the
ANTIBODY-EFFECTOR FUNCTION IN ABMR
detection of HLA antibodies that bind to C3d may have an
Acute ABMR is uncommon in nonsensitized transplant even greater prognostic significance in patients with acute
recipients and is difficult to treat. The diagnosis of ABMR ABMR.396 A number of complement inhibitors, including
has been facilitated by more sensitive methods of alloan- eculizumab and C1 esterase inhibitors, are currently being
tibody detection and by the identification of complement trialed for both the prevention of ABMR in antibody-incom-
activation in allograft biopsies via C4d staining. Clinical patible transplant recipients,397 and for the treatment of
features of ABMR include a decline in allograft function, ABMR.398,399
the presence of donor-specific HLA antibodies, C4d deposi-
tion in peritubular capillaries, and evidence of acute vascu- FcγR Activation
lar injury (e.g., capillaritis, with neutrophils in capillaries). The absence of C4d staining in more than half of biopsies
ABMR, particularly in its chronic form, may occur in the with late ABMR highlights the importance of complement-
absence of C4d deposition on biopsy.83,389,390 Analysis independent mechanisms in mediating the deleterious
of transcripts may also assist in making a more accurate effects of DSAs.83,391 Furthermore, some IgG isotypes (IgG4)
assessment of the type of rejection, particularly in diagnos- cannot fix complement, whereas IgG2 has a limited comple-
ing antibody-mediated rejection, compared with standard ment-activating capacity compared with IgG1 and IgG3.400
histopathological analysis, even in the absence of C4d FcγRs bind to the Fc portion of IgG and mediate activation of
staining.391,392 the immune cells that express them (Fig. 2.8). In humans,
A gradual decline in allograft function with time is almost there are several activating receptors (FcγRIIA, FcγRIIC,
universally observed. This was previously termed chronic FcγRIIIA, and FcγRIIIB) and a single inhibitory receptor,
allograft nephropathy but the more recent Banff Classifica- FcγRIIB, which plays a critical role in suppressing IgG-
tions have sought to distinguish nonimmunologic insults, mediated inflammation.401,402 FcγRs are widely expressed
for example, CNI toxicity, from chronic rejection, particu- on immune cells, including neutrophils, monocytes, mac-
larly chronic ABMR. Chronic ABMR is characterized by rophages, DCs, mast cells, NK cells, and B cells but the effect
vasculopathy and in the kidney is evidenced by glomerulop- of FcγR ligation varies between cells. For example, neu-
athy (double contouring in peripheral capillary loops) and trophils express FcγRIIA and FcγRIIIB, and cross-linking
peritubular capillary basement. There may also be peritu- leading to phagocytosis, cytokine and superoxide produc-
bular capillary C4d staining, although this is not universal. tion, neutrophil adhesion, and extracellular trap formation
Clinically, this usually occurs in patients with detectable (NETosis),403–410 whereas on macrophages and dendritic
HLA DSA, often in the context of noncompliance. cells, FcγR ligation induces proinflammatory cytokine pro-
The histologic features of ABMR result from well-estab- duction, including TNFα and IL-1β, phagocytosis, CCR7-
lished pathways of alloantibody effector function, namely dependent migration, and antigen presentation.411–414
direct activation of endothelial cells via binding to MHC393 There are several lines of evidence implicating FcγR
complement activation (via the classical pathway) or activity in rejection severity and graft survival in ABMR.
recruitment and activation of immune cells that express NK cells, in particular, have received significant atten-
receptors for the Fc portion of IgG or complement receptors tion as an immune cell subset mediating FcγR-dependent
(including neutrophils, macrophages, monocytes, DCs, and inflammation in AMBR. Hirohashi et al. demonstrated that
NK cells394; Fig. 2.8). chronic allograft vasculopathy in murine heat allografts
was NK cell and FcγR dependent, inflammation being
Direct Stimulation of Endothelial MHC attenuated after anti-NK1.1 IgG-treatment or in the pres-
Independently of FcγR engagement, IgG can directly acti- ence of F(ab′)2 fragments of IgG2a DSA.415 Consistent with
vate allograft endothelium. Binding of DSA to HLA induced a role in human ABMR, FCGR3A and other NK cell-asso-
intracellular signaling and endothelial cell survival and ciated transcripts correlate with the presence of DSA and
proliferation, mediated by upregulation of the fibroblast ABMR.416,417 Furthermore, several groups have examined
2 • The Immunology of Transplantation 25

NK cell/neutrophil/
monocyte

iii. Complement activation


i. FcγR ligation on
immune cells

ii. Direct activation


of endothelium

Allograft endothelium

Key

C1q C4d FcγR DSA Donor HLA

A>I A=I A<I


Expression
level

IFN-γ IL-4
TNFα IL-10
FcγRIIIA-V158

FcγRIIIA-F158
FcγRIIA-H131

FcγRIIA-R131
FcγRIIB-T232

FcγRIIB-I232
variation
Genetic
glycosylation
IgG

Fuc SA

B
Fig. 2.8 Mechanisms of antibody-mediated rejection. (A) Donor-specific antibody (DSA) binding to allograft endothelium can activate a variety of
inflammatory mechanisms: (i) engagement of FcγRs on local and circulating immune cells, such as NK cells; (ii) direct activation of allograft endothe-
lium through cross-linking of endothelial cell surface molecules; (iii) activation complement via the classical pathway. (B) Numerous factors influence
the ability of IgG-FcγR engagement to direct inflammation and tissue damage. FcγR cellular expression levels are controlled by the cytokine milieu:
inflammatory mediators, such as IFNγ and TNFα increase expression levels of activating FcγR, increasing the activating-to-inhibitory (A:I) ratio. FcγR
polymorphisms can influence FcγR expression level or signaling capacity. Finally, the IgG glycosylation state alters the capacity of antibodies to interact
with different FcγRs: sialylated IgG exhibits reduced binding to activating type I FcγRs, whereas defucosylated IgG increases FcγRIIIA binding affinity.
Fuc, ucose; SA, sialic acid. Green receptor, activating FcγR (A). Red receptor, inhibitory FcγR (I).
26 Kidney Transplantation: Principles and Practice

activating FcγR single nucleotide polymorphisms (SNPs) allograft. Animal models have suggested that costimulatory
in kidney transplant recipients. A recent study of a cohort blockade may provide, in theory, a mechanism of achieving
of 85 DSA-positive kidney allograft recipients by Arnold this (as discussed in the section on T Cell-Mediated Rejec-
et al. demonstrated that individuals with the high-affinity tion), although this has not directly translated to clinical
FcγRIIIA-V158 variant showed higher rate of peritubular practice in humans.
capillaritis, an effect independent of C1q-binding or capil- A number of immune cell subsets have immunoregula-
lary C4d.418 Functionally, a model NK cell line expressing tory capacity, and may therefore contribute to tolerance
FcγRIIIA-V158 produced over two fold more IFNγ upon induction in transplantation, including CD4 T cells, B cells,
incubation with HLA antibody-coated cells compared with DCs, and macrophages. These cells broadly act via the pro-
those expressing low-affinity FcγRIIIA-F158, consistent duction of antiinflammatory cytokines such as IL-10 and
with the potential role of NK cells in driving ABMR. TGFβ, and contact-dependent inhibition. The best-charac-
FcγR associations have also been identified independently terized regulatory cell subset are CD4+CD25+ Tregs cells
of NK cells and FcγRIIIA. Whereas allograft survival was that modulate immune responses via IL-10, TGFβ, and
increased in patients with the FcγRIIA-R/R131 genotype IL-35,433 adenosine production,434 down-regulation of DC
in one study,419 in subsequent studies the same genotype costimulation, and up-regulation of indoleamine-pyrrole
was associated with acute rejection.420 The FcγRIIA-R131 2,3-dioxygenase (IDO).215 Tregs also may have more direct
variant exhibits reduced binding affinity for IgG1 and IgG2 actions on effector cell viability through mechanisms that
subclasses and has been postulated to contribute to pathol- involve granzyme B.435 The phenotype of CD4+CD25+
ogy through inefficient clearance of deposited IgG within Tregs cells is highly dependent on the expression of the
allografts.421,422 This is consistent with the high levels transcription factor Foxp3, which is required to mediate
FcγRIIA expression on professional phagocytes. Indeed, regulatory activity including suppression of IL-2 and IFNγ
monocytes expressing the high-affinity FcγRIIA-H/H131 production, and expression of CTLA-4 and glucocorticoid-
variant were found to adhere more strongly to HLA anti- induced TNFR-related protein (GITR). Foxp3 is therefore
body-activated endothelium.423 In a larger study of 200 an important marker of Treg activity, although the pheno-
kidney transplant recipients who had lost their grafts, the type of peripherally generated Tregs may not be stable in
FcγRIIA-R/R131 genotype was associated with early graft all circumstances.436 CTLA-4 appears to play an important
loss, particularly in those patients who were DSA positive.424 role in Treg function,233,437 engaging CD80/86 on DCs and
In mouse models, FcγRIIB-deficient mice develop alloan- inducing IDO expression.215,438 There is ample evidence
tibody-driven chronic vasculopathy analogous to human for the importance of Tregs in many models of experimen-
chronic rejection in a cardiac allograft model (BM12 organs tal transplantation tolerance,439–441 well-illustrated in
into C57BL/6 mice).425 This study is consistent with the models using donor-specific transfusion, and nondepleting
known role of FcγRIIB in suppressing humoral immunity, anti-CD4 antibody in which Treg function seems to play
although the exact mechanism of IgG-mediated pathology was a crucial mechanistic role.442 Perhaps more importantly,
not dissected. Indeed, in a model of antibody-mediated nephri- recent evidence in humanized animal models suggests that
tis, myeloid-specific FcγRIIB deficiency is sufficient to exacer- the infusion of nonspecifically expanded Tregs can abro-
bate tissue inflammation.426 A number of nonsynonymous gate the acquisition of transplant arteriosclerosis, opening
SNPs have been identified in the FCGR2B gene in humans, potential translation into the clinical setting.443–446 In this
with one occurring at a notable frequency (rs1050501). context, a clinical trial in kidney transplant is underway,
This SNP encodes an isoleucine-to-threonine substitution at the ONE Study, which expands regulatory cells ex vivo
position 232, located within the transmembrane domain.402 (including Tregs) and aims to initially assess safety (https://
This substitution results in receptor loss of function as a result clinicaltrials.gov/ct2/show/NCT02129881).
on impaired lateral mobility and recruitment to signaling Perhaps one of the most surprising findings on the nature
domains.427–429 Surprisingly, despite being a major risk factor of allograft tolerance in humans has been gained by study-
in SLE and the heightened inflammatory responses of mono- ing the handful of human subjects who fail to reject their
cytes from FcγRIIB-T232 individuals to IgG, no association allografts in spite of little or no immunosuppression. These
was observed between FcγRIIB-T232 and graft or patient observations have arisen in a tiny fraction of noncompli-
survival in a large study of more than 2800 renal transplant ant patients or in cases where immunosuppression has
recipients.430 However, patients were not stratified, for exam- been stopped in the context of malignancy. Analysis of the
ple based on DSA or ABMR status, possibly masking effects. peripheral blood mononuclear cell (PBMC) transcriptome
Curiously, human cultured aortic endothelial cells also in these tolerant transplant recipients identified an excess
express FcγRI and FcγRII and mediated IgG internalization, of B cell gene transcripts and an increase in CD20 mRNA
cytokine production, and the upregulation of adhesion in the urine of tolerant subjects.388,447 In addition, a sig-
molecules directly.431,432 This expression can be further nificant increase in the number of total B cells, particularly
enhanced with IFNγ and TNFα in vitro. However, whether memory B cells and B cells expressing CD1d and CD5, has
this occurs in vivo and whether it contributes to IgG- been observed in these subjects,388 whereas other groups
mediated phenomena in allografts remains to be addressed. have shown a higher proportion of IL-10-producing CD24/
CD38high transitional B cells.384,385 In contrast, a lower
number of IL-10-producing CD24/CD38high transitional B
Transplant Tolerance cells has been associated with worse allograft outcome in
renal transplant recipients.387 Together, these data sug-
Given the adverse effect of long-term immunosuppres- gest that manipulation of the regulatory fraction of the B
sion, one of the key goals in transplantation is the genera- cell compartment may also hold utility in efforts to induce
tion of immunologic nonresponsiveness (tolerance) to the transplant tolerance.
2 • The Immunology of Transplantation 27

Factors Influencing Rejection knowledge of the detail and complexity of both innate and
adaptive immune systems has been transformed by basic
Beyond the Graft—The research. After the initial intense focus on T cell response
Microbiome that has been accompanied by increasing success in tar-
geting this pathway to prevent TCMR, attention has now
The human body is a complex ecosystem of host cells and turned to innate and humoral immunity. Innate immune
commensal microorganisms and it is increasingly appre- responses mediate sterile inflammation in IRI and com-
ciated that the composition of the microbiome has signifi- promise immediate and long-term graft function. Organ
cant consequences for host homeostasis and inflammation. shortage has necessitated the use of allografts from subop-
Indeed, it is clear that the microbiome has a significant timal donors that are more susceptible to the effects of IRI.
influence on the activity of host immune cells, not only Therefore developing treatments that can ameliorate IRI is
within mucosal barrier sites, such as the gastrointestinal of significant clinical interest. With prolonged patient and
tract, but also systemically.448–450 allograft survival and an increasing number of patients
With respect to transplant rejection, environment- being listed for retransplantation, the issue of allosensitiza-
interfacing organs, such as the gut, are the most likely to tion is also a major problem. The presence of DSA is still a
be influenced by the composition of the microbiome. For significant barrier to transplantation, and there is need for
example, monocolonization of germ-free mice with seg- an increased understanding of the mechanisms of B cell and
mented filamentous bacteria is sufficient to induce local antibody effector function to identify immunosuppressants
Th17 cells that contribute to intestinal inflammation.451 that can deal with this arm of the immune system. These
Conversely, tolerogenic DCs and microbiota-derived studies will need to move away from the historical reliance
short-chain fatty acids have direct roles in the priming of on mouse models, in part because of significant differences
local Treg responses to suppress damaging inflammatory in HLA expression between mouse and human endothe-
responses.452 Therefore it is possible that a transplanted lium, and because our increasing genomic and informatic
organ’s microbial flora may dictate subsequent alloimmu- capabilities will enable experimental medicine studies using
nity and rejection, opening the way for targeted manipula- human subjects, with all their complexity and diversity.
tion of the microbiome in these individuals.453
In addition to the importance of the donor organ micro- References
biome, organ failure and transplantation have secondary
1. Naesens M, Sarwal MM. Molecular diagnostics in transplantation.
effects on the recipient microbiome, characterized by a loss Nat Rev Nephrol 2010;6(10):614–28.
of diversity and species richness. Ongoing immunosup- 2. Obhrai J, Goldstein DR. The role of toll-like receptors in solid organ
pression, prophylactic antibiotic administration, dietary transplantation. Transplantation 2006;81(4):497–502.
restrictions, and IRI have effects on commensal dysbio- 3. Nakhaei P, et al. RIG-I-like receptors: sensing and responding to
sis and the emergence of pathobionts, increasing the risk RNA virus infection. Semin Immunol 2009;21(4):215–22.
4. Philpott DJ, Girardin SE. Nod-like receptors: sentinels at host mem-
of enteric infections and inflammation.454 For example, branes. Curr Opin Immunol 2010;22(4):428–34.
advanced renal failure is associated with urea and uric 5. Ricklin D, et al. Complement: a key system for immune surveillance
acid influx into the gastrointestinal tract and alterations and homeostasis. Nat Immunol 2010;11(9):785–97.
in the intestinal microbiota and increases in microbial 6. Matzinger P. Tolerance, danger, and the extended family. Annu Rev
Immunol 1994;12:991–1045.
families, such as Proteobacteria. A similar dysbiosis is 7. Kono H, Rock KL. How dying cells alert the immune system to dan-
observed in individuals with acute and chronic liver dis- ger. Nat Rev Immunol 2008;8(4):279–89.
ease and in patients with small bowel transplant rejection. 8. Mariathasan S, et al. Cryopyrin activates the inflammasome in
In renal allografts, the magnitude of IR-induced acute kid- response to toxins and ATP. Nature 2006;440(7081):228–32.
ney injury may be influenced by the gut microbiota, via 9. Yamasaki K, et al. NLRP3/cryopyrin is necessary for interleukin-
1beta (IL-1beta) release in response to hyaluronan, an endog-
the production of short-chain fatty acids.455 In a mouse enous trigger of inflammation in response to injury. J Biol Chem
model of minor antigen-mismatched skin grafts, pretreat- 2009;284(19):12762–71.
ment of donor and recipient mice with broad-spectrum 10. Murphy SP, Porrett PM, Turka LA. Innate immunity in transplant
antibiotics or the use of germ-free mice significantly pro- tolerance and rejection. Immunol Rev 2011;241(1):39–48.
11. Land WG, et al. Transplantation and damage-associated molecular
longed the survival of allografts, attributed to a reduction patterns (DAMPs). Am J Transplant 2016;16(12):3338–61.
in the ability of antigen-presenting cells to induce allore- 12. Jin C, Flavell RA. Molecular mechanism of NLRP3 inflammasome
active T cells.456 In renal transplant recipients, changes in activation. J Clin Immunol 2010;30(5):628–31.
the urine microbiome have been associated with adverse 13. Zhou R, et al. Thioredoxin-interacting protein links oxidative stress
features on biopsy, including interstitial fibrosis and tubu- to inflammasome activation. Nat Immunol 2010;11(2):136–40.
14. Steri M, et al. Overexpression of the cytokine BAFF and autoimmu-
lar atrophy.457 Therefore further research is required to nity risk. N Engl J Med 2017;376(17):1615–26.
delineate the contribution of the microbiota to allograft 15. Walport MJ. Complement. First of two parts. N Engl J Med
rejection. 2001;344(14):1058–66.
16. Walport MJ. Complement. Second of two parts. N Engl J Med
2001;344(15):1140–4.
17. Zuber J, et al. New insights into postrenal transplant hemolytic ure-
Conclusion mic syndrome. Nat Rev Nephrol 2011;7(1):23–35.
18. Ricklin D, Reis ES, Lambris JD. Complement in disease: a defence sys-
The immunologic basis of transplant rejection was pro- tem turning offensive. Nat Rev Nephrol 2016;12(7):383–401.
posed by Gorer458 and defined by Medawar, who demon- 19. Pickering MC, et al. C3 glomerulopathy: consensus report. Kidney
Int 2013;84(6):1079–89.
strated an immune response that is specific for donor tissue, 20. Servais A, et al. Acquired and genetic complement abnormalities
is consequent upon infiltration by leukocytes, and displays play a critical role in dense deposit disease and other C3 glomeru-
memory.459–462 In the 70 years since these discoveries, our lopathies. Kidney Int 2012;82(4):454–64.
28 Kidney Transplantation: Principles and Practice

21. Zand L, et al. Clinical findings, pathology, and outcomes of C3GN 48. Karmakar M, et al. Neutrophil P2X7 receptors mediate NLRP3
after kidney transplantation. J Am Soc Nephrol 2014;25(5): inflammasome-dependent IL-1beta secretion in response to ATP.
1110–7. Nat Commun 2016;7:10555.
22. McCaughan JA, O’Rourke DM, Courtney AE. Recurrent dense 49. Jones ND, et al. Regulatory T cells can prevent memory CD8+ T-cell-
deposit disease after renal transplantation: an emerging role for mediated rejection following polymorphonuclear cell depletion. Eur
complementary therapies. Am J Transplant 2012;12(4):1046–51. J Immunol 2010;40(11):3107–16.
23. Thurman JM, et al. Lack of a functional alternative complement 50. Kish DD, et al. Neutrophil expression of Fas ligand and perforin
pathway ameliorates ischemic acute renal failure in mice. J Immu- directs effector CD8 T cell infiltration into antigen-challenged skin.
nol 2003;170(3):1517–23. J Immunol 2012;189(5):2191–202.
24. Thurman JM, et al. Acute tubular necrosis is characterized by 51. Kreisel D, et al. Emergency granulopoiesis promotes neutrophil-
activation of the alternative pathway of complement. Kidney Int dendritic cell encounters that prevent mouse lung allograft
2005;67(2):524–30. acceptance. Blood 2011;118(23):6172–82.
25. Thurman JM, et al. Altered renal tubular expression of the comple- 52. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in
ment inhibitor Crry permits complement activation after ischemia/ health and inflammation. Nat Rev Immunol 2013;13(3):159–75.
reperfusion. J Clin Invest 2006;116(2):357–68. 53. Sayah DM, et al. Neutrophil extracellular traps are pathogenic in pri-
26. de Vries B, et al. The mannose-binding lectin-pathway is involved in mary graft dysfunction after lung transplantation. Am J Respir Crit
complement activation in the course of renal ischemia-reperfusion Care Med 2015;191(4):455–63.
injury. Am J Pathol 2004;165(5):1677–88. 54. Fournier BM, Parkos CA. The role of neutrophils during intestinal
27. Moller-Kristensen M, et al. Mannan-binding lectin recognizes struc- inflammation. Mucosal Immunol 2012;5(4):354–66.
tures on ischemic reperfused mouse kidneys and is implicated in tis- 55. Varol C, Mildner A, Jung S. Macrophages: development and tissue
sue injury. Scand J Immunol 2005;61(5):426–34. specialization. Annu Rev Immunol 2015;33:643–75.
28. Farrar CA, et al. Local extravascular pool of C3 is a determinant of 56. Epelman S, Lavine KJ, Randolph GJ. Origin and functions of tissue
postischemic acute renal failure. FASEB J 2006;20(2):217–26. macrophages. Immunity 2014;41(1):21–35.
29. Parker MD, et al. Ischemia-reperfusion injury and its influence on 57. Dannappel M, et al. RIPK1 maintains epithelial homeostasis by
the epigenetic modification of the donor kidney genome. Transplan- inhibiting apoptosis and necroptosis. Nature 2014;513(7516):
tation 2008;86(12):1818–23. 90–4.
30. Damman J, et al. Local renal complement C3 induction by donor 58. Takahashi N, et al. RIPK1 ensures intestinal homeostasis by pro-
brain death is associated with reduced renal allograft function after tecting the epithelium against apoptosis. Nature 2014;513(7516):
transplantation. Nephrol Dial Transplant 2011;26(7):2345–54. 95–9.
31. Zheng X, et al. Protection of renal ischemia injury using combina- 59. Kalliolias GD, Ivashkiv LB. TNF biology, pathogenic mecha-
tion gene silencing of complement 3 and caspase 3 genes. Transplan- nisms and emerging therapeutic strategies. Nat Rev Rheumatol
tation 2006;82(12):1781–6. 2016;12(1):49–62.
32. Zhou W, et al. Predominant role for C5b-9 in renal ischemia/reperfu- 60. Nauseef WM, Borregaard N. Neutrophils at work. Nat Immunol
sion injury. J Clin Invest 2000;105(10):1363–71. 2014;15(7):602–11.
33. de Vries B, et al. Complement factor C5a mediates renal ischemia- 61. Glass CK, Natoli G. Molecular control of activation and priming in
reperfusion injury independent from neutrophils. J Immunol macrophages. Nat Immunol 2016;17(1):26–33.
2003;170(7):3883–9. 62. Chen CJ, et al. Identification of a key pathway required for the
34. de Vries B, et al. Inhibition of complement factor C5 protects against sterile inflammatory response triggered by dying cells. Nat Med
renal ischemia-reperfusion injury: inhibition of late apoptosis and 2007;13(7):851–6.
inflammation. Transplantation 2003;75(3):375–82. 63. Schroder K, Tschopp J. The inflammasomes. Cell 2010;140(6):821–
35. Arumugam TV, et al. A small molecule C5a receptor antagonist pro- 32.
tects kidneys from ischemia/reperfusion injury in rats. Kidney Int 64. Tsung A, et al. The nuclear factor HMGB1 mediates hepatic
2003;63(1):134–42. injury after murine liver ischemia-reperfusion. J Exp Med 2005;
36. Zheng X, et al. Gene silencing of complement C5a receptor using 201(7):1135–43.
siRNA for preventing ischemia/reperfusion injury. Am J Pathol 65. Armstrong DA, et al. Neutrophil chemoattractant genes KC and
2008;173(4):973–80. MIP-2 are expressed in different cell populations at sites of surgical
37. Ichim TE, et al. RNA interference: a potent tool for gene-specific ther- injury. J Leukoc Biol 2004;75(4):641–8.
apeutics. Am J Transplant 2004;4(8):1227–36. 66. Chen Y, et al. ATP release guides neutrophil chemotaxis via P2Y2
38. Patel H, et al. Therapeutic strategy with a membrane-localizing com- and A3 receptors. Science 2006;314(5806):1792–5.
plement regulator to increase the number of usable donor organs after 67. Springer TA. Adhesion receptors of the immune system. Nature
prolonged cold storage. J Am Soc Nephrol 2006;17(4):1102–11. 1990;346(6283):425–34.
39. Sacks S, et al. Targeting complement at the time of transplantation. 68. Wu H, et al. HMGB1 contributes to kidney ischemia reperfusion
Adv Exp Med Biol 2013;735:247–55. injury. J Am Soc Nephrol 2010;21(11):1878–90.
40. Kaabak M, et al. A prospective randomized, controlled trial of ecu- 69. Leemans JC, et al. Renal-associated TLR2 mediates ischemia/reper-
lizumab to prevent ischemia-reperfusion injury in pediatric kidney fusion injury in the kidney. J Clin Invest 2005;115(10):2894–903.
transplantation. Pediatr Transplant 2018;22(2). 70. Wu H, et al. TLR4 activation mediates kidney ischemia/reperfusion
41. Kirby JA. Function of leukcocyte adhesion molecules during injury. J Clin Invest 2007;117(10):2847–59.
allograft rejection. In: Tilney NL, Strom TB, Paul LC, editors. Trans- 71. Iyer SS, et al. Necrotic cells trigger a sterile inflammatory response
plantation Biology: Cellular and Molecular Aspects. Philadelphia, through the Nlrp3 inflammasome. Proc Natl Acad Sci USA
PA: Lippincott-Raven Publishers; 1996. 2009;106(48):20388–93.
42. Stepkowski SM. Therapeutic potential for adhesion antagonists in 72. Melnikov VY, et al. Neutrophil-independent mechanisms of
organ transplantation. Curr Opin Organ Transplant 2002;7:366–72. caspase-1- and IL-18-mediated ischemic acute tubular necrosis in
43. McEver RP, et al. GMP-140, a platelet alpha-granule membrane pro- mice. J Clin Invest 2002;110(8):1083–91.
tein, is also synthesized by vascular endothelial cells and is localized 73. Faubel S, et al. Caspase-1-deficient mice are protected against
in Weibel-Palade bodies. J Clin Invest 1989;84(1):92–9. cisplatin-induced apoptosis and acute tubular necrosis. Kidney Int
44. Pober JS, et al. Overlapping patterns of activation of human endo- 2004;66(6):2202–13.
thelial cells by interleukin 1, tumour necrosis factor and immune 74. Liu HF, et al. Effects of specific interleukin-1beta-converting enzyme
interferon. J Immunol 1986;137:1893–6. inhibitor on ischemic acute renal failure in murine models. Acta
45. Cose S, et al. Evidence that a significant number of naive T cells enter Pharmacol Sin 2005;26(11):1345–51.
non-lymphoid organs as part of a normal migratory pathway. Eur J 75. Kelly KJ, et al. Antibody to intercellular adhesion molecule 1 pro-
Immunol 2006;36(6):1423–33. tects the kidney against ischemic injury. Proc Natl Acad Sci USA
46. Laskowski I, et al. Molecular and cellular events associated with 1994;91(2):812–6.
ischemia/reperfusion injury. Ann Transplant 2000;5(4):29–35. 76. Kelly KJ, et al. Intercellular adhesion molecule-1-deficient mice
47. McDonald B, et al. Intravascular danger signals guide neutrophils to are protected against ischemic renal injury. J Clin Invest 1996;
sites of sterile inflammation. Science 2010;330(6002):362–6. 97(4):1056–63.
2 • The Immunology of Transplantation 29

77. Rabb H, et al. Role of CD11a and CD11b in ischemic acute renal fail- 102. Sonnenberg GF, et al. CD4(+) lymphoid tissue-inducer cells promote
ure in rats. Am J Physiol 1994;267(6 Pt 2):F1052–8. innate immunity in the gut. Immunity 2011;34(1):122–34.
78. Haug CE, et al. A phase I trial of immunosuppression with anti- 103. Zaiss DM, et al. Emerging functions of amphiregulin in orches-
ICAM-1 (CD54) mAb in renal allograft recipients. Transplantation trating immunity, inflammation, and tissue repair. Immunity
1993;55(4):766–72. Discussion 772-3. 2015;42(2):216–26.
79. Salmela K, et al. A randomized multicenter trial of the anti-ICAM-1 104. Hanash AM, et al. Interleukin-22 protects intestinal stem cells from
monoclonal antibody (enlimomab) for the prevention of acute rejec- immune-mediated tissue damage and regulates sensitivity to graft
tion and delayed onset of graft function in cadaveric renal trans- versus host disease. Immunity 2012;37(2):339–50.
plantation: a report of the European Anti-ICAM-1 Renal Transplant 105. Lindemans CA, et al. Interleukin-22 promotes intestinal-stem-cell-
Study Group. Transplantation 1999;67(5):729–36. mediated epithelial regeneration. Nature 2015;528(7583):560–4.
80. Takada M, et al. Early cellular and molecular changes in ischemia/ 106. Bernink JH, et al. Human type 1 innate lymphoid cells accumulate in
reperfusion injury: inhibition by a selectin antagonist, P-selectin gly- inflamed mucosal tissues. Nat Immunol 2013;14(3):221–9.
coprotein ligand-1. Transplant Proc 1997;29(1-2):1324–5. 107. Vely F, et al. Evidence of innate lymphoid cell redundancy in
81. Singbartl K, Green SA, Ley K. Blocking P-selectin protects from humans. Nat Immunol 2016;17(11):1291–9.
ischemia/reperfusion-induced acute renal failure. FASEB J 108. Sallusto F, Lanzavecchia A. Heterogeneity of CD4+ memory T
2000;14(1):48–54. cells: functional modules for tailored immunity. Eur J Immunol
82. Osama Gaber A, et al. YSPSL (rPSGL-Ig) for improvement of early 2009;39(8):2076–82.
renal allograft function: a double-blind, placebo-controlled, multi- 109. Lanzavecchia A, Sallusto F. Regulation of T cell immunity by den-
center Phase IIa study(1,2,3). Clin Transplant 2011;25(4):523–33. dritic cells. Cell 2001;106:263–6.
83. Haas M, et al. Banff. 2013 meeting report: inclusion of c4d-negative 110. Worbs T. Oral tolerance originates in the intestinal immune sys-
antibody-mediated rejection and antibody-associated arterial lesions. tem and relies on antigen carriage by dendritic cells. J Exp Med
Am J Transplant 2014;14(2):272–83. 2006;203(3):519–27.
84. Sund S, et al. Glomerular monocyte/macrophage influx correlates 111. Esterházy D, et al. Classical dendritic cells are required for dietary
strongly with complement activation in 1-week protocol kidney antigen–mediated induction of peripheral Treg cells and tolerance.
allograft biopsies. Clin Nephrol 2004;62(2):121–30. Nat Immunol 2016;17(5):545–55.
85. Pilmore HL, et al. Early up-regulation of macrophages and myofibro- 112. Coombes JL, et al. A functionally specialized population of muco-
blasts: a new marker for development of chronic renal allograft rejec- sal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta
tion. Transplantation 2000;69(12):2658–62. and retinoic acid-dependent mechanism. J Exp Med 2007;204(8):
86. Hanvesakul R, et al. KIR and HLA-C interactions promote dif- 1757–64.
ferential dendritic cell maturation and is a major determinant 113. Denning TL, et al. Functional specializations of intestinal dendritic
of graft failure following kidney transplantation. PLoS One cell and macrophage subsets that control Th17 and regulatory T cell
2011;6(8):e23631. responses are dependent on the T cell/APC ratio, source of mouse
87. Hanvesakul R, et al. Donor HLA-C genotype has a profound impact strain, and regional localization. J Immunol (Baltimore, MD:1950)
on the clinical outcome following liver transplantation. Am J Trans- 2011;187(2):733–47.
plant 2008;8(9):1931–41. 114. den Dunnen J, et al. IgG opsonization of bacteria promotes Th17
88. Uehara S, et al. NK cells can trigger allograft vasculopathy: the responses via synergy between TLRs and FcγRIIa in human den-
role of hybrid resistance in solid organ allografts. J Immunol dritic cells. Blood 2012;120(1):112–21.
2005;175(5):3424–30. 115. Coccia M, et al. IL-1β mediates chronic intestinal inflammation by
89. Murphy WJ, Kumar V, Bennett M. Rejection of bone marrow promoting the accumulation of IL-17A secreting innate lymphoid
allografts by mice with severe combined immune deficiency (SCID): cells and CD4(+) Th17 cells. J Experiment Med 2012;209(9):1595–
evidence that NK cells can mediate the specificity of marrow graft 609.
rejection. J Exp Med 1987;165:1212–7. 116. Revu S, et al. IL-23 and IL-1β drive human Th17 cell differentiation
90. Murphy WJ, Kumar V, Bennett M. Acute rejection of murine and metabolic reprogramming in absence of CD28 costimulation.
bone marrow allografts by natural killer cells and T cells: dif- Cell Rep 2018;22(10):2642–53.
ferences in kinetics and target antigens recognized. J Exp Med 117. Kullberg MC, et al. IL-23 plays a key role in Helicobacter hepat-
1987;166(5):1499–509. icus-induced T cell-dependent colitis. J Exp Med 2006;203(11):
91. Armstrong HE, et al. Prolonged survival of actively enhanced rat 2485–94.
renal allografts despite accelerated cellular infiltration and rapid 118. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage
induction of both class I and class II MHC antigens. J Exp Med activation: time for reassessment. F1000prime Rep. 2014;6:13.
1987;165(3):891–907. 119. Hutchinson JA, et al. Cutting edge: immunological consequences
92. Bradley JA, Mason DW, Morris PJ. Evidence that rat renal allografts and trafficking of human regulatory macrophages administered to
are rejected by cytotoxic T cells and not by nonspecific effectors. renal transplant recipients. J Immunol 2011;187(5):2072–8.
Transplantation 1985;39(2):169–75. 120. Puga I, et al. B cell-helper neutrophils stimulate the diversification
93. Kim J, et al. The activating immunoreceptor NKG2D and its and production of immunoglobulin in the marginal zone of the
ligands are involved in allograft transplant rejection. J Immunol spleen. Nat Immunol 2011;13(2):170–80.
2007;179(10):6416–20. 121. Leliefeld PH, Koenderman L, Pillay J. How neutrophils shape adap-
94. Kirwan SE, Burshtyn DN. Regulation of natural killer cell activity. tive immune responses. Front Immunol 2015;6:471.
Curr Opin Immunol 2007;19(1):46–54. 122. Chu VT, et al. Eosinophils promote generation and maintenance of
95. van Bergen J, et al. KIR-ligand mismatches are associated with immunoglobulin-A-expressing plasma cells and contribute to gut
reduced long-term graft survival in HLA-compatible kidney trans- immune homeostasis. Immunity 2014;40(4):582–93.
plantation. Am J Transplant 2011;11(9):1959–64. 123. Chu VT, et al. Eosinophils are required for the maintenance of plasma
96. Karre K. NK cells, MHC class I molecules and the missing self. Scand cells in the bone marrow. Nat Immunol 2011;12(2):151–9.
J Immunol 2002;55(3):221–8. 124. Wong TW, et al. Eosinophils regulate peripheral B cell numbers
97. Ljunggren HG, Karre K. In search of the ‘missing self’: MHC mol- in both mice and humans. J Immunol (Baltimore, MD: 1950)
ecules and NK cell recognition. Immunol Today 1990;11(7):237– 2014;192(8):3548–58.
44. 125. Hepworth MR, et al. Group 3 innate lymphoid cells mediate intesti-
98. Herberman RB, et al. Natural killer cells: characteristics and regula- nal selection of commensal bacteria-specific CD4+ T cells. Science
tion of activity. Imm Rev 1979;44:43–70. 2015;348(6238):1031–5.
99. Bix M, et al. Rejection of class I MHC-deficient haemopoietic cells by 126. Hepworth MR, et al. Innate lymphoid cells regulate CD4+ T-cell
irradiated MHC-matched mice. Nature 1991;349:329–31. responses to intestinal commensal bacteria. Nature 2013;498(7452):
100. Eberl G, et al. Innate lymphoid cells: a new paradigm in immunol- 113–7.
ogy. Science 2015;348(6237):aaa6566. 127. Oliphant CJ, et al. MHCII-Mediated dialog between group 2
101. Sonnenberg GF, Artis D. Innate lymphoid cell interactions with innate lymphoid cells and CD4+ T cells potentiates type 2 immu-
microbiota: implications for intestinal health and disease. Immunity nity and promotes parasitic helminth expulsion. Immunity
2012;37(4):601–10. 2014;41(2):283–95.
30 Kidney Transplantation: Principles and Practice

128. von Burg N, et al. Activated group 3 innate lymphoid cells pro- 157. Goulmy E. Minor histocompatibility antigens: from transplantation
mote T-cell-mediated immune responses. Proc Nat Acad Sci USA problems to therapy of cancer. Hum Immunol 2006;67(6):433–8.
2014;111(35):12835–40. 158. Gurley KE, Lowry RP, Clarke-Forbes RD. Immune mechanisms
129. Magri G, et al. Innate lymphoid cells integrate stromal and immuno- in organ allograft rejection: II. T helper cells, delayed type hyper-
logical signals to enhance antibody production by splenic marginal sensitivity and rejection of renal allografts. Transplantation
zone B cells. Nat Immunol 2014;15(4):354–64. 1983;36:401–5.
130. Peugh WN, et al. The role of H-2 and non-H-2 antigens and genes 159. Hall BM, DeSaxe I, Dorsch SE. The cellular basis of allograft rejection
in the rejection of murine cardiac allografts. Immunogenetics in vivo: restoration of first set rejection of heart grafts by T helper
1986;23(1):30–7. cells in irradiated rats. Transplantation 1983;36:700–5.
131. Flowers ME, et al. Comparative analysis of risk factors for acute 160. Lowry RP, Gurley KE, Forbes RD. Immune mechanisms in organ
graft-versus-host disease and for chronic graft-versus-host disease allograft rejection. I. Delayed-type hypersensitivity and lympho-
according to National Institutes of Health consensus criteria. Blood cytotoxicity in heart graft rejection. Transplantation 1983;36(4):
2011;117(11):3214–9. 391–401.
132. Dallman MJ, Mason DW, Webb M. Induction of Ia antigens on 161. Loveland BE, et al. Delayed type hypersensitivity and allograft rejec-
murine epidermal cells during the rejection of skin allografts. Eur J tion in the mouse: correlation of effector cell phenotype. J Exp Med
Immunol 1982;12:511–8. 1981;153:1044–57.
133. de Waal RMW, et al. Variable expression of Ia antigens on the 162. Steinman RM, Witmer MD. Lymphoid dendritic cells are potent stim-
vascular endothelium of mouse skin allografts. Nature (Lond) ulations of the primary mixed leucocyte reaction in mice. Proc Natl
1983;303:426–9. Acad Sci USA 1978;75:5132–6.
134. Fellous M, et al. Interferon-dependent induction of mRNA for the 163. Daar AS, et al. Demonstration and phenotypic characterisation
major histocompatibility antigens in human fibroblasts and lympho- of HLA-DR positive interstitial dendritic cells widely distributed
blastoid cells. Proc Natl Acad Sci USA 1982;79:3082–6. in human connective tissue. Transplant Proc 1983;XV(suppl. 1):
135. Fuggle SV, McWhinnie D, Morris PJ. Precise specificity of induced 311–5.
tubular class II antigens in renal allografts. Transplantation 164. Hart DN, Fabre JW. Demonstration and characterisation of Ia posi-
1987;44:214–20. tive dendritic cells in the interstitial connective tissues of the rat
136. Mason DW, Dallman MJ, Barclay AN. Graft-versus-host disease heart and other tissues, but not brain. J Exp Med 1981;154:347–61.
induces expression of Ia antigen in rat epidermal cells and gut epi- 165. Berry MR, et al. Renal sodium gradient orchestrates a dynamic anti-
thelium. Nature 1981;293:150–1. bacterial defense zone. Cell 2017;170(5):860–74.
137. Tewari MK, et al. A cytosolic pathway for MHC class II-restricted 166. Larsen CP, Morris PJ, Austyn JM. Migration of dendritic leukocytes
antigen processing that is proteasome and TAP dependent. Nat from cardiac allografts into host spleens: a novel pathway for initia-
Immunol 2005;6(3):287–94. tion of rejection. J Exp Med 1990;171(1):307–14.
138. Bevan MJ. Cross-priming for a secondary cytotoxic response to 167. Larsen CP, et al. Migration and maturation of Langerhans cells in
minor H antigens with H-2 congenic cells which do not cross-react skin transplants and explants. J Exp Med 1990;172:1483–93.
in the cytotoxic assay. J Exp Med 1976;143(5):1283–8. 168. Reis e Sousa C, Stahl PD, Austyn JM. Phagocytosis of antigens by
139. Chicz RM, et al. Predominant naturally processed peptides bound to Langerhans cells in vitro. J Exp Med 1993;178:509–19.
HLA-DR1 are derived from MHC-related molecules and are hetero- 169. Steinman RM, et al. Dendritic cells are the peripheral stimula-
geneous in size. Nature 1992;358(6389):764–8. tors of the primary mixed leukocyte reaction in mice. J Exp Med
140. O’Brien SJ, et al. Genetic basis for species vulnerability in the chee- 1983;157:613–27.
tah. Science 1985;227:1428–34. 170. Steinman RM, Hemmi H. Dendritic cells: translating innate to adap-
141. Felix NJ, et al. Alloreactive T cells respond specifically to multiple dis- tive immunity. Curr Top Microbiol Immunol 2006;311:17–58.
tinct peptide-MHC complexes. Nat Immunol 2007;8(4):388–97. 171. Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic
142. Ely LK, et al. T-cells behaving badly: structural insights into alloreac- cells. Annu Rev Immunol 2003;21:685–711.
tivity and autoimmunity. Curr Opin Immunol 2008;20(5):575–80. 172. Trombetta ES, Mellman I. Cell biology of antigen processing in vitro
143. Carosella ED, Gregori S, LeMaoult J. The tolerogenic interplay(s) and in vivo. Annu Rev Immunol 2005;23:975–1028.
among HLA-G, myeloid APCs, and regulatory cells. Blood 173. Mahnke K, Enk AH. Dendritic cells: key cells for the induction of
2011;118(25):6499–505. regulatory T cells? Curr Top Microbiol Immunol 2005;293:133–50.
144. Deschaseaux F, et al. HLA-G in organ transplantation: towards clini- 174. Rotschke O, et al. On the nature of peptides involved in T cell allore-
cal applications. Cell Mol Life Sci 2011;68(3):397–404. activity. J Exp Med 1991;174:1059–71.
145. Milner CM, Campbell RD. Genetic organization of the human MHC 175. Golding H, Singer A. Role of accessory cell processing and presenta-
class III region. Front Biosci 2001;6:D914–26. tion of shed H-2 alloantigens in allospecific cytotoxic T lymphocyte
146. Turner D, et al. Cytokine gene polymorphism and heart transplant responses. J Immunol 1984;133:597–605.
rejection. Transplantation 1997;64(5):776–9. 176. Parham P, et al. Inhibition of alloreactive cytotoxic T lymphocytes by
147. Scott DM, et al. Identification of a mouse male-specific transplanta- peptides from the OL2 domain of HLA-A2. Nature 1987;325:625–8.
tion antigen H-Y. Nature 1995;376:695–8. 177. Morris GP, Ni PP, Allen PM. Alloreactivity is limited by the
148. Wang W, et al. Human H-Y: a male-specific histocompatibility anti- endogenous peptide repertoire. Proc Natl Acad Sci USA 2011;
gen derived from the SMCY protein. Science 1995;269:1588–90. 108(9):3695–700.
149. Greenfield A, et al. An H-YDb epitope is encoded by a novel mouse Y 178. Wise M, et al. CD4 T cells can reject major histocompatibility complex
chromosome gene. Nat Genet 1996;14. 474-48. class I-incompatible skin grafts. Eur J Immunol 1999;29(1):156–67.
150. Scott DM, et al. Why do some females reject males? The molecu- 179. Auchincloss H, et al. The role of “indirect” recognition in initiating
lar basis for male-specific graft rejection. J Mol Med 1997;75(2): rejection of skin grafts from major histocompatibility complex class
103–14. II-deficient mice. Proc Natl Acad Sci USA 1993;90(8):3373–7.
151. Superina RA, et al. Assessment of primarily vascularized cardiac 180. Lee RS, et al. Indirect recognition by helper cells can induce
allografts in mice. Transplantation 1986;42:226–7. donor-specific cytotoxic T lymphocytes in vivo. J Exp Med
152. Fabre JW, Morris PJ. Studies on the specific suppression of 1994;179(3):865–72.
renal allograft rejection in presensitised rats. Transplantation 181. Lee RS, et al. CD8+ effector cells responding to residual class I anti-
1975;19:121–33. gens, with help from CD4+ cells stimulated indirectly, cause rejec-
153. Steinmuller D, Wachtal SS. Passenger leukocytes and induction of tion of “major histocompatibility complex-deficient” skin grafts.
allograft immunity. Transplant Proc 1980;12:100–6. Transplantation 1997;63(8):1123–33.
154. Hart DN, Fabre JW. Kidney-specific alloantigen system in the 182. Najafian N, et al. Enzyme-linked immunosorbent spot assay analysis
rat: characterisation and role in transplantation. J Exp Med of peripheral blood lymphocyte reactivity to donor HLA-DR peptides:
1980;151:651–66. potential novel assay for prediction of outcomes for renal transplant
155. Roopenian D, Choi EY, Brown A. The immunogenomics of minor recipients. J Am Soc Nephrol 2002;13(1):252–9.
histocompatibility antigens. Immunol Rev 2002;190:86–94. 183. Poggio ED, et al. Alloreactivity in renal transplant recipients with
156. Simpson E, Roopenian D, Goulmy E. Much ado about minor histo- and without chronic allograft nephropathy. J Am Soc Nephrol
compatibility antigens. Immunol Today 1998;19:108–12. 2004;15(7):1952–60.
2 • The Immunology of Transplantation 31

184. Baker RJ, et al. Loss of direct and maintenance of indirect allore- 211. Schwartz RH. T cell clonal anergy. Curr Opin Immunol 1997;9:
sponses in renal allograft recipients: implications for the pathogenesis 351–7.
of chronic allograft nephropathy. J Immunol 2001;167(12):7199– 212. Carlin LM, et al. Secretion of IFN-gamma and not IL-2 by anergic
206. human T cells correlates with assembly of an immature immune
185. Hornick PI, et al. Significant frequencies of T cells with indirect anti- synapse. Blood 2005;106(12):3874–9.
donor specificity in heart graft recipients with chronic rejection. Cir- 213. Frasca L, et al. Anergic T cells effect linked suppression. Eur J Immu-
culation 2000;101(20):2405–10. nol 1997;27:3191–7.
186. Waanders MM, et al. Monitoring of indirect allorecognition: wishful 214. Lombardi G, et al. Anergic T cells as suppressor cells in vitro. Science
thinking or solid data? Tissue Antigens 2008;71(1):1–15. 1994;264:1587–9.
187. Smith HJ, et al. T Lymphocyte responses to nonpolymorphic HLA- 215. Cobbold SP, et al. Connecting the mechanisms of T-cell regulation:
derived peptides are associated with chronic renal allograft dysfunc- dendritic cells as the missing link. Immunol Rev 2010;236:203–
tion. Transplantation 2011;91(3):279–86. 18.
188. Shirwan H. Chronic allograft rejection. Do the Th2 cells preferen- 216. Linsley PS, Clark EA, Ledbetter JA. T-cell antigen CD28 mediates
tially induced by indirect alloantigen recognition play a dominant adhesion with B cells by interacting with activation antigen B7/
role? Transplantation 1999;68(6):715–26. BB-1. Proc Natl Acad Sci USA 1990;87:5031–5.
189. Yamada A, et al. Further analysis of the T-cell subsets and path- 217. Archdeacon P, et al. Summary of the US FDA approval of belatacept.
ways of murine cardiac allograft rejection. Am J Transplant Am J Transplant 2012;12(3):554–62.
2003;3(1):23–7. 218. Dharnidharka VR. Costimulation blockade with belatacept in renal
190. Cramer DV, et al. Cardiac transplantation in the rat I. The effect of transplantation. N Engl J Med 2005;353(19):2085–6. Author
histocompatibility differences on graft arteriosclerosis. Transplanta- reply:2085–6.
tion 1989;47:414–9. 219. Clarkson MR, Sayegh MH. T-cell costimulatory pathways in allograft
191. Nadazdin O, et al. Contributions of direct and indirect alloresponses rejection and tolerance. Transplantation 2005;80(5):555–63.
to chronic rejection of kidney allografts in nonhuman primates. 220. Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revis-
J Immunol 2011;187(9):4589–97. ited. Annu Rev Immunol 2005;23:515–48.
192. Bestard O, et al. Circulating alloreactive T cells correlate with graft 221. Riley JL, June CH. The CD28 family: a T-cell rheostat for therapeutic
function in longstanding renal transplant recipients. J Am Soc control of T-cell activation. Blood 2005;105(1):13–21.
Nephrol 2008;19(7):1419–29. 222. Kawai K, et al. Skin allograft rejection in CD28-deficient mice. Trans-
193. Sauve D, et al. Alloantibody production is regulated by CD4+ T cells’ plantation 1996;61(3):352–5.
alloreactive pathway, rather than precursor frequency or Th1/Th2 223. Sharpe AH, Abbas AK. T-cell costimulation—biology, therapeutic
differentiation. Am J Transplant 2004;4(8):1237–45. potential, and challenges. N Engl J Med 2006;355(10):973–5.
194. Conlon TM, et al. Germinal center alloantibody responses are medi- 224. Turka LA, et al. T-cell activation by the CD28 ligand B7 is required
ated exclusively by indirect-pathway CD4 T follicular helper cells. for cardiac allograft rejection in vivo. Proc Natl Acad Sci USA
J Immunol 2012;188(6):2643–52. 1992;89(22):11102–5.
195. Lachmann N, et al. Antihumoral rejection therapy by proteasome 225. Pearson TC, et al. Transplantation tolerance induced by CTLA-4 Ig.
inhibitor bortezomib: a case series. Clin Transpl 2009:351–8. Transplantation 1994;57:1701–6.
196. Hourmant M, et al. Frequency and clinical implications of 226. Lenschow DJ, et al. CD28/B7 regulation of Th1 and Th2 subsets in
development of donor-specific and non-donor-specific HLA the development of autoimmune diabetes. Immunity 1996;5:285–
antibodies after kidney transplantation. J Am Soc Nephrol 93.
2005;16(9):2804–12. 227. Collins AV, et al. The interaction properties of costimulatory mol-
197. Herrera OB, et al. A novel pathway of alloantigen presentation by ecules revisited. Immunity 2002;17(2):201–10.
dendritic cells. J Immunol 2004;173(8):4828–37. 228. Schwartz JC, et al. Structural mechanisms of costimulation. Nat
198. Bedford P, Garner K, Knight SC. MHC class II molecules transferred Immunol 2002;3(5):427–34.
between allogeneic dendritic cells stimulate primary mixed leuko- 229. Fallarino F, Fields PE, Gajewski TF. B7-1 engagement of cytotoxic
cyte reactions. Int Immunol 1999;11(11):1739–44. T lymphocyte antigen 4 inhibits T cell activation in the absence of
199. Harshyne LA, et al. Dendritic cells acquire antigens from live cells for CD28. J Exp Med 1998;188(1):205–10.
cross-presentation to CTL. J Immunol 2001;166(6):3717–23. 230. Krummel MF, Allison JP. CTLA-4 engagement inhibits IL-2 accumu-
200. Russo V, et al. Acquisition of intact allogeneic human leukocyte lation and cell cycle progression upon activation of resting T cells.
antigen molecules by human dendritic cells. Blood 2000;95(11): J Exp Med 1996;183(6):2533–40.
3473–7. 231. Schneider H, et al. Reversal of the TCR stop signal by CTLA-4. Sci-
201. Love PE, Hayes SM. ITAM-mediated signaling by the T-cell antigen ence 2006;313(5795):1972–5.
receptor. Cold Spring Harb Perspect Biol 2010;2(6):a002485. 232. Wing K, et al. CTLA-4 control over Foxp3+ regulatory T cell func-
202. Wang H, et al. ZAP-70: an essential kinase in T-cell signaling. Cold tion. Science 2008;322(5899):271–5.
Spring Harb Perspect Biol 2010;2(5):a002279. 233. Ise W, et al. CTLA-4 suppresses the pathogenicity of self antigen-
203. Balagopalan L, et al. The LAT story: a tale of cooperativity, coor- specific T cells by cell-intrinsic and cell-extrinsic mechanisms. Nat
dination, and choreography. Cold Spring Harb Perspect Biol Immunol 2010;11(2):129–35.
2010;2(8):a005512. 234. Vincenti F, et al. Three-year outcomes from BENEFIT, a randomized,
204. Jordan MS, Koretzky GA. Coordination of receptor signaling in mul- active-controlled, parallel-group study in adult kidney transplant
tiple hematopoietic cell lineages by the adaptor protein SLP-76. Cold recipients. Am J Transplant 2012;12(1):210–7.
Spring Harb Perspect Biol 2010;2(4):a002501. 235. Pestana JO, et al. Three-year outcomes from BENEFIT-EXT: a phase
205. Wucherpfennig KW, et al. Structural biology of the T-cell receptor: III study of belatacept versus cyclosporine in recipients of extended
insights into receptor assembly, ligand recognition, and initiation of criteria donor kidneys. Am J Transplant 2012;12(3):630–9.
signaling. Cold Spring Harb Perspect Biol 2010;2(4):a005140. 236. Masson P, et al. Belatacept for kidney transplant recipients. Cochrane
206. Fernandes RA, et al. What controls T cell receptor phosphorylation? Database Syst Rev 2014;11:CD010699.
Cell 2010;142(5):668–9. 237. Kim EJ, et al. Costimulation blockade alters germinal center
207. Lamb JR, et al. Induction of tolerance in influenza virus-immune T responses and prevents antibody-mediated rejection. Am J Trans-
lymphocyte clones with synthetic peptides of influenza hemaggluti- plant 2014;14(1):59–69.
nin. J Exp Med 1983;157:1434–47. 238. Linterman MA, Vinuesa CG. Signals that influence T follicular
208. Jenkins MK, et al. Molecular events in the induction of a nonrespon- helper cell differentiation and function. Semin Immunopathol
sive state in interleukin 2-producing helper T-lymphocyte clones. 2010;32(2):183–96.
Proc Natl Acad Sci USA 1987;84:5409–13. 239. Zhu Y, Yao S, Chen L. Cell surface signaling molecules in the control
209. Jenkins MK, Schwartz RH. Antigen presentation by chemically mod- of immune responses: a tide model. Immunity 2011;34(4):466–
ified splenocytes induces antigen-specific T cell unresponsiveness 78.
in vitro and in vivo. J Exp Med 1987;165:302–19. 240. Larsen CP, et al. Long term acceptance of skin and cardiac
210. Schwartz RH. A cell culture model for T lymphocyte clonal anergy. allografts after blocking CD40 and CD28 pathways. Nature
Science 1990;248:1349–56. 1996;381:434–8.
32 Kidney Transplantation: Principles and Practice

241. Larsen CP, et al. CD40-gp39 interactions play a critical role during 270. Salvadori M, et al. FTY720 versus MMF with cyclosporine in de
allograft rejection: suppression of allograft rejection by blockade of novo renal transplantation: a 1-year, randomized controlled trial in
the CD40-gp39 pathway. Transplantation 1996;61(1):4–9. Europe and Australasia. Am J Transplant 2006;6(12):2912–21.
242. Kirk AD, et al. Treatment with humanized monoclonal antibody 271. Tedesco-Silva H, et al. Randomized controlled trial of FTY720
against CD154 prevents acute renal allograft rejection in nonhu- versus MMF in de novo renal transplantation. Transplantation
man primates. Nat Med 1999;5(6):686–93. 2006;82(12):1689–97.
243. Badell IR, et al. Nondepleting anti-CD40-based therapy pro- 272. Hoitsma AJ, et al. FTY720 combined with tacrolimus in de novo
longs allograft survival in nonhuman primates. Am J Transplant renal transplantation: 1-year, multicenter, open-label randomized
2012;12(1):126–35. study. Nephrol Dial Transplant 2011;26(11):3802–5.
244. Weaver TA, et al. Alefacept promotes co-stimulation blockade-based 273. Butcher EC. The regulation of lymphocyte traffic. Curr Top Microbiol
allograft survival in nonhuman primates. Nat Med 2009;15(7): Immunol 1986;128:85–122.
746–9. 274. Picker LJ, Butcher EC. Physiological and molecular mechanisms of
245. Lo DJ, et al. Selective targeting of human alloresponsive CD8+ lymphocyte homing. Annu Rev Immunol 1992;10:561–91.
effector memory T cells based on CD2 expression. Am J Transplant 275. Santamaria-Babi LF, et al. Migration od skin-homing T cells across
2011;11(1):22–33. cytokine-activated human endothelial cell layers involves interac-
246. Badell IR, et al. LFA-1-specific therapy prolongs allograft survival in tion of the cutaneous lymphocyte-associated antigen (CLA), the
rhesus macaques. J Clin Invest 2010;120(12):4520–31. very late antigen-4 (VLA-4) and the lymphocyte function-associated
247. Grgic I, et al. Blockade of T-lymphocyte KCa3.1 and Kv1.3 channels antigen-1 (LFA-1). J Immunol 1995;154:1543–50.
as novel immunosuppression strategy to prevent kidney allograft 276. Marelli-Berg FM, et al. Antigen recognition influences transendothe-
rejection. Transplant Proc 2009;41(6):2601–6. lial migration of CD4+ T cells. J Immunol 1999;162:696–703.
248. De Jager PL, et al. The role of the CD58 locus in multiple sclerosis. 277. Cosimi AB, Conti D, e.a. Delmonico FL. In vivo effects of monoclo-
Proc Natl Acad Sci USA 2009;106(13):5264–9. nal antibody to ICAM-1 (CD54) in nonhuman primates with renal
249. Bloomgren G, et al. Risk of natalizumab-associated progressive mul- allografts. J Immunol 1990;144:4604–12.
tifocal leukoencephalopathy. N Engl J Med 2012;366(20):1870–80. 278. Dallman MJ, et al. Lymphokine production in allografts-analysis of
250. Nashan B, et al. Randomised trial of basiliximab versus placebo for RNA by northern blotting. Transplant Proc 1989;20:296–8.
control of acute cellular rejection in renal allograft recipients. CHIB. 279. Hourmant M, et al. A randomized multicenter trial comparing leu-
201 International Study Group. Lancet 1997;350(9086):1193–8. kocyte function-associated antigen-1 monoclonal antibody with
251. Vincenti F, et al. Interleukin-2-receptor blockade with daclizumab to rabbit antithymocyte globulin as induction treatment in first kidney
prevent acute rejection in renal transplantation. Daclizumab Triple transplantations. Transplantation 1996;62(11):1565–70.
Therapy Study Group. N Engl J Med 1998;338(3):161–5. 280. Yang HC, et al. In situ expression of platelet-derived growth factor
252. Steiger J, et al. IL-2 knockout recipient mice reject islet cell allografts. (PDGF-beta) during chronic rejection is abolished by retransplanta-
J Immunol 1995;155:489–98. tion. J Surg Res 1995;59:205–10.
253. Saleem S, et al. Acute rejection of vascularized heart allografts in the 281. Morikawa M, et al. Cardiac allografts in rat recipients with simultane-
absence of IFNg. Transplantation 1996;62:1908–11. ous use of anti-ICAM-1 and anti-LFA-1 monoclonal antibodies leads
254. Dai Z, Lakkis FG. The role of cytokines, CTLA-4 and costimula- to accelerated graft loss. Immunopharmacology 1994;28:171–82.
tion in transplant tolerance and rejection. Curr Opin Immunol 282. Stepkowski SM, et al. An oral formulation for intracellular adhe-
1999;11:504–8. sion molecules-1 antisense oligonucleotides. Transplant Proc
255. Sakaguchi S, et al. Foxp3+ CD25+ CD4+ natural regulatory T cells 2001;33(7-8):3271.
in dominant self-tolerance and autoimmune disease. Immunol Rev 283. Nemoto T, et al. Small molecule selectin ligand inhibition
2006;212:8–27. improves outcome in ischemic acute renal failure. Kidney Int
256. Maloy KJ, Powrie F. Fueling regulation: IL-2 keeps CD4+ Treg cells 2001;60(6):2205–14.
fit. Nat Immunol 2005;6(11):1071–2. 284. Subramanian S, et al. Attenuation of renal ischemia-reperfusion injury
257. Warnecke G, et al. CD4+ regulatory T cells generated in vitro with with selectin inhibition in a rabbit model. Am J Surg 1999;178(6):
IFN-{gamma} and allogeneic APC inhibit transplant arteriosclero- 573–6.
sis. Am J Pathol 2010;177(1):464–72. 285. Hancock WW. Chemokines and transplant immunobiology. J Am
258. Sawitzki B, et al. IFN-gamma production by alloantigen-reactive Soc Nephrol 2002;13(3):821–4.
regulatory T cells is important for their regulatory function in vivo. 286. Hancock WW, et al. Chemokines and their receptors as markers of
J Exp Med 2005;201(12):1925–35. allograft rejection and targets for immunosuppression. Curr Opin
259. Wood KJ, Sawitzki B. Interferon gamma: a crucial role in the Immunol 2003;15(5):479–86.
function of induced regulatory T cells in vivo. Trends Immunol 287. Merani S, et al. Chemokines and their receptors in islet allograft
2006;27(4):183–7. rejection and as targets for tolerance induction. Cell Transplant
260. Mackay CR. Homing of naive, memory and effector lymphocytes. 2006;15(4):295–309.
Curr Opin Immunol 1993;5:423–7. 288. Oppenheim JJ, et al. The role of chemokines in transplantation. In:
261. Mackay CR. Immunological memory. Adv Immunol 1993;53:217– Tilney NL, Strom TB, Paul LC, editors. Transplantation Biology: Cellu-
65. lar and Molecular Aspects. Philadelphia: Lippincott-Raven Publishers;
262. Lakkis FG, et al. Immunologic ‘ignorance’ of vascularized organ 1996. p. 187–200.
transplants in the absence of secondary lymphoid tissue. Nat Med 289. Rossi D, Zlotnik A. The biology of chemokines and their receptors.
2000;6(6):686–8. Annu Rev Immunol 2000;18:217–42.
263. Lakkis FG. Where is the alloimmune response initiated? Am J Trans- 290. Lentsch AB, et al. Chemokine involvement in hepatic ischemia/
plant 2003;3(3):241–2. reperfusion injury in mice: roles for macrophage inflammatory pro-
264. Zhou P, et al. Secondary lymphoid organs are important but tein-2 and KC. Hepatology 1998;27:1172–7. [Corrected and repub-
not absolutely required for allograft responses. Am J Transplant lished article originally printed in Hepatology. 1998;27(2):507–12].
2003;3(3):259–66. 291. DeVries ME, Ran L, Kelvin D. On the edge: the physiological and
265. Ingham-Clark CL, et al. Lymphocyte infiltration patterns in rat pathophysiological role of chemokines during inflammatory and
small-bowel transplants. Transplant Proc 1990;22:2460. immunological responses. Semin Immunol 1999;11:95–104.
266. Kim PC, et al. Immunologic basis of small intestinal allograft rejec- 292. Fairchild RL, et al. Expression of chemokine genes during rejection
tion. Transplant Proc 1991;23:830. and long-term acceptance of cardiac allografts. Transplantation
267. Toogood GJ, et al. The immune response following small bowel 1997;63(12):1807–12.
transplantation II: a very early cytokine response in the gut associ- 293. Grandaliano G, et al. Monocyte chemotactic peptide-1 expression
ated lymphoid tissue. Transplantation 1997;63:1118–23. and monocyte infiltration in acute renal transplant rejection. Trans-
268. Toogood GJ, et al. The immune response following small bowel plantation 1997;63(3):414–20.
transplantation I. An unusual pattern of cytokine expression. Trans- 294. Gao JW, et al. Polymorphisms in cytotoxic T lymphocyte associated
plantation 1996;62:851–5. antigen-4 influence the rate of acute rejection after renal transplan-
269. Baddoura FK, et al. Lymphoid neogenesis in murine cardiac allografts tation in 167 Chinese recipients. Transpl Immunol 2012;26(4):
undergoing chronic rejection. Am J Transplant 2005;5(3):510–6. 207–11.
2 • The Immunology of Transplantation 33

295. Hancock WW, et al. Donor-derived IP-10 initiates development of 323. Kerjaschki D, et al. Lymphatic neoangiogenesis in human kidney
acute allograft rejection. J Exp Med 2001;193(8):975–80. transplants is associated with immunologically active lymphocytic
296. O’Boyle G, Ali S, Kirby JA. Chemokines in transplantation: what can infiltrates. J Am Soc Nephrol 2004;15(3):603–12.
atypical receptors teach us about anti-inflammatory therapy? Trans- 324. Wehner JR, et al. B cells and plasma cells in coronaries of chronically
plant Rev (Orlando) 2011;25(4):136–44. rejected cardiac transplants. Transplantation 2010;89(9):1141–8.
297. Hayry P, Defendi V. Mixed lymphocyte cultures produce effector cells: 325. Patel R, Terasaki PI. Significance of the positive crossmatch test in
model in vitro for allograft rejection. Science 1970;168(927):133–5. kidney transplantation. N Engl J Med 1969;280(14):735–9.
298. Hodes RJ, Svedmyr EA. Specific cytotoxicity of H-2-incompatible 326. Gebel HM, Liwski RS, Bray RA. Technical aspects of HLA antibody
mouse lymphocytes following mixed culture in vitro. Transplanta- testing. Curr Opin Organ Transplant 2013;18(4):455–62.
tion 1970;9(5):470–7. 327. Loupy A, Hill GS, Jordan SC. The impact of donor-specific anti-
299. Mason DW, Morris PJ. Inhibition of the accumulation, in rat kidney HLA antibodies on late kidney allograft failure. Nat Rev Nephrol
allografts, of specific—but not nonspecific—cytotoxic cells by cyclo- 2012;8(6):348–57.
sporine. Transplantation 1984;37(1):46–51. 328. Montgomery RA, et al. HLA incompatible renal transplantation.
300. Cobbold SP, et al. Therapy with monoclonal antibodies by elimina- Curr Opin Organ Transplant 2012;17(4):386–92.
tion of T cell subsets in vivo. Nature 1984;312:548–51. 329. Mohan S, et al. Donor-specific antibodies adversely affect kidney
301. Madsen JC, et al. The effect of anti-L3T4 monoclonal antibody treat- allograft outcomes. J Am Soc Nephrol 2012;23(12):2061–71.
ment on first-set rejection of murine cardiac allografts. Transplanta- 330. Hidalgo LG, et al. De novo donor-specific antibody at the time of kid-
tion 1987;44(6):849–52. ney transplant biopsy associates with microvascular pathology and
302. Tilney NL, et al. Mechanisms of rejection and prolongation of vascu- late graft failure. Am J Transplant 2009;9(11):2532–41.
larized organ allografts. Immunol Rev 1984;77:185–216. 331. Norin AJ, et al. Poor kidney allograft survival associated with posi-
303. Selvaggi G, et al. The role of the perforin and Fas pathways of cytotox- tive B cell - Only flow cytometry cross matches: a ten year single cen-
icity in skin graft rejection. Transplantation 1996;62(12):1912–5. ter study. Hum Immunol 2013;74(10):1304–12.
304. Schulz M, et al. Acute rejection of vascular heart allografts by 332. Zou Y, et al. Antibodies against MICA antigens and kidney-transplant
perforin-deficient mice. Eur J Immunol 1995;25(2):474–80. rejection. N Engl J Med 2007;357(13):1293–300.
305. Walsh CM, et al. Cell-mediated cytotoxicity results from, but 333. Dragun D, et al. Angiotensin II type 1-receptor activating antibodies
may not be critical for, primary allograft rejection. J Immunol in renal-allograft rejection. N Engl J Med 2005;352(6):558–69.
1996;156:1436–41. 334. Giral M, et al. Pretransplant sensitization against angiotensin II
306. Mintz B, Silvers WK. “Intrinsic” immunological tolerance in allo- type 1 receptor is a risk factor for acute rejection and graft loss. Am J
phenic mice. Science 1967;158(807):1484–6. Transplant 2013;13(10):2567–76.
307. Mintz B, Silvers WK. Histocompatibility antigens on melanoblasts 335. Sigdel TK, et al. Non-HLA antibodies to immunogenic epitopes pre-
and hair follicle cells. Cell-localized homograft rejection in allophenic dict the evolution of chronic renal allograft injury. J Am Soc Nephrol
skin grafts. Transplantation 1970;9(5):497–505. 2012;23(4):750–63.
308. Snider ME, Steinmuller D. Nonspecific tissue destruction as a conse- 336. Porcheray F, et al. Polyreactive antibodies developing amidst
quence of cytotoxic T lymphocyte interaction with antigen-specific humoral rejection of human kidney grafts bind apoptotic cells and
target cells. Transplant Proc 1987;19(1 Pt 1):421–3. activate complement. Am J Transplant 2013;13(10):2590–600.
309. Sutton R, et al. The specificity of rejection and the absence of suscep- 337. Gao B, et al. Pretransplant IgG reactivity to apoptotic cells corre-
tibility of pancreatic islet beta cells to nonspecific immune destruc- lates with late kidney allograft loss. Am J Transplant 2014;14(7):
tion in mixed strain islets grafted beneath the renal capsule in the 1581–91.
rat. J Exp Med 1989;170(3):751–62. 338. Batista FD, Neuberger MS. B cells extract and present immobi-
310. Rosenberg AS, Singer A. Cellular basis of skin allograft rejection: an lized antigen: implications for affinity discrimination. EMBO J
in vivo model of immune-mediated tissue destruction. Annu Rev 2000;19(4):513–20.
Immunol 1992;10:333–58. 339. Batista FD, Iber D, Neuberger MS. B cells acquire antigen from target
311. Pratt JR, Basheer SA, Sacks SH. Local synthesis of complement cells after synapse formation. Nature 2001;411(6836):489–94.
component C3 regulates acute renal transplant rejection. Nat Med 340. Janeway CA, Ron J, Katz ME. The B cell is the initiating antigen-
2002;8(6):582–7. presenting cell in peripheral lymph nodes. J Immunol 1987;
312. Pavlov V, et al. Donor deficiency of decay-accelerating factor acceler- 138(4):1051–5.
ates murine T cell-mediated cardiac allograft rejection. J Immunol 341. Ron Y, Sprent J. T cell priming in vivo: a major role for B cells in pre-
2008;181(7):4580–9. senting antigen to T cells in lymph nodes. J Immunol 1987;138(9):
313. Peng Q, et al. Local production and activation of complement up- 2848–56.
regulates the allostimulatory function of dendritic cells through 342. Giles JR, et al. B cell-specific MHC class II deletion reveals multiple
C3a-C3aR interaction. Blood 2008;111(4):2452–61. nonredundant roles for B cell antigen presentation in murine lupus.
314. Strainic MG, et al. Locally produced complement fragments C5a and J Immunol 2015;195(6):2571–9.
C3a provide both costimulatory and survival signals to naive CD4+ 343. Zeng Q, et al. B cells mediate chronic allograft rejection indepen-
T cells. Immunity 2008;28(3):425–35. dently of antibody production. J Clin Invest 2014;124(3):1052–6.
315. Jacob J, Kelsoe G. In situ studies of the primary immune response 344. Sarwal M, et al. Molecular heterogeneity in acute renal allograft
to (4-hydroxy-3-nitrophenyl)acetyl. II. A common clonal origin for rejection identified by DNA microarray profiling. N Engl J Med
periarteriolar lymphoid sheath-associated foci and germinal centers. 2003;349(2):125–38.
J Exp Med 1992;176(3):679–87. 345. Khatri P, et al. A common rejection module (CRM) for acute rejection
316. Di Noia JM, Neuberger MS. Molecular mechanisms of antibody across multiple organs identifies novel therapeutics for organ trans-
somatic hypermutation. Annu Rev Biochem 2007;76:1–22. plantation. J Exp Med 2013;210(11):2205–21.
317. Goodnow CC, et al. Control systems and decision making for anti- 346. Deola S, et al. Helper B cells promote cytotoxic T cell survival and
body production. Nat Immunol 2010;11(8):681–8. proliferation independently of antigen presentation through CD27/
318. Vinuesa CG, et al. T cells and follicular dendritic cells in germinal cen- CD70 interactions. J Immunol 2008;180(3):1362–72.
ter B-cell formation and selection. Immunol Rev 2010;237(1):72– 347. Venner JM, et al. Molecular landscape of T cell-Mediated rejection in
89. human kidney transplants: prominence of CTLA4 and PD ligands.
319. Crotty S. Follicular helper CD4 T cells (TFH). Annu Rev Immunol Am J Transplant 2014;14(11):2565–76.
2011;29:621–63. 348. Edwards JC, Cambridge G. B-cell targeting in rheumatoid arthritis and
320. Nutt SL, et al. The generation of antibody-secreting plasma cells. Nat other autoimmune diseases. Nat Rev Immunol 2006;6(5):394–403.
Rev Immunol 2015;15(3):160–71. 349. Claes N, et al. B cells are multifunctional players in multiple sclerosis
321. Cassese G, et al. Inflamed kidneys of NZB / W mice are a major site for pathogenesis: insights from therapeutic interventions. Front Immu-
the homeostasis of plasma cells. Eur J Immunol 2001;31(9):2726– nol 2015;6:642.
32. 350. Hinman RM, Smith MJ, Cambier JC. B cells and type 1 diabetes ...in
322. Thaunat O, et al. Lymphoid neogenesis in chronic rejection: evi- mice and men. Immunol Lett 2014;160(2):128–32.
dence for a local humoral alloimmune response. Proc Natl Acad Sci 351. Gonzalez M, et al. The sequential role of lymphotoxin and B cells in the
USA 2005;102(41):14723–8. development of splenic follicles. J Exp Med 1998;187(7):997–1007.
34 Kidney Transplantation: Principles and Practice

352. Golovkina TV, et al. Organogenic role of B lymphocytes in mucosal 381. Matsushita T, et al. A novel splenic B1 regulatory cell subset sup-
immunity. Science 1999;286(5446):1965–8. presses allergic disease through phosphatidylinositol 3-kinase-Akt
353. Ngo VN, Cornall RJ, Cyster JG. Splenic T zone development is B cell pathway activation. J Allergy Clin Immunol 2016;138(4):1170–82.
dependent. J Exp Med 2001;194(11):1649–60. 382. Shen P, et al. IL-35-producing B cells are critical regulators of
354. Moseman EA, et al. B cell maintenance of subcapsular sinus macro- immunity during autoimmune and infectious diseases. Nature
phages protects against a fatal viral infection independent of adap- 2014;507(7492):366–70.
tive immunity. Immunity 2012;36(3):415–26. 383. Braza F, et al. A regulatory CD9(+) B-cell subset inhibits HDM-induced
355. Angeli V, et al. B cell-driven lymphangiogenesis in inflamed allergic airway inflammation. Allergy 2015;70(11):1421–31.
lymph nodes enhances dendritic cell mobilization. Immunity 384. Viklicky O, et al. B-cell-related biomarkers of tolerance are up-
2006;24(2):203–15. regulated in rejection-free kidney transplant recipients. Transplan-
356. Drayton DL, et al. Lymphoid organ development: from ontogeny to tation 2013;95(1):148–54.
neogenesis. Nat Immunol 2006;7(4):344–53. 385. Chesneau M, et al. Unique B cell differentiation profile in tolerant kid-
357. Luther SA, et al. BLC expression in pancreatic islets causes B cell ney transplant patients. Am J Transplant 2014;14(1):144–55.
recruitment and lymphotoxin-dependent lymphoid neogenesis. 386. Shabir S, et al. Transitional B lymphocytes are associated with pro-
Immunity 2000;12(5):471–81. tection from kidney allograft rejection: a prospective study. Am J
358. Espeli M, et al. Local renal autoantibody production in lupus nephri- Transplant 2015;15(5):1384–91.
tis. J Am Soc Nephrol 2011;22(2):296–305. 387. Cherukuri A, et al. Immunologic human renal allograft injury asso-
359. Shen P, Fillatreau S. Antibody-independent functions of B cells: a ciates with an altered IL-10/TNF-alpha expression ratio in regula-
focus on cytokines. Nat Rev Immunol 2015;15(7):441–51. tory B cells. J Am Soc Nephrol 2014;25(7):1575–85.
360. Kopf M, et al. Impaired immune and acute-phase responses in inter- 388. Pallier A, et al. Patients with drug-free long-term graft function
leukin-6-deficient mice. Nature 1994;368(6469):339–42. display increased numbers of peripheral B cells with a memory and
361. Barr TA, et al. TLR and B cell receptor signals to B cells differentially inhibitory phenotype. Kidney Int 2010;78(5):503–13.
program primary and memory Th1 responses to Salmonella enterica. 389. Sis B, et al. Endothelial gene expression in kidney transplants with
J Immunol 2010;185(5):2783–9. alloantibody indicates antibody-mediated damage despite lack of
362. Barr TA, et al. B cell depletion therapy ameliorates autoimmune C4d staining. Am J Transplant 2009;9(10):2312–23.
disease through ablation of IL-6-producing B cells. J Exp Med 390. Haas M. Pathology of C4d-negative antibody-mediated rejection in
2012;209(5):1001–10. renal allografts. Curr Opin Organ Transplant 2013;18(3):319–26.
363. Menard LC, et al. B cells amplify IFN-gamma production by T cells via 391. Sis B, Halloran PF. Endothelial transcripts uncover a previously
a TNF-alpha-mediated mechanism. J Immunol 2007;179(7):4857– unknown phenotype: C4d-negative antibody-mediated rejection.
66. Curr Opin Organ Transplant 2010;15(1):42–8.
364. Bao Y, et al. Identification of IFN-gamma-producing innate B cells. 392. Halloran PF, et al. Microarray diagnosis of antibody-mediated rejec-
Cell Res 2014;24(2):161–76. tion in kidney transplant biopsies: an international prospective study
365. Rauch PJ, et al. Innate response activator B cells protect against (INTERCOM). Am J Transplant 2013;13(11):2865–74.
microbial sepsis. Science 2012;335(6068):597–601. 393. Jindra PT, et al. HLA class I antibody-mediated endothelial cell prolif-
366. Weber GF, et al. Pleural innate response activator B cells protect eration via the mTOR pathway. J Immunol 2008;180(4):2357–66.
against pneumonia via a GM-CSF-IgM axis. J Exp Med 2014;211(6): 394. Smith RN, Colvin RB. Chronic alloantibody mediated rejection.
1243–56. Semin Immunol 2012;24(2):115–21.
367. Hilgendorf I, et al. Innate response activator B cells aggravate ath- 395. Loupy A, et al. Complement-binding anti-HLA antibodies and
erosclerosis by stimulating T helper-1 adaptive immunity. Circula- kidney-allograft survival. N Engl J Med 2013;369(13):1215–26.
tion 2014;129(16):1677–87. 396. Sicard A, et al. Detection of C3d-binding donor-specific anti-HLA
368. Griffin DO, Holodick NE, Rothstein TL. Human B1 cells in umbilical antibodies at diagnosis of humoral rejection predicts renal graft loss.
cord and adult peripheral blood express the novel phenotype CD20+ J Am Soc Nephrol 2015;26(2):457–67.
CD27+ CD43+ CD70. J Exp Med 2011;208(1):67–80. 397. Cornell LD, et al. Positive crossmatch kidney transplant recipients
369. Martin F, Kearney JF. B1 cells: similarities and differences with other treated with eculizumab: outcomes beyond 1 year. Am J Transplant
B cell subsets. Curr Opin Immunol 2001;13(2):195–201. 2015;15(5):1293–302.
370. Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, and func- 398. Eskandary F, et al. Complement inhibition as potential new therapy
tion. Immunity 2015;42(4):607–12. for antibody-mediated rejection. Transpl Int 2016;29(4):392–402.
371. Khan AR, et al. PD-L1hi B cells are critical regulators of humoral 399. Kulkarni S, et al. Eculizumab therapy for chronic antibody-mediated
immunity. Nat Commun 2015;6:5997. injury in kidney transplant recipients: a pilot randomized controlled
372. Chesneau M, et al. Tolerant kidney transplant patients produce B trial. Am J Transplant 2017;17(3):682–91.
cells with regulatory properties. J Am Soc Nephrol 2015;26(10): 400. Spiegelberg HL. Biological role of different antibody classes. Int Arch
2588–98. Allergy Appl Immunol 1989;90(Suppl. 1):22–7.
373. DiLillo DJ, Matsushita T, Tedder TF. B10 cells and regulatory B cells 401. Nimmerjahn F, Ravetch JV. Fc-receptors as regulators of immunity.
balance immune responses during inflammation, autoimmunity, Adv Immunol 2007;96:179–204.
and cancer. Ann N Y Acad Sci 2010;1183:38–57. 402. Smith KG, Clatworthy MR. FcgammaRIIB in autoimmunity and
374. Mauri C, Bosma A. Immune regulatory function of B cells. Annu Rev infection: evolutionary and therapeutic implications. Nat Rev
Immunol 2012;30:221–41. Immunol 2010;10(5):328–43.
375. Yoshizaki A, et al. Regulatory B cells control T-cell autoimmu- 403. Rosales C, Brown EJ. Signal transduction by neutrophil immuno-
nity through IL-21-dependent cognate interactions. Nature globulin G Fc receptors. Dissociation of intracytoplasmic calcium
2012;491(7423):264–8. concentration rise from inositol 1,4,5-trisphosphate. J Bio Chem
376. Blair PA, et al. CD19(+)CD24(hi)CD38(hi) B cells exhibit regula- 1992;267(8):5265–71.
tory capacity in healthy individuals but are functionally impaired 404. Mayadas TN, Tsokos GC, Tsuboi N. Mechanisms of immune
in systemic lupus erythematosus patients. Immunity 2010;32(1): complex-mediated neutrophil recruitment and tissue injury.
129–40. Circulation 2009;120(20):2012–24.
377. Iwata Y, et al. Characterization of a rare IL-10-competent B-cell 405. Kobayashi SD, et al. Global changes in gene expression by human
subset in humans that parallels mouse regulatory B10 cells. Blood polymorphonuclear leukocytes during receptor-mediated phagocy-
2011;117(2):530–41. tosis: cell fate is regulated at the level of gene expression. Proc Natl
378. Ding Q, et al. Regulatory B cells are identified by expression of TIM-1 Acad Sci USA 2002;99(10):6901–6.
and can be induced through TIM-1 ligation to promote tolerance in 406. Zhou MJ, Brown EJ. CR3 (Mac-1, alpha M beta 2, CD11b/CD18) and
mice. J Clin Invest 2011;121(9):3645–56. Fc gamma RIII cooperate in generation of a neutrophil respiratory
379. van de Veen W, et al. IgG4 production is confined to human IL- burst: requirement for Fc gamma RIII and tyrosine phosphorylation.
10-producing regulatory B cells that suppress antigen-specific J Cell Biol 1994;125(6):1407–16.
immune responses. J Allergy Clin Immunol 2013;131(4):1204–12. 407. Coxon A, et al. Fc gamma RIII mediates neutrophil recruitment to
380. Sun J, et al. Transcriptomics identify CD9 as a marker of murine IL- immune complexes. A mechanism for neutrophil accumulation in
10-competent regulatory B cells. Cell Rep 2015;13(6):1110–7. immune-mediated inflammation. Immunity 2001;14(6):693–704.
2 • The Immunology of Transplantation 35

408. Sur Chowdhury C, et al. Enhanced neutrophil extracellular trap gen- 433. Akdis M, et al. Interleukins, from 1 to 37, and interferon-gamma:
eration in rheumatoid arthritis: analysis of underlying signal trans- receptors, functions, and roles in diseases. J Allergy Clin Immunol
duction pathways and potential diagnostic utility. Arth Res Ther 2011;127(3):701–21. e1-70.
2014;16(3):R122. 434. Deaglio S, et al. Adenosine generation catalyzed by CD39 and CD73
409. Kessenbrock K, et al. Netting neutrophils in autoimmune small- expressed on regulatory T cells mediates immune suppression. J Exp
vessel vasculitis. Nat Med 2009;15(6):623–5. Med 2007;204(6):1257–65.
410. Sadik CD, et al. Neutrophils orchestrate their own recruitment in 435. Gondek DC, et al. Cutting edge: contact-mediated suppression by
murine arthritis through C5aR and FcgammaR signaling. Proc Natl CD4+CD25+ regulatory cells involves a granzyme B-dependent, per-
Acad Sci USA 2012;109(46):E3177–85. forin-independent mechanism. J Immunol 2005;174(4):1783–6.
411. Clatworthy MR, et al. Immune complexes stimulate CCR7-dependent 436. Zhou X, et al. Instability of the transcription factor Foxp3 leads to
dendritic cell migration to lymph nodes. Nat Med 2014;20(12):1458– the generation of pathogenic memory T cells in vivo. Nat Immunol
63. 2009;10(9):1000–7.
412. Guilliams M, et al. The function of Fcγ receptors in dendritic cells and 437. Tivol EA, et al. Loss of CTLA-4 leads to massive lymphoproliferation
macrophages. Nature reviews. Immunology 2014;14(2):94–108. and fatal multiorgan tissue destruction, revealing a critical negative
413. Boruchov AM, et al. Activating and inhibitory IgG Fc recep- regulatory role of CTLA-4. Immunity 1995;3:541–7.
tors on human DCs mediate opposing functions. J Clin Invest 438. Grohmann U, et al. CTLA-4-Ig regulates tryptophan catabolism
2005;115(10):2914–23. in vivo. Nat Immunol 2002;3(11):1097–101.
414. Vogelpoel LT, et al. Fc gamma receptor-TLR cross-talk elicits pro- 439. Schliesser U, Streitz M, Sawitzki B. Tregs: application for solid-organ
inflammatory cytokine production by human M2 macrophages. Nat transplantation. Curr Opin Organ Transplant 2012;17(1):34–41.
Commun 2014;5:5444. 440. Lechler RI, Garden OA, Turka LA. The complementary roles of dele-
415. Hirohashi T, et al. A novel pathway of chronic allograft rejec- tion and regulation in transplantation tolerance. Nat Rev Immunol
tion mediated by NK cells and alloantibody. Am J Transplant 2003;3(2):147–58.
2012;12(2):313–21. 441. Jiang S, Lechler RI. Regulatory T cells in the control of transplantation
416. Hidalgo LG, et al. Interpreting NK cell transcripts versus T cell transcripts tolerance and autoimmunity. Am J Transplant 2003;3(5):516–24.
in renal transplant biopsies. Am J Transplant 2012;12(5):1180–91. 442. Bushell A, et al. The generation of CD25+ CD4+ regulatory T cells
417. Venner JM, et al. The molecular landscape of antibody-mediated that prevent allograft rejection does not compromise immunity to a
kidney transplant rejection: evidence for NK involvement through viral pathogen. J Immunol 2005;174(6):3290–7.
CD16a Fc receptors. Am J Transplant 2015;15(5):1336–48. 443. Nadig SN, et al. In vivo prevention of transplant arteriosclerosis by
418. Arnold ML, et al. Functional Fc gamma receptor gene polymor- ex vivo-expanded human regulatory T cells. Nat Med 2010;16(7):
phisms and donor-specific antibody-triggered microcirculation 809–13.
inflammation. Am J Transplant 2018;1–13. 444. Edinger M, Hoffmann P. Regulatory T cells in stem cell transplan-
419. Pawlik A, et al. The correlation between FcgammaRIIA poly- tation: strategies and first clinical experiences. Curr Opin Immunol
morphism and renal allograft survival. Transplant Proc 2011;23(5):679–84.
2002;34(8):3138–9. 445. Hoffmann P, Eder R, Edinger M. Polyclonal expansion of human
420. Ozkayin N, Mir S, Afig B. The role of Fcgamma receptor gene poly- CD4(+)CD25(+) regulatory T cells. Methods Mol Biol 2011;677:5–30.
morphism in pediatric renal transplant rejections. Transplant Proc 446. Tang Q, Bluestone JA, Kang SM. CD4(+)Foxp3(+) regulatory T cell
2008;40(10):3367–74. therapy in transplantation. J Mol Cell Biol 2012;4(1):11–21.
421. Bruhns P, et al. Specificity and affinity of human Fcgamma recep- 447. Newell KA, et al. Identification of a B cell signature associ-
tors and their polymorphic variants for human IgG subclasses. Blood ated with renal transplant tolerance in humans. J Clin Invest
2009;113(16):3716–25. 2010;120(6):1836–47.
422. Gillis C, et al. Contribution of human FcgammaRs to disease with 448. Turnbaugh PJ, et al. A core gut microbiome in obese and lean twins.
evidence from human polymorphisms and transgenic animal Nature 2009;457(7228):480–4.
studies. Front Immunol 2014;5:254. 449. Qin N, et al. Alterations of the human gut microbiome in liver cir-
423. Valenzuela NM, et al. Monocyte recruitment by HLA IgG-activated rhosis. Nature 2014;513(7516):59–64.
endothelium: the relationship between IgG subclass and Fcgam- 450. Le Chatelier E, et al. Richness of human gut microbiome correlates
maRIIa polymorphisms. Am J Transplant 2015;15(6):1502–18. with metabolic markers. Nature 2013;500(7464):541–6.
424. Arnold ML, et al. Association of a coding polymorphism in Fc gamma 451. Ivanov II , et al. Induction of intestinal Th17 cells by segmented fila-
receptor 2A and graft survival in re-transplant candidates. Hum mentous bacteria. Cell 2009;139(3):485–98.
Immunol 2015;76(10):759–64. 452. Furusawa Y, et al. Commensal microbe-derived butyrate
425. Callaghan CJ, et al. Regulation of allograft survival by inhibitory induces the differentiation of colonic regulatory T cells. Nature
FcgammaRIIb signaling. J Immunol 2012;189(12):5694–702. 2013;504(7480):446–50.
426. Sharp PE, et al. FcgammaRIIb on myeloid cells and intrinsic renal 453. Vindigni SM, Surawicz CM. The gut microbiome: a clinically sig-
cells rather than B cells protects from nephrotoxic nephritis. J Immu- nificant player in transplantation? Expert Rev Clin Immunol
nol 2013;190(1):340–8. 2015;11(7):781–3.
427. Floto RA, et al. Loss of function of a lupus-associated Fcgam- 454. Bromberg JS, et al. Microbiota-implications for immunity and trans-
maRIIb polymorphism through exclusion from lipid rafts. Nat Med plantation. Nat Rev Nephrol 2015;11(6):342–53.
2005;11(10):1056–8. 455. Andrade-Oliveira V, et al. Gut bacteria products prevent AKI
428. Kono H, et al. FcgammaRIIB Ile232Thr transmembrane poly- induced by ischemia-reperfusion. J Am Soc Nephrol 2015;26(8):
morphism associated with human systemic lupus erythematosus 1877–88.
decreases affinity to lipid rafts and attenuates inhibitory effects on B 456. Lei YM, et al. The composition of the microbiota modulates allograft
cell receptor signaling. Hum Mol Genet 2005;14(19):2881–92. rejection. J Clin Invest 2016;126(7):2736–44.
429. Xu L, et al. Impairment on the lateral mobility induced by structural 457. Modena BD, et al. Changes in urinary microbiome populations
changes underlies the functional deficiency of the lupus-associated correlate in kidney transplants with interstitial fibrosis and tubular
polymorphism Fc γ RIIB-T232. J Exp Med 2016;213(12):2707. atrophy documented in early surveillance biopsies. Am J Transplant
430. Clatworthy MR, et al. Defunctioning polymorphism in the immu- 2017;17(3):712–23.
noglobulin G inhibitory receptor (FcγRIIB-T/T232) does not 458. Gorer PA. The antigenic basis of tumour transplantation. J Pathol
impact on kidney transplant or recipient survival. Transplantation Bacteriol 1938;47:231–52.
2014;98(3):285–91. 459. Gibson JM, Medawar PB. The fate of skin homografts in man. J Anat
431. Devaraj S, Du Clos TW, Jialal I. Binding and internalization of 1943;77:299–310.
C-reactive protein by Fcgamma receptors on human aortic endothe- 460. Medawar PB. Behaviour and fate of skin autografts and skin homo-
lial cells mediates biological effects. Arterioscler Thromb Vasc Biol grafts in rabbits. J Anat 1944;78:176–99.
2005;25(7):1359–63. 461. Medawar PB. A second study of the behaviour and fate of skin homo-
432. Pan LF, Kreisle Ra, Shi YD. Detection of Fcgamma receptors on grafts in rabbits. J Anat 1945;79:157–76.
human endothelial cells stimulated with cytokines tumour necrosis 462. Medawar PB. Immunity to homologous grafted skin. III. The fate of
factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma). Clin skin homografts transplanted to the brain, to subcutaneous tissue and
Exper Immunol 1998;112(3):533–8. to the anterior chamber of the eye. Brit J Exp Path 1948;29:58–69.
3 Chronic Kidney Failure:
Renal Replacement
Therapy
ANDREW DAVENPORT

CHAPTER OUTLINE Introduction Dialysis Transport


Definition and Timing Referral Automated Peritoneal Dialysis
Prevalence and Incidence Continuous Ambulatory Peritoneal Dialysis
Etiology Practical Considerations
Treatment of CKD5 Indications for and Advantages of Peritoneal
Dialysis Dialysis
General Aspects Posttransplantation
Hypertension and Fluid and Electrolyte Complications
Balance Loss of Ultrafiltration
Hematopoiesis and Immunity Peritonitis
Calcium, Phosphate, and the Skeleton Exit Site and Tunnel Infection
Nutrition and Metabolism Anatomic Complications
Hemostasis Encapsulating Peritoneal Sclerosis
Skin Metabolic Complications
Neurologic and Musculoskeletal Choice and Planning for the
Manifestations Individual Patient
Endocrine Abnormalities Dialysis Posttransplant
Psychological Problems Hemodialysis
Initiation of Dialysis Continuous Ambulatory Peritoneal Dialysis
Hemodialysis Return to Dialysis After Transplant Failure
Complications RRT Modality and Survival
Peritoneal Dialysis Quality of Life
Dialysis Adequacy

Introduction and reducing cardiovascular risk factors, because many


more patients will die of cardiovascular disease compared
Renal replacement therapy (RRT) is a general term encom- with those who progress to dialysis (CKD5d).4 Uncontrolled
passing a range of different treatment modalities for patients hypertension is the major risk factor for progression fol-
with what was formally termed acute renal failure and end- lowed by proteinuria. Angiotensin-converting enzyme
stage kidney disease, which are now called acute kidney inhibitors and angiotensin receptor blockers are the pre-
injury stage 3 (AKI-3)1 and chronic kidney disease stage ferred antihypertensives, aiming for blood pressure targets
5 dialysis (CKD5d),2 respectively (Table 3.1A and B). RRT of 130 to 140/80 to 90 mmHg, depending on the age of the
includes various forms of dialysis (hemodialysis, hemodiafil- patient. In a minority of cases specific management strate-
tration, and peritoneal dialysis), hemofiltration, and renal gies may be appropriate to halt progression, such as immu-
transplantation. Dialysis has rapidly expanded from a treat- nosuppression for patients with renal vasculitis or lupus
ment restricted to AKI in teaching hospitals in the 1960s nephritis. Thereafter management is directed to control the
to what is now a routine treatment for around 3 million complications of progressive kidney disease, hypervolemia,
patients with CKD worldwide. However, all types of RRT are anemia, acidosis, and renal bone disease.5 In progressive
incomplete solutions for CKD, with a 5-year life expectancy CKD patient education is vital, so that patients can make
for a dialysis patient in the United Kingdom (UK) of around an informed decision about whether to have RRT or opt
60%, somewhere between that of patients with ovarian and for a conservative nondialysis approach, accepting that
bowel cancer (www.renalreg.com).3 they will die of azotemia. For those patients opting for RRT,
The management of patients with CKD centers on trying it is important to plan ahead, asking about potential live
to slow down the progression of underlying kidney disease organ donors, creating vascular access for those choosing
36
3 • Chronic Kidney Failure: Renal Replacement Therapy 37

TABLE 3.1A Staging for Chronic Kidney Diseasea quality for the individual patient at the lowest possible cost.
Quality standards and clinical practice guidelines have
CKD Stage eGFR (mL/min/1.73 m2) been developed by both national and international organi-
Stage 1 >90 zations (e.g., British Transplantation Society, Renal Associ-
Stage 2 60–90 ation, European Renal Association – European Dialysis and
Stage 3a 45–60 Transplant Association, Kidney Disease Improving Global
Stage 3b 30–44
Stage 4 15–29
Outcomes).
Stage 5 <15 mL/min
Stage 5d Dialysis
aStaging for chronic kidney disease (CKD), based on an estimate of glomerular
Definition and Timing Referral
filtration rate (eGFR) after the modification of diet in renal disease (MDRD)
4 value equation. The suffix (p) can be used to denote the presence of The estimate of glomerular filtration rate (eGFR) system
proteinuria as defined by a spot urinary albumin:creatinine ratio of ≥30 (Table 3.1A) was introduced into the UK to alert nonre-
mg/mmol, which is approximately equivalent to a protein:creatinine ratio nal specialists that patients with what appeared to be high
of ≥50 mg/mmol (≥0.5 g/24 h). normal or mildly elevated serum creatinine had CKD, and
therefore are more at risk from nephrotoxic drugs. Unfortu-
TABLE 3.1B Staging for Acute Kidney Injurya nately, serum creatinine is not linearly associated with glo-
merular filtration rate and is affected by diet, exercise, and
Stage Serum Creatinine Criteria Urine Criteria
drugs (see Table 3.2). In the UK patients with CKD stage 3 are
1 ↑ SCreat ≥0.3 mg/dL or 27 μmol/L <0.5 mL/kg/h for >6 h routinely managed by primary care physicians, unless they
above baseline within 48 h or ↑ have rapidly progressive kidney disease, or proteinuria or
SCreat ≥1.5–1.9× above baseline
within 7 days hematuria, whereas those in stage 4 and 5 should be under
2 ↑ SCreat 2.0–2.9× above baseline <0.5 mL/kg/h for >2 h the care of specialist nephrologists. However, there is still a
within 7 days relatively high number of avoidable late referrals of patients
3 SCreat 3× above baseline or <0.3 mL/kg/h for 24 h with CKD5.6 In some cases this situation is unavoidable—
SCreat ≥4.0 mg/dL (350 μmol/L) or anuria for 12 h
or RRT within 7 days
the patients may have had a truly silent illness or an acute
presentation of an irreversible renal injury (e.g., myeloma,
aThe baseline serum creatinine (SCreat) measurement should be the admission antiglomerular basement membrane disease, or renal vas-
serum creatinine after resuscitation, or a recent clinic value. The change in culitis). The consequences for patients of late presentation
serum creatinine should occur within 7 days. All patients who are treated are many, ranging from increased risk of infection because
by renal replacement therapy are staged as stage 3.
of the use of temporary central venous access catheters to
the psychological effect of suddenly requiring dialysis, with
TABLE 3.2 Examples of Nonrenal Factors That Affect major changes in lifestyle (dietary and fluid restriction, loss
Serum Creatinine Measurements, Without Changing of employment). Not surprisingly, these unplanned starters
Renal Function have a worse prognosis,7 incur greater healthcare costs,8
Effect on Serum
and are less likely to be activated on the renal transplant
Factor Creatinine waiting list.
General factors Age Decreased
Female sex Decreased
Ethnicity Black Increased Prevalence and Incidence
compared with Asian Decreased
Caucasoids The point prevalence of CKD is unknown because most
Body habitus Muscular Increased patients are asymptomatic and unaware of its presence.
Obese Decreased
Chronic illness Cirrhosis Reduced Referral patterns increased in the UK after the introduction
Cancer Reduced of the eGFR reporting system, because of the alerts sent back
Heart failure Reduced from pathology laboratories to primary care physicians.9
Endocrine Hypothyroidism Increased Similarly, the prevalence of CKD increases as populations
diseases
Diet Vegetarian Reduced
increase in body habitus and prevalence of diabetes.10 In
Meat stews Increased the latest UK renal registry report the prevalence of patients
Drugs Antibiotics Trimethoprim receiving RRT was 913 per million population (pmp), but
Gastroprotection Cimetidine varies within the UK, being higher in inner-city ethnic
Diuretics Amiloride populations and lower in predominantly affluent caucasoid
Spironolactone
suburbs, with the prevalence varying between 560 and
1680 pmp (http://www.renalreg.org/).
The differences in incidence and prevalence of CKD
hemodialysis, and considering the timing of inserting a requiring RRT (Table 3.3) depend both on patient factors,
peritoneal dialysis catheter. such as the incidence of diabetes and hypertension in the
RRT programs continue to expand (Table 3.2), and, population studied, and healthcare spending.11 In 2014 the
although <0.04% of the UK population has CKD requir- highest incidence of CKD5d reported came from Taiwan,
ing RRT, this consumes 2% to 3% of the overall UK health Jalisco (Mexico), United States, Thailand, and Singapore,
budget. The challenge for nephrologists and transplant sur- whereas Iceland, Iran, Russia, and Bangladesh reported
geons is to provide the most appropriate RRT of the highest <80 per million (http://www.usrds.org). However, it must
38 Kidney Transplantation: Principles and Practice

TABLE 3.3 Incidence and Prevalence of Chronic Kidney Disease Treated by Dialysis, and Transplantation Prevalence of
Functioning Grafts and Transplant Rate (Rates Expressed per Million Population)
Dialysis Dialysis Dialysis Dialysis Transplant Transplant
Year 2010 2012 2014 2014 2014 2014
Country Incidence Incidence Incidence Prevalence Prevalence Rate
Argentina 152 156 155 856 191 29.8
Australia 106 114 111 944 431 40.4
Austria 140 143 122 1062 549 46.8
Bangladesh 23 45 49 113 0.6
Belgium 200 191 175 1242 515 36.0
Brazil 147 172 180 773 224 27.8
Canada 178 182 193 1291 534 41.5
Chile 156 170 157 1301 207 16.7
Denmark 198 191 906 449 43.9
France 153 155 161 1203 540 49.1
Iceland 107 62 58 675 452 24.4
Iran 74 74 78 617 294 29.7
Israel 186 183 203 1169 405 31
Japan 291 285 285 2505 65 12.6
Mexico Jalisco 404 467 421 1568 590 60.3
Netherlands 118 121 115 967 583 59.2
New Zealand 118 189 122 962 364 31.7
Philippines 104 127 161 289 4 4.0
Qatar 133 99 116 689 358 21.9
Russia 45 53 60 283 51 5.7
Saudi Arabia 100 100 104 771 258 21.0
Singapore 243 285 294 1891 33 15.0
Spain 121 120 132 1168 605 56.8
Taiwan 439 446 455 3219 126
Thailand 146 221 299 1203 106 8.5
Turkey 223 139 147 918 143 37.6
UK (England and 108 111 118 932 494 48.5
Wales)
USA 367 360 370 2076 630 56.2
Uruguay 153 150 151 1122 366 39.9

be recognized that there is a difference between the inci- replacement therapy for conditions such as Fabry’s dis-
dence of the disease and the incidence of patients starting ease. The incidence of diseases that cause CKD change
RRT, because more affluent countries will start more elderly with age, such that childhood CKD may be associated
patients with additional comorbidities,12 and the incidence with congenital abnormalities, including urethral valves,
of CKD increases exponentially with age, whereas less afflu- prune-belly syndrome, vesico-ureteric reflux, whereas
ent countries tend to restrict RRT to younger patients with patients older than age 65 are more likely to have reno-
fewer comorbidities. Similarly, transplantation rates also vascular disease, hypertensive nephropathy, myeloma,
differ between countries, and although this is dependent on and prostatic obstruction.
the provision and access to dialysis, it can also be affected The prevalence of inherited diseases, such as polycys-
by cultural and religious practices that limit cadaveric tic kidney disease, appears to be similar in different ethnic
transplantation. populations; however, diabetic nephropathy tends to fol-
The numbers of patients on RRT worldwide have yet to low the rate of diabetes in the underlying population. In
reach a steady state, particularly in developing economies. addition, some forms of interstitial nephritis have distinc-
This will only occur when the number of new patients tive geographic patterns or predilection for particular eth-
accepted into programs is balanced by the number of nic groups, such as Balkan nephropathy, Chinese herbal
deaths. nephropathy, associated with ingestion of Aristolochic
herbs, South Asian interstitial nephropathy, Meso Ameri-
can nephropathy, and Sri Lankan nephropathy.
Etiology Screening programs, coupled with prompt treatment
and investigation of urosepsis, have reduced the number of
Because RRT is an expensive supportive treatment, the children with vesico-ureteric reflux progressing to CKD5d.
key goal would be to prevent or limit the progression of However, these causes comprise only a minority of cases,
CKD. Unfortunately, only a minority of conditions can with the vast majority of patients developing progressive
be halted, and even then, only if treated early, such as CKD because of small-vessel disease in association with
withdrawal of nephrotoxic analgesics and lithium and hypertension and diabetes. Such health screening programs
appropriate treatment of infections, such as tuberculosis, have centered on the measurement of eGFR to establish
inflammatory conditions, including sarcoid and auto- an earlier diagnosis of CKD, and then active treatment of
immune diseases, most commonly vasculitis and sys- cardiovascular risk factors, because hypertension and pro-
temic lupus erythematosus, and more recently, enzyme teinuria are the key risk factors for progression.5 Because
3 • Chronic Kidney Failure: Renal Replacement Therapy 39

angiotensin-converting enzyme inhibitors and angiotensin TABLE 3.4 Proportion (%) of Chronic Kidney Disease
receptor blockers are potent antihypertensive agents, and Patients Treated by Different Dialysis Modalities in 2014
also appear to have an additional effect in terms of reduc-
ing proteinuria, these agents are preferentially prescribed. Center Home Peritoneal
Modality Hemodialysis Hemodialysis Dialysis
As patients develop progressive CKD, appetite declines, so
patients tend to self-restrict protein, and protein-restricted Argentina 94.2 0 5.8
diets do not appear to have any additional benefit, provided Australia 70.2 9.4 20.4
Austria 90.9 0.1 9.0
that blood pressure is adequately controlled.13 In addition Bangladesh 93.6 0 6.4
to controlling blood pressure, prescription of sodium bicar- Belgium 90.9 1.3 7.8
bonate, to correct metabolic acidosis has been shown to Brazil 92.7 0.1 7.2
reduce progression of CKD. Canada 75.1 4.8 19.1
Chile 94.2 0 5.8
For most patients, establishing the cause of CKD can be Denmark 78.7 0 21.3
determined by history, physical examination, simple urine France 92.6 0.6 6.8
dipstick testing coupled with specific biochemical and Iceland 78.0 21.9
immunologic investigations, and renal imaging. Patients Iran 94.1 0 5.9
may have a family history of adult polycystic kidney dis- Israel 89.0 0 11.0
Japan 96.9 0.2 2.9
ease, glomerulonephritis, reflux nephropathy, and hyper- Mexico (Jalisco) 52.3 0.8 46.9
tension. Physical examination may reveal femoral arterial Netherlands 82.7 3.8 13.5
bruits and signs of cholesterol embolization in renovascular New Zealand 51.1 18.3 30.6
disease, or skin and joint changes in vasculitis and auto- Philippines 95.2 0 4.8
Qatar 73.3 0 26.7
immune diseases. Patients with glomerular hematuria Russia 93.6 0 6.4
and proteinuria require renal biopsy because glomerulo- Saudi Arabia 88.3 11.6
nephritis or systemic disease may be amenable to specific Singapore 93.5 0.1 6.4
therapies. Spain 88.3 0.3 11.4
Transplant surgeons prefer to know the cause of CKD in Taiwan 69.6 30.4
Thailand 69.6 0 30.4
potential kidney transplant recipients, because some condi- Turkey 72.3 0.3 7.4
tions can recur posttransplantation, including some forms UK (England 82.1 4.4 13.5
of focal segmental glomerular sclerosis; hemolytic uremic and Wales)
syndrome and dense deposit disease, a form of membra- USA 88.6 1.8 9.6
Uruguay 91.0 0 9
noproliferative glomerulonephritis; and IgA nephropathy.
Similarly, other renal conditions may be inherited and need
to be screened in family members volunteering as living
donors. In addition, patients with urogenital malformations helpers, because home hemodialysis provides patients with
or recurrent urosepsis may require surgical reconstruction an option for more frequent or longer overnight dialysis ses-
or nephrectomy before transplantation to reduce the risk of sions. Although in-center hemodialysis may be the default
subsequent infections. option, ideally hemodialysis patients should dialyze close
to home, in local hospital satellite or free-standing self-care
centers, to minimize traveling time.
Treatment of CKD5 Apart from Mexico (Table 3.4), and some Southeast Asian
countries that have a peritoneal dialysis–first policy, hemo-
Ideally patients with CKD5 should be involved in the deci- dialysis remains the most common mode of RRT worldwide.
sion to choose a form of RRT, whether conservative man- The differences between countries are at times difficult to
agement or dialysis. In practice, economic and cultural understand and are a result of the balance between medical
pressures, the limit in kidney donor supply, and medical and financial resources, reimbursement, and patient local-
prejudices often dictate what treatment patients receive. ity. For example, in New Zealand and Canada many patients
This is particularly the case for patients who have had no live in rural environments distant from hospitals, and as
or minimal predialysis nephrologic care, who by default such home-based therapies, peritoneal dialysis, and home
typically start hemodialysis using a central venous access hemodialysis are more favored. On the other hand, coun-
catheter. tries such as Japan, the United States, and Germany, which
Because life expectancy and quality of life are typically offer greater financial reimbursement for hemodialysis, typ-
greater with transplantation, then transplantation should ically have more center-based hemodialysis programs.
be considered for all patients with an expected 5-year sur-
vival or greater. Preemptive transplantation is an option DIALYSIS
for CKD patients attending nephrologic care if a suitable
living donor is available; otherwise patients opting for General Aspects
dialysis should be allowed to choose between hemodialy- The aims of dialysis are to maintain homeostasis, in terms
sis and peritoneal dialysis, so that vascular access can be of electrolyte, acid–base, volume status, and removal of
created before initiation of hemodialysis; and for peritoneal the waste products of nitrogen metabolism that accumu-
dialysis, embedding a peritoneal dialysis catheter is also an late in CKD. Dialysis adequacy is typically measured by
option, but the longer the catheter remains buried the less urea clearance, although urea itself is only one of many
likely it is to work when released. Home dialysis should be azotemic toxins that accumulate in CKD5d patients as a
encouraged for patients with appropriate home circum- consequence of nitrogen turnover. The original National
stances, especially if patients have partners who can act as Cooperative Dialysis Study14 showed that a minimal
40 Kidney Transplantation: Principles and Practice

amount of urea clearance was required for patients to sus- advances in the management of patients with CKD5. Hemo-
tain well-being. To correct for different patient sizes, urea globin targets have been reduced recently (www.kdigo.org),
clearance was normalized and expressed as a dimension- because of the report of increased risk of stroke in patients
less formula, termed Kt/Vurea, where for hemodialysis K is with higher hematocrit, with most clinical guidelines now
the dialyzer urea clearance, and for peritoneal dialysis the advocating a target of 10.0 to 12.0 g/dL.17 Patients who
combination of urinary and peritoneal urea clearances,­ fail to respond to ESAs adequately are usually found to be
t the time, and Vurea the volume of urea distribution in the iron-deficient or have active bleeding or to have foci of infec-
body. For hemodialysis Kt/Vurea is expressed per dialysis tion and inflammation. Oral iron supplements are often
session, whereas for peritoneal dialysis it is expressed per inadequate and most patients require intravenous iron
week. Over time the target Kt/Vurea value for adequate preparations. Too rapid an increase in hematocrit can lead
dialysis has increased from 1.0 to 1.4 for thrice-weekly to uncontrolled hypertension and seizures; high hemato-
hemodialysis treatments,15 and a minimum weekly value crits are associated with increased risk of vascular access
of 1.7 for peritoneal dialysis.16 Whereas peritoneal dialysis thrombosis, particularly arteriovenous grafts, and occa-
is a continuous therapy, hemodialysis is typically an inter- sional hyperkalemia. Since the introduction of ESAs, blood
mittent treatment with three sessions of 4 hours’ duration, transfusion requirements have generally reduced, reducing
scheduled either Monday, Wednesday, Friday or Tuesday, third-party human leukocyte antigen (HLA) sensitization.
Thursday, Saturday. Platelet counts are normal in CKD, but their function is
However, it should be recognized that what is considered abnormal (see under Hemostasis).
to be adequate dialysis provides the equivalent of only 6 to Infections are the second major cause of death in CKD5
10 mL/min of glomerular filtration rate so that there are patients. Cell-mediated immunity is depressed in chronic
many consequences of CKD for which additional measures renal failure, which explains the failure to clear hepatitis
are needed. B infection and the higher risk of reactivating tuberculo-
sis and varicella-zoster, and reduced response to hepatitis
Hypertension and Fluid and Electrolyte Balance B and other vaccinations. Despite reduced responsiveness,
Hypertension is common in CKD patients both before start- most countries advocate vaccination against hepatitis B,
ing and when being treated by dialysis, and it is usually pneumococcus, varicella-zoster, and influenza.
associated with expansion of the extracellular fluid volume
caused by positive sodium and water balance. Hemodialysis Calcium, Phosphate, and the Skeleton
patients find it difficult to stick to target dry weights and fre- Changes in calcium and phosphate homeostasis occur early
quently gain 2 kg or more between hemodialysis sessions. in the course of progressive CKD as a result of reduced renal
Dietary sodium restriction is essential to reduce thirst, but phosphate clearance, despite an increase in phosphotonins,
most patients also require ultrafiltration with dialysis, unless such as fibroblast factor 23 and reduced production of the
they have good residual renal function. Excessive water active form of vitamin D3, 1,25-dihydroxycholecalciferol,
intake can lead to dilutional hyponatremia. Longer-acting which depends on 1α-hydroxylation in the kidney. Phosphate
antihypertensive drugs are preferred because short-acting retention and relative deficiency of 1,25-dihydroxychole-
vasodilators may exacerbate intradialytic hypotension calciferol cause an increase in the secretion of parathyroid
when the rate of ultrafiltration during dialysis exceeds that of hormone (PTH) by the parathyroid glands, which eventu-
plasma refilling from the extracellular space. Choice of anti- ally become hyperplastic and then autonomous. Although
hypertensives is important because water soluble β-blockers the increased concentration of PTH enhances phosphate
and angiotensin converting enzyme inhibitors are cleared excretion, it increases bone turnover by disturbing the bal-
by hemodialysis. Although there are target blood pressures ance between osteoblast and osteoclast activity, so disorga-
for the general population, there are no agreed targets for nizing bone structure, with Looser’s zones, microfractures,
hemodialysis patients, because blood pressure typically falls and increased risk of tendon rupture. As such, most CKDd
with hemodialysis, and blood pressure measurements taken patients require supplemental 1-hydroxycholecalciferol to
immediately before dialysis do not correspond to 24-hour control hyperparathyroidism. Hyperphosphatemia needs to
ambulatory blood pressure or home blood pressure mea- be controlled by taking agents designed to bind phosphate
surements during the interdialytic interval. in the gastrointestinal tract.18 These phosphate binders
Dialysis only removes around 60 mmol of potassium per may contain elemental calcium alone or combined with
day so that dietary restriction is essential. Hyperkalemia magnesium, the rare earth lanthanum, inert plastic-based
has effects on cardiac muscle (arrhythmias) and on skeletal ion-exchange resins, or iron and aluminum. Although
muscle (weakness and ultimately paralysis). More hemo- aluminum is a potent phosphate binder, aluminum accu-
dialysis patients die toward the end of the 2-day interval mulation can lead to a microcytic anemia, fracturing renal
between dialysis sessions, just before the first dialysis ses- osteodystrophy, and in extreme cases encephalopathy and
sion of the week, at a time when they are most likely to be death. Patients with CKD are prone to soft-tissue calcification
most volume-overloaded and hyperkalemic. resulting from a reduction in GLA matrix proteins, typically
leading to medial arterial calcification, which can lead to
Hematopoiesis and Immunity soft-tissue calcinosis with skin and fat ischemia and necrosis
Erythropoiesis is reduced in patients with CKD, who also in severe cases. This typically occurs in the setting of inflam-
have higher iron requirements than the general popula- mation with an increased serum calcium phosphate prod-
tion because of increased hepcidin, which reduces gas- uct, in patients with vitamin K deficiency (often secondary
trointestinal iron absorption and iron release from the to warfarin therapy) or 25-hydroxy-vitamin D3 deficiency,
reticuloendothelial system. The introduction of erythropoi- which reduce hepatic GLA matrix protein synthesis, and
etin-stimulating agents (ESAs) has been one of the major either oversuppressed or very overactive parathyroid glands.
3 • Chronic Kidney Failure: Renal Replacement Therapy 41

Nutrition and Metabolism deposition, is a specific form of uremic mononeuropathy,


Malnutrition is common in dialysis patients and is caused typically affecting the arm with an arteriovenous fistula or
by inappropriate dietary restrictions, anorexia because of graft. This complication is declining with the introduction
reduction in orexigenic hormones acyl-ghrelin with corre- of ultrapure-quality dialysates and high-flux hemodialyz-
sponding increased desacyl ghrelin, leptin, obestatin, acido- ers. Gout and pseudogout resulting from pyrophosphate
sis, and insulin resistance, and is aggravated by intercurrent crystals cause a painful crystal arthropathy. Aluminum
infections, and reduced sense of taste and smell. Appetite toxicity resulting from contaminated dialysate or occa-
may be further suppressed in peritoneal dialysis patients sionally after consumption of large numbers of aluminum-
because of acid reflux and constipation, and glucose absorp- based medications can rarely cause a pseudoparkinsonian
tion from the dialysate. Patients may have simple malnutri- syndrome, coma, and even death.
tion, which responds to enteral feeding, or in more severe
cases protein energy wasting, with loss of muscle mass and Endocrine Abnormalities
hypoalbuminemia. Because there are additional protein There are diverse abnormalities in hormone production,
losses in the dialysate, dialysis patients are recommended to control, protein binding, catabolism, and tissue effects in
eat 1.2 g protein/kg/day.19 Hypercholesterolemia is more dialysis patients. The most obvious examples are those of
common with peritoneal dialysis patients, resulting from the vitamin D–PTH axis and erythropoietin caused by loss of
absorption of glucose from the dialysate and hypertriglyc- renal function, but thyroid hormone, growth hormone, and
eridemia from heparin administration with hemodialysis. both prolactin and sex hormones are also affected. Women
usually are infertile, and men often have erectile dysfunc-
Hemostasis tion. Sildenafil and other phosphodiesterase-5 inhibitors
Untreated CKD5 patients have a bleeding diathesis, which have proved effective but should be used with caution in
is, in part, a consequence of abnormal platelet function, patients with heart disease and avoided in patients pre-
with prolonged bleeding times. However, once patients are scribed nitrates for angina. However, many of these “appar-
adequately dialyzed, they are more at risk of thrombosis. ent” endocrine abnormalities are reversed by more frequent
and longer hemodialysis sessions, suggesting that they
Skin are consequent upon inadequate delivery of RRT. Indeed,
Itching is a common symptom in dialysis patients and is greater RRT dosing restores female fertility, and with daily
aggravated by dry skin, secondary to reduced apocrine hemodialysis treatments pregnancies can be sustained and
function, heat, and stress. In some cases itching is caused intrauterine growth retardation minimized.
by deposition of calcium phosphate crystals secondary to a
high calcium (Ca) inorganic phosphate (Pi) product (>6.25 Psychological Problems
mmol2/L2, >70 mg2/dL2), or similarly high magnesium, or The psychological problems of dialysis patients, usually
caused by abnormal demyelinated nerve fiber ending sensi- anxiety and depression, are the predictable and understand-
tivity. Treatment with moisturizing skin lotions with menthol able consequences of loss of health, control, and pleasure.21
sometimes helps, but in cases of neuropathy patients often Depression is more common in unplanned dialysis starters
require trials of oral gabapentin, ondansetron, or naltrexone compared with those who have been counseled in predialy-
to downregulate pain receptor sensitivity and local treat- sis nephrologic clinics. For most patients dialysis dictates
ments with capsaicin cream or ultraviolet B phototherapy. their lifestyle, and not surprisingly, depressed patients have
The widespread use of gadolinium chelates as contrast increased mortality.22
agents for magnetic resonance imaging (MRI) scans has led
to reports of gadolinium deposition in the skin and other tis- Initiation of Dialysis
sues, causing nephrogenic systemic fibrosis (NSF). There is When should dialysis be started in a patient with CKD?
no current treatment, and therefore gadolinium-enhanced Symptomatic patients, those who are losing weight, and
MRI scans should be limited in CKDd patients, and mac- patients with volume overload, uncontrolled acidosis, or
rocyclic gadolinium chelates preferred because there are hyperkalemia should initiate dialysis (Table 3.5). However,
fewer reports of NSF compared with linear chelates.20 recent studies have shown that there is no advantage to
starting the apparently stable asymptomatic patient who is
Neurologic and Musculoskeletal Manifestations maintaining weight, not fluid-overloaded, with controlled
Uremic encephalopathy usually manifests as subtle cogni- biochemistry (not acidotic or hyperkalemic) on dialysis until
tive impairment and is an indication for increasing dialysis the glomerular filtration rate falls to around 5 mL/min.23
dose. Coma and seizures are rare, except in noncompli-
ant patients who skip treatments. An asymmetric distal
polyneuropathy is common in patients with CKD5. The TABLE 3.5 Indications to Initiate Dialysis in Patients
With Chronic Kidney Disease
symptoms are those of dysesthesia—a prickling or burning
sensation and, rarely, foot drop. Restless legs, especially at Biochemical Refractory hyperkalemia >6.5 mmol/L
night, are a nuisance and may respond to low-dose dopa- indications
mine agonists, including pramipexole and cabergoline at Serum urea >50 mmol/L not due to hypovolemia
Refractory metabolic acidosis pH ≤7.1
night. An autonomic neuropathy is variable in its effects— Clinical End organ damage: pericarditis, encephalopathy,
manifesting mostly as sexual dysfunction and sluggish car- indications neuropathy, myopathy, uremic bleeding,
diovascular reflexes during hemodialysis, with increased weight loss
risk of intradialytic hypotension. Median nerve compression Refractory volume overload
in the carpal tunnel, caused by β2-microglobulin amyloid
42 Kidney Transplantation: Principles and Practice

HEMODIALYSIS preferably three, shifts per day, 6 days per week, with one
nurse or dialysis technician being responsible for 4 to 6 dial-
During hemodialysis, water-soluble solutes that are small ysis patients at a time. An important consideration is the
enough to pass through the pores of a semipermeable dia- production of large volumes of water in the generation of
lyzer membrane diffuse and move down a concentration dialysate. Tap water is purified by filtration to remove bac-
gradient. In addition, the application of a pressure gradient teria, softened to remove calcium, carbon filtered to remove
across the semipermeable membrane drives ultrafiltration small organic compounds such as chloramines, and reverse
or convection because of a bulk water movement across osmosis treated to remove other contaminating substances,
the membrane; this leads not only to water loss, but also including metals and nitrites. The water system should run
to a convective loss of those water-soluble solutes that are 24 hours per day to prevent stagnation and biofilm depo-
able to pass through the membrane pores. Whereas dif- sition, with ideally multiple passes through the reverse
fusion is effective for clearing small solutes, this convec- osmosis system to improve final water quality to achieve
tive clearance increases middle-sized solute clearances. In ultrapure water, which is essential if high-flux and hemo-
routine practice, this requires a dialysis membrane with diafiltration treatments are used. Water ring-mains require
a surface area of 1.0 to 2.3 m2 (now conveniently pack- regular cleaning and disinfection.
aged as single-use sterilized hollow-fiber dialyzers), a blood Patients who are hepatitis B antigen–positive should be
flow of 250 to 400 mL/min, and a countercurrent flow of dialyzed in isolation and on dedicated dialysis machines.
dialysate, generated by a proportioning machine, of 500 Hepatitis B vaccinations are recommended and are most
to 800 mL/min. The dialysate can be made in a batch and effective if administered before CKD5 develops. Universal
pumped to the dialysis machine, or made by the dialysis precautions are considered sufficient to prevent spread
machine by adding concentrated electrolytes and sodium of human immunodeficiency virus and hepatitis C, but
bicarbonate to the dialysis water, warming it, checking many centers cohort these patients to prevent nosocomial
conductivity, and pumping it through the dialyzer and transmission.
then to waste. The dialysis machine pumps the blood from
the patient through the dialyzer and returns it via a venous Complications
air detector, which is alarmed to prevent air embolism. Hypotension occurs in up to 30% of dialysis sessions and
Anticoagulation can be achieved by a single bolus of low- most commonly is secondary to intravascular hypovole-
molecular-weight heparin or by a bolus of unfractionated mia. It is more common when large volumes of fluid have
heparin followed by a continuous infusion. Modern dialy- to be removed during a short hemodialysis session after
sis machines can be programmed to remove fluid gained excessive gains between dialyses. Eating meals during
in the interdialytic interval, and by altering the dialysate dialysis increases the risk of hypotension. Hypotension usu-
sodium concentration, temperature, and the rate of ultra- ally responds to laying the patient flat or head-down, stop-
filtration, reduce the risk of intradialytic hypotension. ping ultrafiltration, or giving a volume of normal saline
Dialysis prescription can be altered by choice of dialyzer or hypertonic glucose. Muscle cramps (approximately
membrane, surface area, blood and dialysate flow rates, 5%–20% of dialyses) often accompany hypotension, an
dialysate composition and temperature, and frequency excessively low target weight, or the use of a low-sodium
and duration of the dialysis sessions. dialysate. Occasionally cramps may be caused by carnitine
Hemodialysis treatments traditionally used low-flux dia- deficiency. Correction of these or the administration of a
lyzers, which effectively cleared small-sized solutes, such as hypertonic solution (saline or glucose) usually is effective
urea, but were not effective in clearing middle-sized solutes treatment. Regular sufferers may be helped by prophylactic
such as β2-microglobulin. As such dialyzer membranes quinine sulfate. Nausea, vomiting, and headache also are
were developed with larger pore sizes designed to remove fairly common during hemodialysis. Mild chest and back
more middle-sized solutes, termed high-flux, or expanded pains may be related to complement activation by the dialy-
hemodialysis, and studies have suggested a survival advan- sis membrane and are less common with biocompatible
tage for high-flux dialysis.24 Hemodiafiltration, which membranes. Some patients may have allergic reactions to
involves the ultrafiltration of fluid across a high-flux dia- heparins, or some of the organic chemicals that are released
lyzer with compensatory reinfusion of ultrapure dialysate, from the glues in the dialyzer cap, or from the plastic poly-
increases middle-sized molecule clearances further. This mers used in the blood lines, particularly if there has been
approach is often tolerated better by patients with unstable minimal rinsing of the dialysis circuit. Pyrogen or microbial
cardiovascular systems, with less frequent intradialytic contamination of the dialysate occasionally may occur.
hypotension.25 In addition to achieving larger convective Patients developing fever and/or rigors on dialysis must
exchange, hemodiafiltration has been reported to improve be examined carefully for evidence of infection of dialysis
patient survival compared with high-flux hemodialysis.26 access; blood cultures should be taken and patients empiri-
The key to adequate hemodialysis is reliable vascular cally treated with broad-spectrum antibiotics while await-
access (see Chapter 5). Poorly functioning access inevitably ing cultures.
leads to poor dialysis and increases morbidity. Reliance on
central venous catheter access exposes patients to increased
PERITONEAL DIALYSIS
risk of bacterial infection, venous thrombosis, and stenoses.
The organization and management of hemodialysis units Peritoneal dialysis is the process by which solutes, buffer,
are major exercises. The aim is to use the capital equipment waste products, and fluid are exchanged between the blood
and staff in the most economical way. Dialysis units gener- in the peritoneal capillaries and the dialysate instilled into
ally should have no fewer than 10 stations running two, the peritoneal cavity. This exchange takes place across
3 • Chronic Kidney Failure: Renal Replacement Therapy 43

Creatinine dialysate to plasma ratio 1


1.2

D4h/D0 glucose ratio


0.9
1 1.03 0.8
Fast 0.7
0.8 0.68 0.64
Fast average 0.6 Slow
0.6 0.61 0.5 0.5
Slow average 0.4 Slow average
0.47 0.38
0.4 0.3 Fast average
0.34 Slow 0.26
0.2 0.2 Fast
0.1 0.11
0 0
0 1 2 3 4 0 1 2 3 4
Dwell time (hours) Time (hours)
A B
Fig. 3.1 Changes in creatinine (A) and glucose (B) during a standard 2-L peritoneal dialysis exchange during a 4-hour dwell are used to categorize
patients in terms of transporter status. Although fast transporters have higher creatinine clearances, as the glucose gradient falls quicker, they are prone
to sodium retention due to reduced osmotically driven ultrafiltration. D0 refers to the initial glucose concentration; otherwise dialysate concentrations
refer to 4-hour drain.

the peritoneal barrier, which comprises the capillary wall, or net zero ultrafiltration with a 2.27% glucose exchange.
interstitial matrix, the visceral mesothelial cells, and over- Patients differ in the speed at which creatinine moves from
lying glycocalyx. The efficiency of solutes cleared depends capillary blood to dialysate and corresponding loss of the
on the vascularity and surface area of the peritoneum, the glucose from the dialysate to the patient (Fig. 3.1). On the
blood flow, the permeability of the capillary-matrix barrier, one hand, fast transporters clear small water-soluble ure-
the volume and frequency of the dialysate instilled, and the mic toxins faster than slow transporters, but as they also
osmotic or oncotic gradient generated by glucose, glucose lose the glucose osmotic gradient, they are more likely to
polymer, or amino acid content of the dialysate. In addition, have lower ultrafiltration, and increased risk of becoming
there will be a variable lymphatic absorption of glucose and volume-overloaded.
glucose polymer from the dialysate.
Automated Peritoneal Dialysis
Dialysis Adequacy Automated peritoneal dialysis (APD) is an increasingly
The efficacy of peritoneal dialysis can be altered only by popular form of peritoneal dialysis. Patients connect
changes in the volume and frequency of exchanges. Weekly themselves to a peritoneal dialysis machine for a series of
creatinine clearance and urea clearance (measured using overnight cycling exchanges. The duration of therapy is
the weekly Kt/V) are used to assess adequacy of peritoneal typically between 7 and 10 hours, with 4 to 7 cycles of 1.5
dialysis.16 The CANUSA study prospectively followed 680 to 3.0 L. Slow transporters require longer cycle dwell times,
patients after starting continuous ambulatory peritoneal with fewer larger-volume cycles, whereas faster transport-
dialysis (CAPD).27 Although patient survival was related to ers require shorter cycle dwell times, with more frequent
Kt/Vurea (5% increase in relative risk of death for every 0.1 cycles. If patients do not have adequate residual renal func-
decrease in Kt/Vurea), this reduced over time as a result of the tion for solute clearance or volume control, then they may
loss of residual renal function with no change in peritoneal also require one long daytime exchange, sometimes termed
dialysis clearance. A larger study also found a correlation continuous cycling peritoneal dialysis, or even two daytime
with residual function but not peritoneal dialysis clearance, additional exchanges, opti-choice peritoneal dialysis.
and currently there is no evidence that increasing perito- APD is a good treatment for children and the elderly
neal dialysis dose reduces morbidity or mortality.28 Most because it can be set up at home by a relative or caregiver
circumstantial data suggest that more dialysis is better, and and allowed to run automatically overnight. It is also a good
the current consensus is that the target should be above a treatment for patients who are active and working, because
combined Kt/Vurea (dialysis and residual renal function) of it frees them from daytime exchanges.
1.7 per week. V is estimated from height, weight, age, and
sex, but the ideal body weight should be used to avoid inac- Continuous Ambulatory Peritoneal Dialysis
curacies in obese or fluid-overloaded patients. Similarly, CAPD is the most widely used form of peritoneal dialysis
a combined urinary and peritoneal creatinine clearance worldwide and relies on the longer dwell phase to make up
of 50 L/1.73 m2 also has been suggested as a target.16 As for the lower frequency of exchanges. Dialysis is continu-
residual renal function reduces, it may become impossible ous, exchanges following one another, usually without
to achieve target clearance, even with larger and more fre- interruption. To deliver sufficient dialysis, three to five 1.5-
quent exchanges, especially for larger patients, and transfer to 3-L exchanges are performed every 24 hours. The usual
to hemodialysis is necessary. routine starts in the morning with the drainage of the over-
night dialysis fluid and is followed by the installation of the
Dialysis Transport first bag of dialysate of the day. The next change is before
After infections, the next common cause of peritoneal dialy- lunch, then late afternoon, and the last immediately before
sis treatment failure is loss of ultrafiltration. Transport status retiring to bed. Each exchange typically takes 30 to 40
should be assessed by a standard 4-hour dwell with a 2.0-L minutes to complete. If patients have a tendency to absorb
exchange, and ultrafiltration failure defined as <200 mL net fluid from the overnight exchange, they should drain out
ultrafiltrate with a 3.86% or greater glucose concentration, and cap off the catheter overnight and start dialysis on a
44 Kidney Transplantation: Principles and Practice

dry peritoneum in the morning. The volume of dialysate The disadvantages include peritonitis, exit site infections,
instilled depends on the abdominal capacity of the patient ultrafiltration failure, the absorption of glucose leading to
(1.5 L for small adults and children, 2 L for regular-sized weight gain, and the amount of time required to go through
adults, and 3 L for large men). The glucose concentra- the disciplined exercise of performing an aseptic exchange,
tion depends on the amount of ultrafiltration required to and storage space for dialysis fluids and disposables. Thus
keep the extracellular fluid volume constant. To generate patients without suitable accommodation or ready access
a higher volume, strong bags of 2.27% and even 3.86% to clean running water, or those unable or unwilling to
glucose are provided, usually as the first exchange of the perform aseptic exchanges, are not suitable candidates
day and only overnight in slow transporters. However, for peritoneal dialysis. However, peritoneal dialysis can be
7.5% icodextrin dialysate solutions can be used to replace successfully performed in the favelas of Brazil, and in some
hypertonic glucose (2.27% or 3.86%) for a long daytime or countries assistance is provided to either set up automated
overnight dwell and are particularly useful for fast trans- peritoneal dialysis cyclers and connect patients or per-
porters. Icodextrin exerts an oncotic pressure and has a form CAPD exchanges. Some patients, particularly young
more gradual and sustained effect on ultrafiltration than women, refuse peritoneal dialysis, because of embarrass-
glucose-based dialysates. ment with the catheter and an esthetic dislike of the discom-
fort and appearance of a distended abdomen.
PRACTICAL CONSIDERATIONS
POSTTRANSPLANTATION
For all types of peritoneal dialysis, a properly placed perito-
neal dialysis catheter is required (see Chapter 5). The intra- Some practices choose to remove peritoneal dialysis cath-
peritoneal tip should be in the pelvis, free from the omentum. eters at the time of renal transplantation, particularly if
The internal cuff must be watertight and sewn outside the immediate graft function occurs or is expected, such as in
peritoneum. The track should be diagonal, and the catheter living donor transplants. In other circumstances, the cath-
emerge away from the belt line with either a downward or eter is left after transplantation, and provided that graft
lateral pointing exit site and the external cuff 2 cm from the function is adequate, the peritoneal dialysis catheter can be
skin exit site. The major peritoneal dialysis companies pro- capped off and left. The catheter exit site should be dressed
vide a selection of peritoneal dialysis solutions, varying in as per usual, and the catheter typically removed after 3
volume, osmotic strength, pH, and calcium concentration. months, when the transplant function is stable. If the graft
The key issue in peritoneal dialysis is the avoidance of fails within 3 months, the catheter needs to be flushed to
infection introduced at the time of connection of the dialysis remove debris and fibrin. Infection occasionally is intro-
bag to the transfer set, which is attached to the peritoneal duced at this first exchange after a break (see also subse-
dialysis catheter. Originally the connection was made by quent section on Dialysis Posttransplant).
spiking the port of the peritoneal dialysis bag, and it was
associated with increased risk of introducing infection. COMPLICATIONS
Improvements in device connections have led to the intro-
duction of the flush-before-fill technique, when about 30 Loss of Ultrafiltration
mL of sterile fresh dialysate from the new dialysate bag at A proportion of patients (10%–30% by 5 years but increas-
the beginning of each exchange is flushed to the waste efflu- ing with time) retain fluid after more than 4 hours’ dwell
ent dialysate bag and not infused into the peritoneal cavity, and have poor ultrafiltration, even with more hypertonic
so that bacteria introduced at the time of the connection of dialysates. This may occur transiently after peritonitis and
the new bag are rinsed away. In addition, skin microorgan- is more likely to become a chronic problem after multiple
isms can migrate along the catheter track, and thus exit site attacks. Patients present with hypertension and fluid over-
care is important to limit skin colonization. Chlorhexidine load, and it is important to exclude constipation, loss of
or alcohol-based antiseptic wipes are typically used in com- residual renal function, or poor compliance with dialysis or
bination with topical antiseptic or antibiotic creams. excessive fluid intake. Ultrafiltration failure because of fast
transport status is referred to as type 1 failure. Low ultrafil-
INDICATIONS FOR AND ADVANTAGES OF tration associated with slow transport is termed type 2 fail-
ure, and is associated with loss of surface area, usually as a
PERITONEAL DIALYSIS
result of encapsulating peritoneal sclerosis (EPS).
Peritoneal dialysis has some major advantages over hemo- In the early stages, reduced ultrafiltration can be man-
dialysis, but there are equally many limitations and draw- aged by using shorter dwell times and leaving the perito-
backs. In the basic CAPD mode, peritoneal dialysis is at least neum dry overnight. The use of an overnight solution of
25% cheaper than hemodialysis in that it does not require 7.5% icodextrin (a glucose polymer) that is oncotically
an expensive dialysis machine and large numbers of techni- active, but too large to be rapidly absorbed, is an alterna-
cally expert trained nursing staff. Because peritoneal dialy- tive strategy. When fluid retention occurs during daytime
sis is a home-based therapy, patients are not constrained exchanges, even with medium or strong bags, the only
by their dialysis slots. However, peritoneal dialysis becomes option to avoid abandoning peritoneal dialysis is to convert
expensive if beset by complications, such as peritonitis, that from CAPD to APD, sometimes with an additional daytime
require hospital admission. CAPD is particularly useful in icodextrin exchange.
patients experiencing practical difficulties with hemodialy- Standard peritoneal dialysates are acidic solutions con-
sis, for example, vascular access problems or cardiovascular taining high concentrations of glucose and lactate. Heat
instability. It has social advantages for children and moth- sterilization generates a number of glucose degradation
ers of young children. products (GDPs). Neutral pH dialysates are now available,
3 • Chronic Kidney Failure: Renal Replacement Therapy 45

replacing some or all the lactate with bicarbonate. These into each CAPD exchange (gentamicin loading dose 8 mg/L
come as twin- or three-chamber bags, mixing the chambers then 4 mg/L, and first- or second-generation cephalosporin
immediately before use. By separating the glucose and acid 250 mg/L loading dose then 125 mg/L). Patients treated
solution from bicarbonate, it reduces GDPs, and these more by APD can convert to CAPD until the pathologic organ-
biocompatible dialysates may help to preserve peritoneal ism is identified with antibiotic specificities, or patients can
ultrafiltration. be treated with vancomycin and ciprofloxacin as discussed
earlier or given intraperitoneal antibiotics in a single day-
Peritonitis time exchange (gentamicin 0.4 mg/kg/day then adjusted
Peritonitis is a potentially serious problem and the most according to antibiotic levels in combination with a first-
common complication of peritoneal dialysis, with an aver- or second-generation cephalosporin 1.5 g/day). In centers
age incidence of one episode per 24 to 36 patient-months. with a high risk of fungal peritonitis, antifungal prophylaxis
The incidence of peritonitis is reduced by using the flush- with fluconazole should be coadministered either at 50 mg
before-fill technique, and some reports suggest that using daily, or 200 mg for 3 days.29
antibiotic exit site creams can further reduce the risk of Unless patients are ill, they are generally treated as out-
peritonitis. patients (approximately 80%). Depending on the results
Around 57% of cases are caused by gram-positive organ- of the microbiology cultures, one or both antibiotics are
isms, 18% caused by gram-negative organisms, and about continued, or new antibiotics started in cases of resistance,
15% are culture-negative. Coagulase-negative staphylo- and in cases of no bacterial growth both antibiotics are
cocci are the most common cause of peritonitis, and typi- continued. If the patient fails to respond within 48 hours
cally cause a relatively mild symptomatic peritonitis, which and is unwell, microbacteriology advice should be sought.
usually responds promptly to treatment with intraperito- Further delay in response should raise the possibility of fun-
neal antibiotics, but can relapse because of penetration of gal peritonitis. Occasionally (<5%), peritonitis is secondary
organisms into the catheter biofilm. Staphylococcus aureus to intraabdominal disease, such as diverticulitis, appendi-
usually causes a severe peritonitis with marked systemic citis, or perforated viscus, and if suspected, laparotomy is
features, including high fever and hypotension. Response indicated.
to antibiotics is less good, and the catheter is more likely Patients with recurrent episodes of peritonitis from the
to be removed for a nonresolving infection. Pseudomonas same organism may have persistent infection of the cath-
accounts for approximately 5% of cases and also tends to eter or its track or it may be a result of the buildup of a bio-
invade the catheter biofilm, often necessitating catheter film on the catheter that provides protection for bacteria
removal, because infections often recur. from antibiotics. This buildup can be treated by removal of
Peritonitis usually presents as cloudy bags (apparent the peritoneal dialysis catheter with a rest on hemodialysis
with >50 leukocytes/μL) and abdominal pain, but the lat- and later placement of a fresh catheter or by removal and
ter may precede the former by one exchange or 1 to 2 days. replacement at the same operation. Recurrent peritonitis
There may be fever (more common in children), nausea and may be a result of poor technique, and this should be reas-
vomiting (approximately 30%) and, when severe, hypoten- sessed carefully.
sion. Abdominal tenderness with peritonism is the major
clinical finding. The effluent peritoneal fluid after a 4-hour Exit Site and Tunnel Infection
dwell contains greater than 100 leukocytes (>50% neutro- Nearly half of all exit sites are colonized by skin bacteria. S.
phils) per μL (normally <10/μL, although the number may aureus colonization of the exit site is usually associated with
be higher after a dry period, for example, during the day in nasal carriage and used to account for more than 50% of all
patients on APD).29 Peritonitis may be present with lower exit site infections. Prevention is the best strategy; perito-
cell counts, particularly with short dwells. Cloudy bags are neal catheters should be placed so that the peritoneal dialy-
occasionally a result of macrophages and/or eosinophils sis catheter exits preferably vertically downward or laterally
and are not associated with peritonitis. to aid drainage of the subcutaneous tunnel, and the exit site
The diagnosis is made in the presence of any two of the is positioned to avoid mechanical damage from skirt or pant
following: abdominal pain, cloudy bags, or the presence waist bands and belts. Patients should be educated about
of bacteria on Gram stain.29 The differential diagnosis the importance of daily washing, drying, avoiding force-
includes intraperitoneal infections, including appendicitis, ful removal of scabs, and avoiding tugging the catheter by
cholecystitis, diverticular disease, pancreatitis, and bowel careful tethering.
perforations. Typically, intraabdominal pathology is Exit site infections can be further reduced by using
accompanied by increased peritoneal effluent, red blood antiseptic creams and gels and topical antibiotics, such
cells, and leukocytes, whereas peritoneal dialysis perito- as mupirocin and gentamicin.30 Because mupirocin will
nitis has very few red blood cells, despite increased leuko- reduce gram-positive organism colonization, then propor-
cytes. In cases of clinical doubt or in cases failing to respond tionately more infections will be gram-negative, in particu-
promptly to therapy, a computed tomography (CT) abdomi- lar Pseudomonas. Although the use of topical antibiotics is
nal scan should be performed to exclude additional intraab- recommended by the International Society for Peritoneal
dominal pathology, before proceeding to catheter removal Dialysis,30 some centers have not followed this advice
in refractory cases. because of concerns of introducing antibiotic resistance.
A variety of regimens are successful, for example, intra- Exit site infections should be guided by advice on local
peritoneal vancomycin, 2 g (1 g if <60 kg), left to dwell for 4 microbiology; because staphylococci are common, 2 weeks
to 6 hours and repeated after 5 to 7 days depending on sys- of flucloxacillin is typically first-line treatment. Recur-
temic levels, and oral ciprofloxacin, or antibiotics injected rence may be treated effectively by adding rifampicin to
46 Kidney Transplantation: Principles and Practice

flucloxacillin. Sometimes the outer cuff extrudes, and shav- hemodialysis and also after transplantation, with patients
ing this may solve the problem, but catheter replacement presenting with signs of small-bowel obstruction. Although
may be necessary, especially if there is evidence of a tunnel EPS can be precipitated by severe peritonitis, the only com-
infection or recurrent peritonitis. mon factor linked to its development is the duration of peri-
toneal dialysis therapy, with an incidence of 3% at 5 years,
Anatomic Complications then rapidly rising to around 15% at 10 years. Diagnosis is
Abdominal fluid increases intraabdominal pressure and often made at laparotomy, but CT scan appearances may
predisposes to the development of hernias, which are rela- show small-bowel tethering, small-bowel dilatation with
tively common (10%–15% of all peritoneal dialysis patients septation, and calcification of the mesentery. Tamoxifen
and higher in patients with polycystic kidneys). Although and steroids may have a role during the inflammatory
often asymptomatic, prompt repair is essential because phase, but not once fibrosis is established, then total par-
they inevitably enlarge, and there is the risk of obstruction enteral nutrition and surgery in specialized centers may be
and/or strangulation. Acid reflux, indigestion, and a feel- required.18
ing of fullness also are more common than in patients on
hemodialysis. Hernias can be confirmed by performing a CT Metabolic Complications
peritoneogram. The peritoneal fluid contains high concentrations of glu-
Infusion pain on draining fluid into the abdomen is rela- cose. It has been estimated that 200 g of glucose (more with
tively common early on but usually settles. This pain can be many high-concentration bags and during peritonitis) may
reduced by using neutral pH dialysate at body temperature be absorbed daily, equivalent to 800 kcal. This absorption
and may be improved by slowing the inflow rate. Drain pain may cause obesity, hyperglycemia, and hyperinsulinemia.
at the end of a cycle is more common with cyclers. It may The glucose load stimulates fat synthesis, and hypertriglyc-
improve by treating constipation; if not, then clamping the eridemia is common. Cholesterol levels also rise during the
line after the fast drain has finished and starting the next first year on CAPD. Albumin levels often are low. The expla-
cycle usually resolves pain. If air is introduced into the sys- nation is multifactorial, including simple dilution resulting
tem, it relocates under the diaphragm, causing severe pleu- from volume expansion, losses in the fluid, and malnutri-
ritic pain referred to the shoulder. tion because of poor appetite, especially during and after
Scrotal or labial edema occurs when fluid leaks from the peritonitis.
peritoneum to subcutaneous tissues. This condition usu- About 25% of CAPD patients are hypokalemic, and they
ally occurs soon after insertion of the Tenckhoff catheter may need potassium-sparing diuretics or supplements.
and spontaneously settles after resting for a few weeks. A few need a stringent low-potassium diet. Some patients
Occasionally, patients have a congenital communication using 2.5- to 3-L lactate-based exchanges run high bicar-
between the pleura (usually right side) and peritoneum, bonate levels, which usually settle when switched to neu-
resulting in a hydrothorax. Because these leaks tend to be tral pH bicarbonate solutions. Alkalosis has been associated
small, diagnosis can be difficult because biochemical analy- with lethargy, nausea, and headaches.
sis of the pleural fluid is typically equivocal—neither peri-
toneal dialysate nor serum—with no leak demonstrated
on CT peritoneogram. However, if 2 L of 7.5% icodextrin
Choice and Planning for
is instilled, then sampling the pleural fluid 4 hours later, the Individual Patient
and checking for a starch reaction with iodine, may help
confirm that the fluid is dialysate, but a negative test does Patients with CKD are never cured, and often change RRT
not exclude a peritoneal leak. Some centers will attempt modality during their lifetime. This requirement for different
chemical or surgical pleurodesis with a temporary transfer modalities of RRT, each with its own limitations and risks,
to hemodialysis. calls for a coordinated and multidisciplinary approach.
Other problems include lower back pain caused by the Because renal transplantation offers better survival and
change in posture secondary to large-volume daytime quality of life, patients should first be assessed for suitability.
exchanges. There are few absolute contraindications, and the decision
has to be based on an overall assessment of the patient’s
Encapsulating Peritoneal Sclerosis current fitness and ability to withstand surgery and immu-
Repeated exposure to peritoneal dialysis fluids causes nosuppression, and subjective assessment of compliance
changes to the peritoneum, typified by increased interstitial and estimated life expectancy. Availability of kidneys may
matrix deposited in the parietal peritoneum, and brown- also influence selection policy.
ing discoloration resulting from deposition of advanced The best first option is a preemptive transplant from a liv-
glycosylation end-products. As the peritoneum thickens, ing related or living donor because this avoids the need for
numerous small capillaries and lymphatics develop, which an initial period of dialysis and the creation of permanent
increase the effective vascular surface area, and patients vascular access. It also enables planning of transplantation,
typically become faster transporters. In addition to this nat- such that blood group and even HLA-mismatched donors
ural thickening of the peritoneum, some patients develop can also be considered. Planning can avoid major disrup-
an inflammatory reaction, which can lead to deposition of tion to the life of young patients at a crucial stage in their
a cocoon-like fibrosis around the small bowel, with teth- education or career. Nephrologists should emphasize the
ering leading to small-bowel obstruction, malabsorption, much longer survival of living donor transplants, the lower
and 50% mortality in severe cases.18 EPS can occur many morbidity, and the reduced doses of immunosuppression.
years after switching modalities from peritoneal dialysis to Although it must be acknowledged that the transplants
3 • Chronic Kidney Failure: Renal Replacement Therapy 47

ultimately may fail, families of young patients should be TABLE 3.6 Drugs That Require Dose Reduction or
reminded of the benefits of 10 years of dialysis-free life in Avoidance in Patients With Chronic Kidney Disease Stage 5d
the early years after the development of CKDd, which would
allow completion of education, establishment of a career, Drug Class and
Specific Drugs Comment
and having a family.
If living donor transplantation is not possible, the next ANTIMICROBIAL
best option is to put the patient forward for a preemptive Aminoglycosides Reduce dose, ototoxic
cadaver donor transplant, if possible. Apart from avoid- Acyclovir Reduce dose
Cephalosporins Reduce dose
ing dialysis, this option allows the patient to spend a Cotrimoxazole Reduce dose, increases creatinine
longer period in the recipient pool; because there is less Ethambutol Reduce dose, retinal toxicity
urgency, the patient can wait for a better-matched kid- Ganciclovir Reduce dose
ney. However, some nephrologists object to this policy on Nitrofurantoin Avoid, neuropathy
the grounds that it is unfair to patients already on dialysis Penicillins Avoid high doses, fits
Vancomycin Reduce dose, ototoxic
who may have waited a long time for a transplant. Given
the shortage of cadaver kidneys and the superior survival ANESTHETIC
of well-matched grafts, this objection can be rejected. It Pancuronium Avoid, prolonged paralysis
Gallamine Avoid, prolonged paralysis
always has to be accepted that preemptive transplanta- Opiates Reduce dose, prolonged effect
tion cannot be guaranteed success, and patients should Suxamethonium Increases plasma potassium concentrations
be counseled that they may need dialysis postoperatively ANALGESICS
for hyperkalemia, or in cases of delayed graft function and
Nonsteroidal Avoid, risk losing residual renal function
graft failure. inflammatories
If dialysis is necessary before transplantation can be per- Opiates Reduce dose and frequency as metabolites
formed, then the best holding treatment depends on the accumulate, causing toxicity. Less likely
length of time waiting for a graft. If the patient is physically with fentanyl and oxycodone
fit, not sensitized, and has a potential living donor available, GASTROINTESTINAL
then peritoneal dialysis is a good holding treatment because Metoclopramide Be aware of extrapyramidal effects
it avoids the need for creating permanent or temporary vas- H2 antagonists Reduce dose
cular access. Occasionally, such patients would be held on Magnesium salts Monitor magnesium concentration
hemodialysis using a temporary internal jugular venous CARDIAC
catheter, but this should only be considered a short-term Digoxin Reduce dose and frequency
option and discouraged because of the risks of infection and β-Blockers May need to reduce dose
the development of venous stenoses, which may exclude Spironolactone Avoid, hyperkalemia
ACE inhibitors Monitor creatinine and potassium
the arm from future arteriovenous fistula formation. If the Clofibrate Avoid, muscle injury
wait for a transplant is indeterminate, then patients should
ORAL HYPOGLYCEMICS
go down the local patient pathways for creating vascular
Chlorpropamide Avoid, hypoglycemia
access for those opting for hemodialysis, and for peritoneal Tolbutamide Reduce dose
dialysis either elective insertion of an embedded peritoneal Biguanides Avoid, lactic acidosis
dialysis catheter, or timely catheter insertion closer to the
time of dialysis initiation. ACE, angiotensin-converting enzyme.

saline flushing (100 mL every 30 minutes) and citrate-


Dialysis Posttransplant based dialysate (Citrasate), which removes the need for
systemic anticoagulation. For the patient with an unstable
The need for dialysis after transplantation is determined by cardiovascular system, hemofiltration or hemodiafiltration
early graft function, which in turn depends on many fac- is preferable, with higher dialysate sodium concentration (5
tors related to the donor organ (see Chapter 14). During mmol/L above serum sodium up to 145 mmol/L) and dialy-
any period when graft function is absent or compromised, sate cooled to 35°C.32 The usual vascular access should be
it is essential to modify doses of drugs for which the pre- used for intermittent hemodialysis treatments, but fistulae
dominant route of excretion is the kidney. Particular care should not be used for continuous forms of RRT.
is required for aminoglycoside antibiotics (see Chapter 31), Vascular access should be monitored in the postopera-
certain muscle relaxants (see Chapter 13), and opiates, the tive phase. Periods of hypotension predispose to collapse
active metabolites of which accumulate in CKD5. Impor- and thrombosis of fistulae and arteriovenous grafts, espe-
tant examples are listed in Table 3.6.31 cially those with preexisting stenoses. The accesses must be
explored and flow restored as soon as possible, not only for
HEMODIALYSIS their immediate use, but also for the long term should the
graft fail. Rehabilitation of patients with failed renal trans-
Hemodialysis should be performed as for any postopera- plants is prolonged greatly if they lose their vascular access
tive patient (i.e., as remote from the surgical procedure as well.
as is safe for the control of potassium and fluid balance). Postoperatively, because of the inflammatory reaction
In practice, this is usually more than 24 hours after the initiated by the transplanted kidney, capillary endothelial
operation. The main risk is hemorrhage from the operation leak leads to patients being several kilograms above their
wound and biopsy sites, but in practice the risks are small dialysis target weight, as sodium-containing intrave-
with short daily dialyses, coupled with regular predialyzer nous fluids are administered to maintain systemic blood
48 Kidney Transplantation: Principles and Practice

pressure and central venous pressure. It is reasonable then this should be withdrawn gradually, over months. Even
to dialyze patients to above their so-called pretransplant then the patient is at risk of hypoadrenalism and should
target weight in the postoperative period. This approach be warned that he or she will require an increased dose of
minimizes the risk of intradialytic hypotension, which steroids to cover any intercurrent illness. It is reasonable to
may delay the graft opening up and even predispose to perform a short Synacthen test if there is doubt about the
thrombosis of the anastomosis. However, it must be rec- adrenal status to ensure that withdrawal has been effected
ognized that some of the anti-T-cell agents predispose to safely.
an acute pulmonary leak, leading to pulmonary edema, Patients with failing grafts should have the same nephro-
which can be life threatening in volume-overloaded logic care as CKD4 patients in terms of volume, blood pres-
patients (see Chapter 20). sure, cardiovascular risk factor, phosphate control, and
renal bone disease and renal anemia management, with
CONTINUOUS AMBULATORY PERITONEAL appropriate dietary advice.5
DIALYSIS
Peritoneal dialysis can be performed safely after the trans- RRT Modality and Survival
plant procedure, provided that the peritoneum has not been
breached; the risk can be minimized by draining the peri- Although the overall survival for patients receiving trans-
toneum before surgery. However, because of inflammatory plants is greater than for those remaining on dialysis, it
milieu posttransplantation, many patients behave as fast must be recognized that there is a selection bias of the
transporters, requiring shorter dwell times and higher glu- younger, fitter patients for transplantation; for example,
cose dialysates to prevent further fluid retention. unadjusted 2014 US Renal Data Survey (USRDS)19 month
If clear fluid is observed leaking from the transplant survival was lowest for hemodialysis (78.9%), compared
wound, measuring the glucose concentration will con- with peritoneal dialysis (90.3%) and first cadaveric trans-
firm peritoneal dialysate, in which case dialysis should be plantation (96.5%). Even after adjusting for age, gender,
stopped immediately, the peritoneum should be drained, ethnicity, and primary renal disease, then 2-year survival
and hemodialysis then used to support the patient. Perito- is greater for both living donor (96.1%) and cadaveric
neal dialysis can be reinstituted at about 7 days but should transplantation (91.7%), compared with peritoneal dialysis
be with low volumes or preferably using a cycling machine (79.2%) and hemodialysis (67.5%). Wolfe and colleagues
that allows small volumes and short dwell times. Patients reported that long-term mortality was 48% to 82% lower
should not be transplanted during an episode of peritonitis. among transplant recipients than among dialysis patients
on the waiting list,34 and a later analysis showed better
5- and 10-year survival for transplanted patients who had
Return to Dialysis After dialysis for 6 months (78% and 63% respectively) or less
Transplant Failure compared with those who had dialyzed for 2 years (58%
and 28% respectively).35 Selection bias has to be considered
The return to dialysis after transplantation is a difficult when interpreting survival for different RRT modes. How-
period for the patient and the nephrologist. Not only are ever, there have been very few studies actually randomizing
there understandable psychological difficulties, but there patients to either peritoneal dialysis and hemodialysis, and
are frequently physical problems. Early graft failure is less the largest study (NECOSADS) did not show major differ-
traumatic for the patient who has not enjoyed indepen- ence between standard dialysis modes.36 Home hemodialy-
dence from dialysis. Immunosuppression, particularly sis patients have better survival; this may be due not only
steroid doses, should be reduced rapidly and the patient to patient selection, but also because they have access to
returned to the usual dialysis schedule. Grafts that fail early better-quality dialysis in terms of more frequent and longer
usually are removed surgically, but the need for this often dialysis sessions.
is precipitated by the reduction in immunosuppression. It Survival on dialysis depends not only on patient factors,
has long been recognized that patient survival is affected but also on acceptance criteria and transplantation rates.
adversely by graft failure. In the UK and other countries there is an increasing trend
Patients whose long-term grafts have failed present more toward offering elderly comorbid patients conservative or
complex problems. The temptation to maintain a failing palliative care (no dialysis). This can then confound dialysis
graft should be resisted because it is better for the patient to survival data, from countries such as Germany, which have
be established on dialysis than to suffer the continual effects limited conservative management programs. As expected,
of uremia and immunosuppression.33 The physician has to age has the greatest effect on survival; for example the UK
discuss with the patient the appropriate modality, which Renal Registry reported 1- and 10-year survival for patients
may be different from that employed before transplantation. aged 18 to 64 of 94.2% and 53.0% respectively, which fell
Time spent on peritoneal dialysis pretransplantation should to 81.9% and 7.6% for those aged >65 years (http://www.
be taken into account when estimating the risk of EPS. If renalreg.org/publications-reports/). Thereafter additional
the graft is failing slowly, then the appropriate patient path- comorbidities, in particular diabetes, reduce life expec-
way for establishing vascular or peritoneal access should be tancy, as with the general population. However, despite
followed. advances in medicine and dialysis technology, the overall
Patients frequently have many side effects from immu- 5-year survival of RRT patients lies between that of ovar-
nosuppression, including osteoporosis, skin atrophy and ian and bowel cancer, which reinforces the need to improve
malignancy, hyperglycemia, hypertension, and secondary RRT and tackle the causes of accelerated cardiovascular
hyperparathyroidism. If patients are prescribed prednisolone, disease in CKD patients.3,4
3 • Chronic Kidney Failure: Renal Replacement Therapy 49

4. Webster AC, Nagler EV, Morton RL, Masson P. Chronic kidney dis-
Quality of Life ease. Lancet 2017;389(10075):1238–52.
5. Vanholder R, Fouque D, Glorieux G, et al.; European Renal Association
European Dialysis; Transplant Association (ERA-EDTA) European Renal;
Whereas mortality is a hard end point, patients also want Cardiovascular Medicine (EURECA-m) working group. Clinical manage-
information about expected quality of life when trying to ment of the uraemic syndrome in chronic kidney disease. Lancet Diabetes
decide upon treatment modalities. Unfortunately, qual- Endocrinol 2016;4(4):360–73.
ity of life is difficult to quantitate, especially because 6. Smart NA, Titus TT. Outcomes of early versus late nephrology
referral in chronic kidney disease: a systematic review. Am J Med
there are major differences in the ages and other circum- 2011;124(11):1073–80.
stances of patients being managed by the various modali- 7. Chan KE, Maddux FW, Tolkoff-Rubin N, et al. Early outcomes among
ties. Employment is an important criterion by which the those initiating chronic dialysis in the United States. Clin J Am Soc
quality of life or outcome of RRT can be judged, and in Nephrol 2011;6(11):2642–9.
8. Black C, Sharma P, Scotland G, et al. Early referral strategies for man-
this respect transplant recipients are at a major advan- agement of people with markers of renal disease: a systematic review
tage. There is no doubt that the quality of life of a trans- of the evidence of clinical effectiveness, cost-effectiveness and eco-
plant patient with minimal complications is far greater nomic analysis. Health Technol Assess 2010;14(21):1–184.
than that of even the most well-adjusted hemodialysis 9. Kagoma YK, Weir MA, Iansavichus AV, et al. Impact of estimated GFR
patient.37 Despite the initiation of dialysis, many patients reporting on patients, clinicians, and health-care systems: a system-
atic review. Am J Kidney Dis 2011;57(4):592–601.
continue to have multiple symptoms, with pain, fatigue, 10. Juutilainen A, Kastarinen H, Antikainen R, et al. Trends in estimated
pruritus, and constipation present in more than 50%. kidney function: the FINRISK surveys. Eur J Epidemiol 2012;27:
Potentially treatable symptoms include bone and joint 305–13.
pains, insomnia, mood disturbance, sexual dysfunction, 11. Hossain MP, Palmer D, Goyder E, et al. Social deprivation and preva-
lence of chronic kidney disease in the UK: workload implications for
paresthesia, and nausea. primary care. QJM 2012;105(2):167–75.
The advantages are less clear-cut if comparison is made 12. Roderick P, Rayner H, Tonkin-Crine S, et al. A national study of prac-
between an independent home hemodialysis patient and tice patterns in UK renal units in the use of dialysis and conservative
a transplant patient who suffers major complications of kidney management to treat people aged 75 years and over with
immunosuppression. The ANZDATA Registry is one of the chronic kidney failure. Southampton (UK): NIHR Journals Library;
2015. PMID 25855842.
few to examine the issue of quality of life systematically. The 13. Klahr S, Levey AS, Beck GJ, et al. The effects of dietary protein restric-
1998 report showed that 85% of transplant recipients were tion and blood-pressure control on the progression of chronic renal
judged to have a normal quality of life in that they were able disease: modification of diet in Renal Disease Study Group. N Engl J
to carry on normal activities with only minor symptoms.38 Med 1994;330:877.
14. Lowrie EG, Laird NM, Parker TF, et al. Effect of haemodialysis prescrip-
Only 44% of dialysis patients were in this category; how- tion on patient morbidity: report from the National Cooperative Dialy-
ever, 83% of dialysis patients were self-caring; 9% required sis Study. N Engl J Med 1981;305:1176.
considerable or special assistance. However, more recent 15. National Kidney Foundation. NKF-DOQI clinical practice guidelines
reports did not show any difference, but this again may be for hemodialysis adequacy. Am J Kidney Dis 1997 Sep;30(3 Suppl.
biased by RRT acceptance criteria.39 2):S15–66.
16. Woodrow G, Davies S. Peritoneal dialysis. Renal Association Clinical
Guidelines. 5th version. 2010. Available online at: www.renal.org/
USEFUL WEBSITES clinical/guidelinessection/peritoneal [accessed 11.12.18].
17. Mikhail A, Srivastora R, Richardson R. Anaemia. Renal Association
Acute Dialysis Quality Initiative, www.adqi.net. clinical guidelines. 5th version. 2010. Available online at: www.renal.
org/clinical/guidelinessection/AnaemiaInCKD.aspx [accessed 11.12.18].
Australia and New Zealand Renal Registry, www.anzdata. 18. Alston H, Fan S, Nakayama M. Encapsulating peritoneal sclerosis.
org.au. Semin Nephrol 2017;37(1):93–102.
British Transplant Society clinical guidelines, www.bts. 19. Ikizler TA, Cano NJ, Franch H, et al. Prevention and treatment of pro-
org.uk. tein energy wasting in chronic kidney disease patients: a consensus
European Dialysis and Transplantation clinical practice statement by the International Society of Renal Nutrition and Metab-
olism. Kidney Int 2013;84(6):1096–107.
guidelines, http://www.ndt-educational.org/. 20. Runge VM. Critical questions regarding gadolinium deposition in
International Society for Peritoneal Dialysis Clinical Guide- the brain and body after injections of the gadolinium-based contrast
lines, www.ispd.org. agents, safety, and clinical recommendations in consideration of the
Kidney Disease Improving Global Outcomes, www. EMA’s Pharmacovigilance and Risk Assessment Committee recom-
mendation for suspension of the marketing authorizations for 4 linear
kdigo.org. agents. Invest Radiol 2017;52(6):317–23.
National Institute for Health and Clinical Excellence, www 21. King-Wing Ma T, Kam-Tao Li P. Depression in dialysis patients.
2.evidence.nhs.uk. Nephrology (Carlton). 2016;21(8):639–46.
Renal Association clinical guidelines, https://renal.org/. 22. Chilcot J, Davenport A, Wellsted D, et al. An association between
UK Renal Registry, http://www.renalreg.com. depressive symptoms and survival in incident dialysis patients.
Nephrol Dial Transplant 2011;26(5):1628–34.
United States Renal Data System, www.usrds.org. 23. Cooper BA, Branley P, Bulfone L, et al. A randomized, controlled trial
  
of early versus late initiation of dialysis. N Engl J Med 2010;363(7):
609–19.
References 24. Locatelli F, Altieri P, Andrulli S, et al. Hemofiltration and hemodiafil-
1. Srisawat N, Hoste EE, Kellum JA. Modern classification of acute kidney tration reduce intradialytic hypotension in ESRD. J Am Soc Nephrol
injury. Blood Purif 2010;29(3):300–7. 2010;21(10):1798–807.
2. Levey AS, Eckardt KU, Tsukamoto Y, et al. Definition and classification of 25. Locatelli F, Martin-Malo A, Hannedouche T, et al. Effect of membrane
chronic kidney disease: a position statement from Kidney Disease: Improv- permeability on survival of hemodialysis patients. J Am Soc Nephrol
ing Global Outcomes (KDIGO). Kidney Int 2005;67(6):2089–100. 2009;20(3):645–54.
3. Nordio M, Limido A, Maggiore U, Nichelatti M, Postorino M, Quin- 26. Davenport A, Peters SA, Bots ML, et al. Higher convection volume
taliani G. Italian dialysis and transplantation registry. Survival in exchange with online hemodiafiltration is associated with survival
patients treated by long-term dialysis compared with the general pop- advantage for dialysis patients: the effect of adjustment for body size.
ulation. Am J Kidney Dis 2012;59(6):819–28. Kidney Int 2016;89(1):193–9.
50 Kidney Transplantation: Principles and Practice

27. Canada-USA (CANUSA) Peritoneal Dialysis Study Group. Adequacy 34. Wolfe RA, Ashby VB, Milford EL, et al. Comparison of mortality in all
of dialysis and nutrition in continuous peritoneal dialysis: association patients on dialysis, patients on dialysis awaiting transplantation, and
with clinical outcomes. J Am Soc Nephrol 1996;7:198. recipients of a first cadaveric transplant. N Engl J Med 1999;341:1725.
28. Paniagua R, Amato D, Vonesh E, et al. Effects of increased peri- 35. Meier-Kriesche HU, Kaplan B. Waiting time on dialysis as the stron-
toneal clearances on mortality rates in peritoneal dialysis: ADE- gest modifiable risk factor for renal transplant outcomes: a paired
MEX, a prospective, randomized, controlled trial. J Am Soc Nephrol donor kidney analysis. Transplantation 2002;74:1377–81.
2002;13(5):1307–20. 36. Korevaar JC, Feith GW, Dekker FW, et al. Effect of starting with
29. Li PK, Szeto CC, Piraino B, et al. ISPD peritonitis recommenda- hemodialysis compared with peritoneal dialysis in patients new
tions: 2016 update on prevention and treatment. Perit Dial Int on dialysis treatment: a randomized controlled trial. Kidney Int
2016;36(5):481–508. 2003;64(6):2222–8.
30. Szeto CC, Li PK, Johnson DW, et al. ISPD catheter-related infection 37. Griva K, Stygall J, Ng JH, et al. Prospective changes in health-related
recommendations: 2017 update. Perit Dial Int 2017;37(2):141–54. quality of life and emotional outcomes in kidney transplantation over
31. Ashley C, Currie A, editors. The renal drug handbook. 4th ed. Oxford, 6 years. J Transplant 2011;2011:671571.
UK: Radcliffe Publishing; 2014. 38. Disney APS, Russ GR, Walker R, et al., editors. ANZDATA registry
32. Vinsonneau C, Camus C, Combes A, et al. Continuous venovenous report 1998. Adelaide, South Australia: Australia and New Zealand
haemodiafiltration versus intermittent haemodialysis for acute renal Dialysis and Transplant Registry; 1998.
failure in patients with multiple-organ dysfunction syndrome: a mul- 39. Sayin A, Mutluay R, Sindel S. Quality of life in hemodialysis, peri-
ticentre randomised trial. Lancet 2006;368(9533):379–85. toneal dialysis, and transplantation patients. Transplant Proc
33. Ong SC, Gaston RS. Medical management of chronic kidney dis- 2007;39:3047–53.
ease in the renal transplant recipient. Curr Opin Nephrol Hypertens
2015;24(6):587–93.
4 The Recipient of a Renal
Transplant
JEREMY R. CHAPMAN

CHAPTER OUTLINE The Patient with Chronic Kidney Disease Miscellaneous Infections—Syphilis,
General Concepts Strongyloides, Toxoplasmosis,
Fitness to Transplant Trypanosoma
Appropriateness to Transplant Malignancy (See Also Chapter 35)
Waitlisting and Allocation of Deceased Psychiatric Disease and Drug Dependency
Donor Organs (See Also Chapter 40)
Counseling Bone
What the Patient Needs to Know Gastrointestinal Tract
Consent Diabetes
What the Potential Living Donor Needs to Type 1 Diabetes Mellitus
Know Type 2 Diabetes Mellitus
What the Family Needs to Know Renal Disease
Specific Medical Considerations Recurrent Renal Disease
Cardiac Urogenital Tract Abnormalities
Vascular Coagulation Disorders
Respiratory Obesity
Hepatic Disease Psychosocial Factors
Hepatitis B Sensitization and Transfusion Status
Hepatitis C Previous Transplantation
Other Liver Disease Preparation for Transplantation
Infectious Disease (See Also Chapter 31) To Join and Remain on the Deceased Donor
Vaccination Strategies Waiting List
Human Immunodeficiency Virus To Undergo Elective Living Donor
Transplantation
Other Viral Infections—CMV, EBV, HHV6/7,
HHV8 To Undergo Deceased Donor
Transplantation
Dental

extreme the poorly advised, badly educated, frightened, or


The Patient with Chronic Kidney noncompliant tread a hazardous course to dialysis and all
Disease too often, to an earlier death.
It is not surprising that those who plan their treatment
The insidious nature of progressive uremia deludes many well and receive a living donor transplant preemptively
patients with chronic kidney disease (CKD) into failing to before the requirement for dialysis tend to have the best
take the opportunity to understand their disease; learn outcomes.1,2 It should also not be surprising that access
about their dialysis, transplant, and palliative care options; to preemptive living donor transplantation is better for
and plan their future. Relatively asymptomatic decline in patients with higher socioeconomic status3 (Fig. 4.1). Most
renal function may explain why many ignore the early people are, however, treated by hemodialysis or peritoneal
warnings of disease and fail to optimize their medical care dialysis for weeks, months, or years before finally being
until they are in a hospital emergency department, hyper- transplanted. There are some benefits to pretransplant
kalemic, acidotic, and confused. The physician’s primary dialysis, especially if the patient is chronically debilitated by
aims in patients with CKD are to prevent progression of their CKD, or if time is needed to enable a family member to
disease and to ensure that the patient and their family understand the benefits of offering a kidney. For some peo-
understand the prognosis and their need to make longer ple, the experience of dialysis provides an important dem-
term decisions. Intelligent, well researched, and financially onstration of life without a renal transplant, which usually
stable members of the community tend to prepare better for strengthens their resolve both to undergo the operation and
the onset of end-stage kidney disease, whereas at the other accept the long-term consequences of immunosuppression.
51
52 Kidney Transplantation: Principles and Practice

Living donor (including preemptive) Deceased donor


0.2 0.2

Cumulative incidence
Relative survival

0.1 0.1

Q1 (disadvantaged) Q1 (disadvantaged)
Q2 Q2
Q3 Q3
Q4 (advantaged) Q4 (advantaged)

0 0
0 2 4 6 8 10 0 2 4 6 8 10
Years Years
Fig. 4.1 The role of socioeconomic status in access to living and deceased donor transplantation in Australia. Q, Quartile of socioeconomic disadvan-
tage. (Reproduced from Grace BS, Clayton PA, Cass A, McDonald SP. Transplantation rates for living- but not deceased-donor kidneys vary with socioeconomic
status in Australia. Kidney Int. 2012;83(1):138–145.)

This chapter attempts to identify the issues that the patient, individual might expect6 instead of a measured comparison
his or her family, and the community must consider before of quantity of life.7,8
deciding to have a renal transplant. In countries without sufficient access to dialysis, the deci-
sion to receive a renal transplant is obvious for almost all
patients, because transplantation is the only alternative to
a slow death from uremia.4 This does assume access both
General Concepts to lifelong immunosuppressant drugs and specialist medical
follow-up, either of which may not be available or afford-
FITNESS TO TRANSPLANT able and without which transplantation becomes a futile
The patient has, in principle, only one simple question to delusion. Understanding by a patient and their family of
consider: will the quality and quantity of life be better after the lifelong commitment needed for a successful transplant
a transplant than on dialysis? For many people the answer is critical because patient and graft survival rates are sub-
is clear and unequivocal—either because the alternative of stantially affected by compliance with follow-up and adher-
long-term dialysis treatment is not available or unafford- ence to medication protocols.9 Transplantation has been
able,4 or because transplantation is the obvious solution promoted as a cure, when it is actually a complicated treat-
because they are young and otherwise fit. For some people, ment requiring regular follow-up by specialists working in
however, the answer is clouded in uncertainty because of sophisticated medical centers using expensive drugs. If the
the relative unavailability of organs for transplantation, or patient and his of her family fail to understand the costs,
because they have comorbid conditions that will be exacer- level of follow-up, and the adherence that will be required of
bated by the operation or the ensuing immunosuppression. them, then it is likely that the published statistics of average
Quality of life is perhaps the single most important issue survival will not apply to them.
for most people and yet the field has found the social sci- Predicting the success rate after transplantation relies on
ences difficult to grasp, and there are few studies comparing characteristics of both the recipient and the donor. Prob-
quality of life on dialysis and after transplantation.5 Most ably the most comprehensive studies comparing transplant
clinicians find it hard to identify living individuals with a recipients with those remaining on dialysis come from the
lower quality of life after a successful transplant than they US where it has been possible to track the outcomes of all
had or would have had on dialysis, and thus find it easy to individuals entered onto the transplant waiting list and
advocate for transplantation. This approach ignores the compare those who were transplanted with those who
patient deaths and graft failures, and the diminished qual- remained on dialysis.7,8,10 These studies show that trans-
ity of life for those who struggle with the consequences of plantation carries the greater risk of death for the first
immune deficiency, infections, and malignancies. Both 3 months or so, and reverses after that point in time so
governments and transplant programs tend to substitute that the risk of death is equal by 6 to 9 months, thereafter
graft survival data for true quality of life data and use it as favoring the transplant recipient dependent on donor and
a surrogate, but objective, measure of the success that each patient risk factors.11–13 Similar analyses show that the
4 • The Recipient of a Renal Transplant 53

Prevalent Dialysis Mortality


Australian patients vs. general population
Female Male
.6
.4

Annual death rate (95% CI)


.2
.1

.01

.001

20 40 60 80 20 40 60 80
Age
Dialysis patients General population
A

Prevalent Transplant Mortality


Australian patients vs. general population
Female Male

.6
Annual death rate (95% CI)

.4
.2
.1

.01

.001

20 40 60 80 20 40 60 80
Age
Transplant patients General population
B
Fig. 4.2 (A) Prevalent dialysis patient mortality compared with the general age matched population. (B) Prevalent transplant patient mortality
compared with the general age matched population. CI, confidence interval. (ANZDATA Registry 2016 Annual Report, reproduced with permission.)

patient who is transplanted preemptively carries an advan- transplantation and received a transplant survive better
tage compared with one who has needed a period of time on than those remaining on dialysis. The presumption, from
dialysis; furthermore, the longer the period of dialysis the the recipient’s perspective, should thus be to be assessed for
worse the outcome.1 fitness to be transplanted.
The patients’ expectations from a successful transplant The next issue, for those fit enough for the procedures, is
may be that they will die in old age with a perfectly func- the source of a donated kidney. In many countries this will
tioning kidney. The reality is that they will die much ear- be restricted to a living donor kidney from a relative or an
lier than an age- and sex-matched individual without renal altruistic friend17 (Fig. 4.3). Where the luxury of deceased
disease, and more than half will have lost their graft before donation exists, organ allocation policies become critical to
they die.14,15 the options. Careful counseling and education is needed to
Analysis of patient perspectives of the most feared risks arrive at a decision. Consider an elderly father or mother
after a transplant are a little at odds with physician views in their late 60s without any comorbid conditions, decid-
because graft loss is feared more than death.5 ing whether or not to accept a donation from a 30-year-
old son or daughter. The son or daughter may consider it
appropriate to offer a kidney to their parent, acknowledging
APPROPRIATENESS TO TRANSPLANT
the small but real immediate and possible long-term risks
The patient with access to both dialysis and transplanta- to themselves. The parent may consider it highly inap-
tion can compare their prognosis using simple outcome propriate to place their offspring at even the slightest risk
statistics of dialysis and transplant patients,16 as seen in to provide a few years of better quality of life for them. In
Fig. 4.2. Even elderly people who have been accepted for the reverse situation it may be considered obvious for a
54 Kidney Transplantation: Principles and Practice

35000

30000

25000

20000

15000

10000

5000

0
Africa Americas Europe Eastern South Western Pacific
Mediterranean East Asia

Living donor Deceased donor


Fig. 4.3 Numbers of living and deceased donor kidney transplants by World Health Organization region as reported by the Global Observatory on
Donation and Transplantation, 2015. (From http://www.transplant-observatory.org/data-charts-and-tables/chart/.)

20-year-old recipient to be transplanted, but not to accept all of these guidelines is to provide objective and explicit
a donor offer from an elderly parent, both because of the advice to the clinician assessing an individual patient.
increased risk of donation by an older person and because Most achieve a comprehensive perspective, but there are
of the worse outcome predicted from an older kidney. It is substantial differences over the methodology behind each
the responsibility of the transplant unit to provide indepen- recommendation. There is a largely consistent approach to
dent medical advice to ensure that both the donor and the the required level of renal impairment and medical comor-
recipient can arrive at a considered and individual decision. bidities, with most also providing broad statements on age
criteria, life expectancy, and both psychological and social
WAITLISTING AND ALLOCATION OF DECEASED factors. The effect of high body mass index (BMI) is inter-
preted as both a criterion for surgical acceptability and a
DONOR ORGANS
medical criterion predicting cardiovascular and other mor-
In those countries with deceased organ donor programs bidity and mortality; however, advice is based on expert
the community must decide how to manage the alloca- opinion rather than hard evidence. There is as yet little
tion of deceased donor kidney offers. In most countries effect of formal assessment of frailty for which the data are
national, regional, or local waiting lists provide access to developing.20 Shortage of donor organs in most environ-
the offer, although in some places where deceased donors ments leaves a tension between maximizing utility on the
are rare, selection may be ad hoc. The percentage of dialysis one hand and equity on the other. In resolving this tension,
patients actively listed for transplantation varies quite sur- most guidelines attempt to promote a transparent decision-
prisingly.18 There are not only barriers because of age and making process in the absence of a robust formula that is
comorbidities but also from demographic, geographic, and universally applicable to each individual.
socioeconomic factors. The community—when asked— Considerable statistical effort has been expended on pre-
almost always regards “equity of access” as the most domi- dicting patient outcomes based on pretransplant factors in
nant factor, but the reality is a complex balance of medical both the donor and the recipient.21,22 The donor element is
utility and sometimes conflicting notions of equity. Health- assessed using the kidney donor profile index (KDPI), which
care professionals are far from agreed on what should con- uses only data derived from the donor before donation.
stitute the criteria for medical eligibility and whether or not The recipient element is derived by the Scientific Registry
it is appropriate to include consideration of social and life- of Transplant Recipients (SRTR) from age, dialysis dura-
style factors, ability to understand and adhere to treatment, tion, prior transplant, and diabetes to create an expected
and medical estimates of prognosis. posttransplant survival (EPTS). Using these elements it was
There are many published guidelines on assessment possible to derive a new kidney allocation system in the
for transplant waiting lists.19 The stated aim of almost US—the early results of which are becoming evident.23
4 • The Recipient of a Renal Transplant 55

In association with new evidence-based reviews,24 there


is now detailed advice on specific issues in recipient assess- BOX 4.1 Checklist for Pretransplant
ment. The EPTS and KDPI can be calculated for an individ- Education of the Patient and Their Family
ual using a published calculator.25 Different countries have 1. Medical condition
assessed local adaptations of these formulae with varying General cardiorespiratory fitness for operation
reproducibility, demonstrating the principles are widely Effect of obesity
applicable but the detail has to be adjusted to the demo- Vascular system suitability for operation
graphics of each environment. Urologic complications
Risks of recurrent renal disease
2. Fitness for lifelong immunosuppression
Infections
Counseling Malignancies, especially skin cancers
Cardiovascular risk factors
WHAT THE PATIENT NEEDS TO KNOW 3. Histocompatibility and organ donor source; effect on out-
comes
The transplant unit has the responsibility to provide each 4. Waiting times and allocations systems on the deceased organ
patient with advice based on his or her own medical condi- donor waiting list
tions and education about the options for long-term treat- 5. Availability and donor outcomes of living donor procedures
ment (Box 4.1). The starting point for such education is 6. Financial costs and specific risks of the donor and transplant
thus a comprehensive evaluation of the suitability, avail- procedures including disease transmission from the donor
ability, and financial cost of dialysis options. The quality 7. Financial and adverse event costs of prophylactic immuno-
of physical and emotional well-being provided by dialysis suppressive and antiinfective drugs
therapy usually becomes abundantly clear to most patients, 8. Long-term follow-up protocol
9. Short- and long-term risks of graft failure and death after
either through meeting other patients already on dialysis,
transplantation
or through dialysis education programs and finally, defini- 10. Consideration of acceptance of extended criteria donor
tively, through direct personal experience, except in that organs
small minority able to undergo preemptive transplantation. 11. Patient specific issues (e.g., options for pancreas transplanta-
A comprehensive evaluation needs to be provided by the tion in diabetics and liver/kidney in primary oxalosis)
transplant unit of each individual’s medical risks if he or
she were to undergo a renal transplant. Much of the rest of
this chapter details the medical assessment, but a checklist
is provided in Table 4.1. This list includes those issues that
affect the individual patient’s technical transplantability in
addition to the short- and long-term factors that influence TABLE 4.1 Screening Tests That Should Be Routinely
such outcomes as graft and patient survival. In assessing Considered Before a Living Donor Transplant or
Acceptance Onto a Transplant Waiting List
the patient’s suitability for a transplant operation the physi-
cian will be focused on the heart and lungs, the surgeon on GENERAL HISTORY AND PHYSICAL EXAMINATION
the blood vessels and bladder. The surgeon will need to dis- DIAGNOSIS OF CAUSE OF RENAL DISEASE, WITH SPECIFIC TESTS
cuss the various complications and risks of the surgical pro- AS REQUIRED
cedure, whereas the physician discussion should revolve
VIRUS EXPOSURE
around the drugs, long-term risks, and follow-up protocols.
HIV antibody HIV 1 and HIV 2
Providing the patient with sufficient knowledge on organ Hepatitis B HBsAg, HBcAb, HBsAb
allocation processes, the pros and cons of particular donor Hepatitis C HCVAb, (HCV RNA if HCVAb positive)
kidney offers, and on the financial costs that he or she will Cytomegalovirus CMV IgG
be expected to bear, are easily left out of a traditional medi- Herpes virus Herpes simplex IgG, Herpes varicella
cal consultation. Most established transplant programs zoster IgG, HHV6, and HHV7 IgG
Epstein–Barr virus EBV IgG
thus have additional formal education sessions provided
by a range of specialized coordinators, social workers, and OTHER INFECTIOUS DISEASE
pharmacists. The Internet and social media increasingly In endemic areas: PPD skin testing
provide a wide range of both good and bad information, Trypanosoma cruzi serology
Coccidioides serology
which patients will certainly access extensively. A guide to
Strongyloides serology
the better material and warnings against the bad sources of West Nile Virus serology
information should also play a part in the advice provided HTLV I and II serology
by the transplant program. HHV8 serology
Toxoplasma screening
Syphilis screening
CONSENT Chest x-ray with follow-up tests as required if abnormal

It is, of course, normal practice to seek written informed Other disease


consent just before undergoing any surgical procedure Electrocardiogram
and all transplant operations are preceded by such a ritual Echocardiogram/stressed cardiac test—with follow-up tests as
signing of a legalistically phrased document. Somewhere required if abnormal
Abdominal ultrasound (kidneys, gall bladder, liver, spleen)
among this scant and hastily signed documentation will be Vascular duplex ultrasound (femoral/carotid)
the expectation that the individual has accepted all the risks
56 Kidney Transplantation: Principles and Practice

of transplantation, from transmission of serious disease as emotionally involved onlookers and supporters for the
from the donor, through to the side effects of every drug that patient. Transplant units vary in the way in which infor-
they will be given. Many patients will also be presented with mation is provided about the potential to donate a kidney,
a dazzling array of research protocols to sign up for, with with some distributing information packets directly to all
patient information sheets of many pages of closely typed known family members or encouraging attendance at edu-
and densely constructed language more designed to pro- cation sessions covering the issue, whereas others await
tect the researcher, hospital, and pharmaceutical company specific individual approaches before providing information
than inform the patient. This documentation of “consent” on living related donation. In countries with low deceased
often takes place under pressure of time and in the middle organ donation rates, the increasing attention being placed
of the night, sometimes even via the telephone. It is hard upon living donation creates the ambience for routine dis-
to see how anything provided by the patient in the haste semination of information to family members and friends.29
of the anesthetic workup, no matter what it is written on, Accurate provision of specific relevant information is of
can be argued to be informed consent. Legal opinions have course dependent on the consent of the recipient to release
been given that suggest that no reliance can be placed on his or her private medical details. Asking the question: Is
a patient’s decision taken under the duress of an immedi- there anyone in the family who would donate you a kid-
ate pretransplant consent, unless backed by extensive prior ney? elicits some interesting insights into the dynamics of
education and information. In constructing education pro- families with members with serious chronic illness. Some
grams, it would be wise for the transplant unit to consider patients refuse to consider a discussion of their illness with
the traditional “operation consent form” a legally valueless their families, whereas others are glad that an independent
protection. person is prepared to raise awareness in their family regard-
ing the seriousness of their illness.
What the Potential Living Donor Needs to Know Lack of information is almost always the starting point for
A potential living donor usually needs to be provided infor- a breakdown in trust and communication between patients,
mation on both the recipient outcomes and the donor oper- their families, and their medical attendants. For this reason
ation with its attendant risks to decide on whether or not to it is important that even the most distant of families are
proceed. There has been considerable recent data analysis aware of the possibility of a poor outcome from transplanta-
of the risks of donating a kidney (see Chapter 7). tion and the importance of compliance with medication and
A donor who expects only a successful outcome from his follow-up to the long-term success of the transplant.
or her donation has a chance of being badly disappointed.
It is thus essential that the best estimates of the risks of
recipient death and graft failure are clearly laid before the
donor.26,27 It is also important for most donors to appreci-
Specific Medical Considerations
ate the dialysis alternatives available to the patient and the CARDIAC
deceased donor waiting list times. In countries with sub-
stantial waiting lists and long waiting times, living dona- The first consideration of any CKD patient undergoing a
tion clearly offers huge advantages that are not so clearly major operation is the state of their heart. Dialysis patients
apparent where deceased donor waiting times are short. In and especially diabetic dialysis patients have high inci-
countries with high deceased donor rates, the advantages dences of both symptomatic and asymptomatic ischemic
of providing a better kidney with better long-term survival heart disease and thus a careful evaluation of the heart is
may be less obvious, but just as real. essential as summarized in the recent American Heart Asso-
A living donor must provide fully informed consent to a ciation guidelines.30 The diversity of evidence and opinion
surgeon with no conflicts of interest through his or her care leaves open the alternative approaches as identified in the
of the potential recipient.28 In addition it is relevant for the European Best Practice Guidelines.31
donor to appreciate his or her blood group and histocom- There is agreement that all patients need a careful clini-
patibility match with the recipient and any concerns that cal history and examination, including an electrocardio-
there might be about the crossmatching data. A general gram and chest x-ray. In addition, most units will perform
overview of the risks that the recipient faces will help ensure an echocardiogram to assess left ventricular function and
that the small percentages of procedures that end in disas- a stressed ECG, echocardiogram, or myocardial perfu-
ter are not followed by endless recrimination and litigation. sion study to exclude significant ischemic heart disease in
More importantly, a well prepared donor is better able psy- asymptomatic high-risk patients. Whereas CKD itself is the
chologically to face the future after a failed transplant or strongest risk factor for coronary artery disease, it is also
even death of the recipient. important to assess obesity, family history, lipid profile,
blood pressure, smoking history, and diabetes to define risk.
What the Family Needs to Know Attitudes to smoking history vary among transplant units
The families of pediatric patients are best regarded as if from outright refusal to transplant those patients that con-
they were the patient, with respect to the information and tinue to smoke, through to more liberal approaches.32
counseling that they require, though there are special Some transplant programs require routine coronary
considerations that young age brings to bear on the deci- angiography before acceptance onto a waiting list. There
sion making in renal transplantation. The family of the is certainly a rationale for such an approach given the
adult patient is in a special situation compared with other high levels of coronary disease uncovered by such a strat-
areas of medicine, because the family represent a potential egy.33 The only randomized trial of surgical or medical
source of organ donation and cannot simply be thought of intervention in this situation (diabetics with CKD) was so
4 • The Recipient of a Renal Transplant 57

unequivocal about the value of intervention that the trial the surgeon planning to perform the transplant. Absence
was halted and the nonintervention arm offered surgery or of intermittent claudication and presence of palpable femo-
angioplasty.34 The weaknesses of this study (it only assessed ral and pedal pulses may be sufficient to confirm that renal
diabetics and the optimum medical therapy would not be transplantation is technically feasible. There are, however,
considered optimum today) and the lack of alternative ran- many potential recipients with a high risk of severe vascu-
domized studies leave the field with uncertainties, but also lar disease, where duplex ultrasound scanning of the femo-
a very clear view that diabetic patients need comprehensive ral and carotid vessels will identify those at significant risk
cardiac evaluation. of peripheral or cerebrovascular events either during or
A lesser strategy is to use a noninvasive test such as a after transplantation.41
stressed dopamine echocardiogram,35 or stressed nuclear Selection of patients with known preexisting peripheral
study,36 or more recently, CT coronary angiography37 as a vascular disease must include a general assessment of their
screening method for asymptomatic and low-risk patients, prognosis and specific assessment of the vascular supply
thus reserving coronary angiography for those with symp- needed for the transplant operation. The largest numbers of
toms, significant risk factors, or a positive screening test. A patients commencing dialysis in most developed countries
systematic review of the predictive value of the screening are elderly obese type 2 diabetics, and many have severe
tests suggested a dobutamine stress echocardiogram had peripheral vascular disease. Only a very small proportion
the best accuracy.38 Despite careful screening close to the of such patients prove to be suitable for transplantation
time of the transplant, patients with disease in minor coro- because of the combined effects of obesity, cardiac, and vas-
nary branches will be operated on, sometimes leading to cular disease on their operative mortality and 3- to 5-year
postoperative chest pain, troponin leakage into the blood, survival rates.42 Two-thirds of dialysis patients requiring
and cardiovascular instability. lower limb amputations are dead within 2 years, implying
Proceeding to transplantation in patients with normal that this group of patients have such a poor prognosis that
left ventricular function and normal coronary vasculature only few, very highly selected patients should be accepted
is the easy decision. The more complex issue is deciding who for transplantation.43
to transplant despite known cardiac disease, which needs to Symptomatic cerebrovascular disease presents a sepa-
be considered not only on its own merits, but also because of rate problem in selection for transplantation. A history of
the implications that it carries for widespread vascular dis- transient ischemic attacks obviously promotes a search for
ease.17 There is no evidence-based answer to this question a cardiac or carotid vascular cause, which if diagnosed and
as yet, and clinicians must thus rely on local opinion based resolved or treated, need not contraindicate subsequent
decisions, guided by some general principles: transplantation.44
  
One group of patients that need particular attention are
Treatable coronary and valvular disease is almost always
□ 
those with adult polycystic kidney disease, especially if they
worth treating before transplantation rather than after-
have a personal or family history of cerebral aneurysm.5
ward, both because of the risks posed by the cardiac dis-
Evaluation of such high-risk patients requires cerebral
ease during the transplant procedure and because of the
vascular imaging such as cerebral computed tomography
risks that cardiac interventional procedures carry in the
(CT) angiography or a magnetic resonance image angio-
presence of immunosuppression and a functioning trans-
gram to exclude berry aneurysms before proceeding to
plant.
transplantation.45
It is usually wiser to avoid transplantation if, despite
□ 

treatment of coronary artery and/or valvular disease,


there remains a substantial risk of infarction of a large RESPIRATORY
area of myocardium, or there is substantial left ventricu-
Assessment of respiratory disease in the potential transplant
lar dysfunction. Cardiac disease is the single largest cause
candidate has two purposes: to identify patients at risk from
of mortality in both the dialysis and transplant popula-
the anesthetic and to identify patients who will be at risk
tions and there is little evidence that transplantation will
of life-threatening infection in the long term as the result
beneficially alter the outcome of ischemic heart disease.
of immunosuppression. The former is based around assess-
There is less certainty with respect to congestive cardiac
ment of smoking and both acute reversible and chronic
failure, where poorly dialyzed patients may recover sig-
obstructive airway disease. It is no different from the assess-
nificant function when uremia and chronic fluid overload
ment that must be made before any elective operation.46
are corrected by transplantation.39
The latter is a more complex decision and remains largely
In patients with severe and irreversible cardiac dysfunc-
□ 
subjective. The diseases of importance are bronchiecta-
tion, the remaining consideration is the option of com-
sis, tuberculosis, and prior fungal infections, all of which
bined heart and kidney transplantation, available to a
may quickly become uncontrollable under the influence
limited number of young and otherwise healthy individu-
of immunosuppression. Formal evaluation of the degree of
als transplanted in highly specialized centers.40,41
respiratory compromise and the frequency and severity of
infective exacerbations will determine the advisability of
transplantation of the patient with bronchiectasis.
VASCULAR
Active pulmonary tuberculosis must be identified from
There is an absolute requirement for an available recipient routine chest x-ray and effectively treated before consid-
artery for anastomosis of the transplant renal artery. Ather- eration of transplantation.47 Patients at high risk of reac-
omatous iliac arteries that have been ossified through years tivation of tuberculosis after transplantation include those
of CKD management must thus be carefully assessed by from areas with high endemic rates, a history of exposure,
58 Kidney Transplantation: Principles and Practice

calcified lesions on chest x-ray or elsewhere, and a positive patients with all genotypes of hepatitis C and severe renal
QuantiFERON Gold test.48 Transplant units in endemic impairment.59
areas tend to advise high-risk patients to have a full treat- The best strategy for the future will be to treat patients
ment course for tuberculosis after transplantation. In devel- before transplantation, but excellent results can be achieved
oped countries, based on slender evidence,43 management by posttransplantation treatment if either the recipient or
usually involves adding a prophylactic course of isoniazid the donor or both have been infected with hepatitis C. This
for 6 months.49 has transformed the effect of hepatitis C and relegated it to
an expensive irritant rather than a major threat to patients’
HEPATIC DISEASE lives.
Hepatitis B Other Liver Disease
The majority of dialysis patients with past or current hepa- Potential renal transplant recipients may suffer from other
titis B will have been identified through routine testing of causes of significant liver disease (see Chapter 32), such as
serum for hepatitis B surface antigen (HBsAg) and antibod- alcoholic liver disease, polycystic liver in association with
ies to hepatitis B core and surface antigens. Many dialysis polycystic kidney disease, or cholelithiasis. It is thus impor-
programs have a routine hepatitis vaccination policy to tant and relatively simple to assess both liver function and
improve protection from cross infection, even though vac- appearance of the liver on ultrasound. Fatty infiltration of
cination is much more effective if administered before the the liver is the commonest finding of such screening proto-
need for dialysis.48 Thus most patients being assessed for cols and may be associated with diabetes but is not in itself a
transplantation have been screened for prior exposure to contraindication to transplantation. Severe liver disease, no
hepatitis B. matter what the cause, inhibits acceptance for renal trans-
Data from transplantation of chronically infected HBsAg- plantation of most patients. There is diversity of opinion on
positive patients, predominantly gained in the 1980s and the role of prophylactic cholecystectomy in dialysis patients
1990s, demonstrate worse outcomes than for HBsAg-neg- with known gallstones, with the larger studies not support-
ative patients.50 ing this approach, but some advocate routine pretransplant
Knowledge of the status of the liver histology is impor- screening and surgery for known gallstones.60
tant in predicting outcomes after renal transplantation,
with mortality correlating with preexisting chronic hepatic INFECTIOUS DISEASE (SEE ALSO CHAPTER 31)
inflammation grades.51 It is now clear that use of posttrans-
plant lamivudine therapy has not carried the survival ben- Vaccination Strategies
efits hoped for but entecavir was associated with reduced General community protection from infectious disease will,
mortality.51 Choice of immunosuppression after trans- in most countries, have led to routine childhood vaccina-
plantation may influence the progression of hepatitis with tion against measles, mumps, polio, rubella, diphtheria,
concern expressed about steroids, azathioprine, and cyclo- tetanus, pertussis, Haemophilus influenzae B, varicella-zos-
sporine reactivating hepatitis B in the chronic carrier.52 ter, and more recently, human papilloma virus has been
Hepatitis B is not a contraindication to renal transplanta- added to the list. Pneumococcal and hepatitis B vaccina-
tion, but established cirrhosis raises the option of combined tion programs, though widespread, are far from universal.
liver and renal transplantation.53 Data at present indicates It is especially important in pediatric practice to ensure
that survival advantage is conferred by combined renal that vaccination has not been forgotten among the prob-
and liver transplantation only in those patients with a low lems of pediatric renal failure.61,62 In adult practice it is also
model for end-stage liver disease (MELD) score of 16 to 20.54 important to understand each patient’s vaccination history
and to remedy deficiencies as soon as possible because the
Hepatitis C responses to vaccines are generally impaired in the dialy-
Hepatitis C represents different challenges to transplant sis population.63 Vaccination for meningococcus is espe-
programs in different countries, with high prevalence in cially important in patients for whom eculizumab may be
dialysis programs where there is reuse of dialysis consum- considered.64
ables and in patients transfused in the 1980s. The natural Vaccination of patients after transplantation is either
history of hepatitis C infection leading to high proportions contraindicated with live vaccines, or may fail with killed
of patients eventually developing significant liver disease55 antigen vaccines, because the medication used to prevent
has now been transformed by the advent of the new direct- allograft rejection is well designed to suppress production
acting antiviral agents (DAAs).56 of an antibody response to a viral antigen. Mycophenolate
Limitations to treatment of patients with CKD stages 4 mofetil, for example, is especially capable of preventing
and 5D have been compounded by the responsiveness of antibody production after vaccination.65 Live vaccines are
different hepatitis C genotypes to the first wave of DAAs. At absolutely contraindicated after transplantation, with the
this moment there are no ribavirin-free and interferon-free commonest errors being the use of yellow fever vaccination
treatments available for genotypes 2, 3, 5, and 6. Sofosbu- in travelers to South America and chicken pox vaccination
vir regimens cannot be used safely in patients with severe with attenuated virus, leading to life-threatening dissemi-
renal impairment or on dialysis because of renal excretion nated pox virus infection in transplant recipients.
of active metabolites57 though treatment after transplanta-
tion is both safe and effective.58 Recent advances in DAAs Human Immunodeficiency Virus
have, however, resolved both these problems with a com- The advent of highly active antiretroviral therapy (HAART)
bination of glecaprevir and pibrentasvir providing a 98% in the mid 1990s both transformed the outcome of patients
sustained virologic response after 12 weeks of treatment in infected with human immunodeficiency virus (HIV) and
4 • The Recipient of a Renal Transplant 59

challenged the renal community to treat CKD with both Syphilis, Strongyloides, and toxoplasmosis have all been
dialysis and transplantation.66 The outcomes of HIV-posi- reported as opportunistic reactivations after transplanta-
tive (HIV+) patients after transplantation are acceptable in tion. In most areas of the world transplant programs require
the short- and mid-term.67 It has been demonstrated that a heightened awareness and lower threshold for suspicion
use of HIV+ donors is acceptable for HIV+ patients and has of these diseases, rather than specific strategies for these
been pioneered in countries with high prevalence of HIV.68 uncommon problems. Testing for syphilis serology is still
The accepted criteria for listing HIV+ patients include practiced by many programs, but is not seen as essential in
stable CD4+ T cell levels >200/μL and compliant on recipients from most developed countries. It is good practice
HAART with HIV RNA undetectable and without oppor- routinely to check the patient’s eosinophil count and pur-
tunistic infection.69 The main posttransplant complexity is sue a diagnosis of infection in anyone with a raised count.
the extremely potent pharmacokinetic interaction between
HAART drugs and immunosuppressive drugs via cyto-
MALIGNANCY (SEE ALSO CHAPTER 35)
chrome p450, requiring careful planning and close moni-
toring by both transplant and infectious disease teams. There is a clear and defined additional risk of malignancy
Despite the advances, it is now clear that there are poten- in patients with CKD, especially after transplantation. This
tial long-term consequences of HIV infection of the trans- increased risk is assumed to be a result of an effect of immu-
planted kidney, which have yet to be resolved.70 nosuppression either on normal mechanisms for control of
neoplastic cells or more likely the effect on viral carcinogen-
Other Viral Infections—CMV, EBV, HHV6/7, HHV8 esis.74 This knowledge has been translated to a reluctance
Knowledge of a recipient’s status with respect to all herpes to transplant patients who have had a prior cancer for fear
viruses has become increasingly relevant because of their that immunosuppression will allow recurrence that might
potential effect after transplantation. otherwise not happen.
Chemoprophylaxis for CMV, which also protects recipi- Recent data question this set of assumptions. First, a
ents for HHV6 and HHV7, is usually based upon knowledge number of cancers are increased in CKD patients, in patients
of the donor and recipient CMV serologic status. Transplan- on dialysis, and in patients after transplantation.75 Second,
tation of an EBV-positive organ into an EBV-negative recip- the cancers with substantially increased risk are restricted
ient carries an increased risk of active EBV infection after to skin and lip cancers, renal tract cancers, and those for
transplantation and also of development of posttransplant which a viral etiology is either established or suspected.
lymphoproliferative disease (PTLD). All patients should The implication for the potential transplant recipient is that
thus be tested for antibody status with respect to each of cancers that are now understood to occur at the same rate
the herpes viruses. The Transplantation Society guidelines as in the normal population should probably be considered
on CMV provide a useful basis for understanding the alter- differently to those where the risk is increased.
native testing and therapeutic options available.71 HHV8 It has been standard advice not to transplant a patient
remains a potent risk factor for development of Kaposi’s sar- within 2 to 5 years of diagnosis and definitive treatment
coma posttransplant. of cancer, depending on which cancer is under consider-
ation. Most guidelines suggest careful screening for cancers
Dental in patients on the transplant waiting list.19 Unfortunately
The traditional approach to evaluation of the transplant such blanket rules, though easy to apply, do not take into
recipient includes ensuring adequate dental hygiene and consideration the variability of the biology of the different
review of dentition before acceptance for transplantation. cancers and especially do not consider the individual risks
It is certainly true that gingival hypertrophy was a con- of recurrence. Table 4.1 gives a list of cancer types that are
sequence of higher doses of cyclosporine, especially when known to be increased in dialysis and transplant patients
combined with nifedipine, and that infected dentition may and should thus be viewed with considerable caution in
cause problems after transplantation. This has been amelio- patients being assessed for transplantation. Melanoma, for
rated with use of tacrolimus and alternative antihyperten- example, is known to respond to T cell immunotherapy and
sives and it would now be an unusual candidate in whom has a substantially increased risk after transplantation.76 It
the dentition provides a risk of transplantation sufficient is known to recur in normal individuals and to metastasize
to outweigh the risk of continued dialysis compared with aggressively. Melanoma has also been observed to recur
transplantation, though bacterial endocarditis is certainly after transplantation with long disease-free intervals pre-
important to avoid through prophylactic antibiotics at the transplantation and must thus be approached very conser-
time of dental interventions. vatively despite the advent of checkpoint inhibitors.77 Breast
and prostate cancer, on the other hand, are not increased in
Miscellaneous Infections—Syphilis, Strongyloides, most studies of dialysis and transplant populations but have
Toxoplasmosis, Trypanosoma substantial metastatic potential. To avoid transplanting a
Transplant programs must pay heed to the particular infec- patient who will succumb to metastatic cancer soon after
tious risks that are both endemic and prevalent in their transplantation, it is thus prudent to advise a waiting period
geographic region, to properly evaluate the posttransplant of at least 2 years, depending to a certain extent on the pre-
risks for their transplant recipients. Trypanosoma cruzi, the dicted risk of spread in any given individual.
causative organism of Chagas’ disease, is for example prev- Common cancers occur frequently in dialysis and
alent in South and Central America.72 It may be transmit- transplant patients. It is important not to shift the clinical
ted by donation and reactivated by immunosuppression, emphasis from common cancers to rare cancers, such as
requiring serology and blood polymerase chain reaction Kaposi’s sarcoma, just because these rare cancers occur
(PCR) surveillance and early treatment.73 with a greatly increased risk compared with the general
60 Kidney Transplantation: Principles and Practice

population. The common cancers in the Australian CKD TABLE 4.2 Suggested Disease-Free Time Intervals
population include kidney, bladder, colon, lung, mela- Before Transplantation of Patients With Prior Cancers
noma, breast, and prostate (Table 4.2). There is no spe-
cific guideline for cancer screening before listing a dialysis SITE (ICD 10 Codes)
patient for transplantation. It would, however, be reason- CKD-ASSOCIATED CANCER
able to at least ensure that patients are aware of cancer risks Kidney (C64) >2 years
and advise that screening guidelines recommended in the Renal pelvis (C65) >2 years
general population for cervical, breast, and bowel screening Ureter (C66) >2 years
Bladder (C67) >2 years
have been undertaken.78 Other urinary organs (C68) >2 years
NON-CKD ASSOCIATED
PSYCHIATRIC DISEASE AND DRUG Nonmelanoma skin (Local treatment)
DEPENDENCY (SEE ALSO CHAPTER 40) Lip (C00) (Local treatment)
Tongue (C01–C02) >2 years
Compliance after transplantation and the patient’s Mouth (C03–C06) >2 years
responses to the psychological stresses of transplantation Salivary gland (C07–C08) >2 years
should be uppermost in the minds of clinical teams evalu- Esophagus (C15) >2 years
Stomach (C16) >2 years
ating recipients. Noncompliance with both medication and Small intestine (C17) >2 years
clinical follow-up are among the most distressing and dev- Colon (C18) >2 years
astating causes of loss of grafts. Prevention of this problem Rectum (C19–C20) >2 years
starts with understanding the patient before transplanta- Anus (C21) >2 years
tion and responding to the different risks for noncompli- Liver (C22) (Contraindicated without liver Tx)
Gallbladder (C23–C24) >2 years
ance.79 It is clear that the majority of noncompliant patients Pancreas (C25) >2 years
do not have a psychiatric disorder, but many with a psychi- Larynx (C32) >2 years
atric disorder are at risk of poor compliance. Trachea; bronchus and lung >2 years
There are two dominant reasons for careful evaluation (C33–C34)
Melanoma (C43) >5 years—assess risk of metas-
of the psychiatric state of the potential recipient: their ability tasis
to understand and consent to the transplant procedure, and Mesothelioma (C45) >2 years
the influence of psychiatric disease after transplantation. For- Kaposi sarcoma (C46) >2 years—use TORi immunosup-
mal psychological testing and psychiatric assessment may be pression
required to evaluate a given individual’s capacity to provide Connective and other soft tissue >2 years
(C47–C49)
properly informed consent. Alcohol and drug abuse raise Breast (C50) >5 years
many practical, medical, ethical, and moral questions that also Vulva (C51) >2 years
have to be evaluated carefully in each individual. Abstinence Cervix uteri (C53) >2 years
from chemical dependency would be regarded as essential for Corpus uteri (C54) >2 years
Ovary (C56) >2 years
acceptance to the transplant waiting list by most transplant Penis (C60) >2 years
programs, but it is difficult to assure and monitor in practice. Prostate (C61) >2 years
Testis (C62) >2 years
Eye (C69) >2 years
BONE Brain (C71) >2 years
Thyroid (C73) >2 years
Renal bone disease status and the degree of control of the Hodgkin disease (C81) >5 years
calcium phosphate product are both important indicators Non-Hodgkin lymphoma (C82– >5 years
of bone disease and vascular risk after transplantation.80 In C85)
children the additional consideration of growth potential Leukemia (C91–C95) >5 years
and the effect of uremia on the one hand and corticosteroids Duration before considering transplantation = The period after apparent
on the other are relevant considerations.81 successful cure of the individual cancer when transplantation may be
The past few years have seen an explosion in available considered if investigations substantiate cure of the cancer. Note also
therapies for renal bone disease and the exact status of comments for individual cancers.
hyperparathyroidism at the time of transplantation is less Recurrence of cancer has been recorded despite disease-free periods
exceeding those suggested here. Each individual patient must be
critical than it has been. It is important to optimize control assessed individually and these intervals may be too long or too short for
of the features of renal bone disease, with special attention individual circumstances.
to attempting to normalize the calcium phosphate product Multiple myeloma needs specific consideration of prior bone marrow trans-
to minimize hyperparathyroidism, osteoporosis, and vascu- plantation.
Data from Martín-Dávila P, Fortún J, López-Vélez R, et al. Transmission of
lar calcification after transplantation. tropical and geographically restricted infections during solid-organ trans-
plantation. Clin Microbiol Rev. 2008;21(1):60–96.
GASTROINTESTINAL TRACT
Perforation of a peptic ulcer has led to many transplant avoidance of steroids combined with a proton pump inhibi-
recipient deaths in the era of high corticosteroid use and tor to prevent peptic ulceration, despite the potential for
before routine introduction of H2 receptor blockers and then interaction with immunosuppressant drug absorption.82
proton pump inhibitors after transplantation. The incidence Gastroesophageal reflux, malabsorption syndromes, celiac
of untreated Helicobacter pylori/peptic ulcer disease is now disease, diverticulosis, and cholelithiasis may all present
quite low and many units use either low dose or complete issues for specific consideration in individual patients.
4 • The Recipient of a Renal Transplant 61

Adjusted for recipient age and gender Adjusted for recipient age and gender
1.00 1.00

0.75 0.75

0.50 0.50

No DM, DD, K only


0.25 0.25 No DM, LD, K only DM1, LD, K only
DM1, DD, K only DM2, DD, K only
DM1, LD, SPK only DM2, LD, K only
0 0
0 2 4 6 8 10 0 2 4 6 8 10
A Years B Years
Fig. 4.4 The outcome of renal transplantation based on diabetes status, type of kidney, and presence of a simultaneous pancreas transplant. (A) Kidney
graft survival. (B) Patient survival. DM1, type 1 diabetes mellitus; DM2, type 2 diabetes mellitus; LD, living donor transplant; DD, deceased donor trans-
plant; K, kidney alone; SPK, simultaneous pancreas kidney. (Data courtesy Australian National Pancreas Transplant and ANZDATA Registries.)

DIABETES
consideration of islet transplantation, before, after, or with a
Recipients with both type 1 and 2 diabetes require special simultaneous kidney, remains the subject of formal clinical
consideration. Transplantation rates of diabetic patients trials in a limited number of centers globally. Although the
fluctuate markedly by era and by the approach of the trans- results in those specialized centers are encouraging there is
plant program to this large group of high-risk patients83 still insufficient evidence to lead to widespread adoption of
and the development and availability of simultaneous pan- islet transplants outside of clinical trials.86
creas and kidney transplantation (SPK).84
Type 2 Diabetes Mellitus
Type 1 Diabetes Mellitus Transplantation of the majority of patients with end-stage
The first decision for patients with type 1 diabetes is renal failure resulting from type 2 diabetes represents a chal-
whether or not to seek a simultaneous kidney and pancreas lenge to both surgical and medical expertise. The epidemic
(SPK) transplant. In countries where this expertise is avail- of type 2 diabetes that is sweeping both the developed and
able the two options that provide the best patient survival developing world has led to more than threefold increases
are preemptive living related renal transplantation and in the number of people commencing renal replacement
SPK transplantation (Fig. 4.4). Acceptance criteria for SPK therapy.87 The disease is treacherous for both patients and
transplantation usually include a rather stricter age cut-off physicians especially because of the effect of ischemic heart
than for kidney transplants and almost all units use rou- disease, and to exacerbation of the clinical effect of comor-
tine cardiac catheterization for cardiac screening. Approxi- bid peripheral vascular disease.88 Only a small proportion
mately half of the type 1 recipients with end-stage renal of type 2 diabetics are suitable for transplantation because
failure prove suitable for SPK, which compares favorably of the effect of age, obesity, and these comorbid conditions,
with living donor kidney transplantation for both patient and transplant units need to have specific policies for evalu-
and kidney outcomes in the first 10 years.84 Selection of ation of cardiac and vascular disease of those deemed suit-
patients for SPK transplants is focused even more on vas- able otherwise. Obesity is being tackled in some centers by
cular and cardiac operative risks, but is otherwise similar to pretransplant bariatric surgery, with some success in moti-
selection for kidney transplantation. The procedure is more vated patients.89 A small minority of type 2 diabetics may
demanding on both surgeon and patient, it takes longer, be helped by SPK transplantation, but this is not widely
and involves the additional risk of pancreas exocrine drain- adopted as a therapy.90
age either into the bladder or, more commonly, into the
bowel. Postoperative recovery takes longer because of the RENAL DISEASE
ileus induced by the bowel surgery and immunosuppres-
sion is on the whole more intense than for a simple kidney The range of underlying renal diseases of patients on dialy-
transplant. Against these issues, the patient must set the sis and those accepted for transplant waiting lists are simi-
benefits of good glucose control without exogenous insulin lar, because few diseases actually prevent successful renal
administration, reduced long-term complications of diabe- transplantation. The exceptions to this rule are from recur-
tes, and improved survival compared with a deceased donor rent disease in primary oxalosis and in the presence of anti-
kidney alone.85 glomerular basement membrane antibodies in Goodpasture
Detailed consideration of SPK transplantation is beyond syndrome.
the scope of this chapter, but it is undoubtedly a good solu- The causes of renal failure in Australian patients com-
tion for type 1 diabetics without severe cardiac disease mencing dialysis and those receiving a renal transplant are
and suitable for kidney transplantation (see Chapter 36). shown in Table 4.3. These data demonstrate the skewed
The role of islet transplantation is still evolving such that distribution of proportions of each type of disease in the
62 Kidney Transplantation: Principles and Practice

TABLE 4.3 Causes of Renal Failure in Patients TABLE 4.4 The Risks of Recurrence of Renal Disease
Commencing End-Stage Renal Disease Therapy and after Transplantation and the Risks of Graft Loss as a
Receiving a Renal Transplant in Australia, 2010 Result of Recurrence, Derived from Literature Review
Dialysis Transplant 10-Year Risk of
Diagnosis Patients (%) Recipients (%) Risk of Recur- Graft Loss From
Disease rence (%) Recurrence (%)
Glomerulonephritis 20 42.5
Analgesic nephropathy 1 0.3 GLOMERULONEPHRITIS
Polycystic kidney 6 12.5 Focal segmental sclerosis 20–30 8–15
Reflux nephropathy 2 7.4 IgA nephropathy 40–50 5–15
Hypertensive nephropathy 14 7.3 Henoch-Schoenlien purpura 10–20 5–10
Diabetes mellitus 37 13 Mesangiocapillary type I 20–30 10–15
Miscellaneous 13 13 Mesangiocapillary type II 80–90 5–10
Unknown 7 4 Membranous 10–20 10–25
Hemolytic uremic syndrome 10–30 10–15
Data courtesy ANZDATA Registry, Adelaide, SA, Australia. ANCA +ve vasculitis 10–15 5
Pauci-immune 10–20 5–10
Goodpasture syndrome (Ab +ve) 100 80
transplant population, especially noting the underrepre- Systemic lupus erythematosus 1 1
sentation of type 2 diabetes. METABOLIC AND OTHER DISEASES
Diabetic nephropathy 100 Low
Recurrent Renal Disease Amyloidosis 30 Low
Glomerulonephritis. Recurrence of glomerulonephri- Oxalosis 90–100 80
Cystinosis 0 0
tis (GN) in the renal transplant is an issue that needs Fabry disease 100 0
routine discussion with patients who have a diagnosis of Alport syndromea 3–4 2
focal and segmental glomerular sclerosis, IgA nephropa- Light chain nephropathy 10–25 10–30
thy, and to a lesser extent, other immune-mediated glo- Mixed essential cryoglobulinemia 50 40
merular diseases. It is important to distinguish between Scleroderma 20 5–10
the risk of recurrence and the prognosis of the graft with aTherisk of de novo antiglomerular basement membrane antibody-medi-
recurrence. A seminal analysis of the ANZDATA data- ated Goodpasture syndrome.
base demonstrated GN recurrence that is a significant ANCA, antineutrophil cytoplasmic antibodies.
cause of late graft loss, causing twice as many losses over
a 10-year period as acute rejection, but half as many as among the commonest of recurrent diseases, but is generally
chronic allograft nephropathy or death with a functioning slow to cause renal impairment and graft loss.93 It is more
graft.91 There have been many attempts, over the years, to common after living related donor grafts, but recurrence
summarize the risks for different diseases,92 and a further does not seem to affect early or medium term graft survival,
attempt is shown in Table 4.4 in which the risks of disease though use of steroid-free regimens may increase recurrence
recurrence and graft failure are presented from general lit- rates.97 Assessment of the family donor, however, needs
erature review. to include consideration of the possibility that IgA may be
familial disease and thus also affect the potential donor.
Focal Segmental Glomerulosclerosis. Recurrence of
primary focal segmental glomerulosclerosis (FSGS) is one of Thrombotic Microangiopathies. There have been sub-
the more difficult issues that must be addressed by trans- stantial advances in the understanding of a range of diseases
plant units. Risk factors for recurrence include young age that produce hemolytic uremic syndromes.98 Definition
of the recipient, the duration of native disease from onset to of typical and atypical syndromes and assessment of the
development of end-stage renal failure, mesangial prolifera- genetic complement defects that lead to the latter disease
tive pathology, and the possibility that the risk is higher in have been transformative. The option of posttransplant
related donor grafts.93,94 There is a very high risk of recur- treatment of such patients with eculizumab has released
rence in a second graft after loss of the first graft from FSGS, these patients from a life of failed transplants and dialysis.99
questioning the wisdom of retransplantation under those
circumstances. The disease may result from a circulating Membranous Nephropathy. Membranous GN may occur
glomerular permeability factor, encouraging use of plasma as either de novo or recurrent disease after transplantation.
exchange to control disease.95 A number of interventions It is becoming clear that de novo appearance of membra-
have been designed using various combinations of steroids, nous histology is related to chronic alloimmune antibody-
plasma exchange, cyclosporin, intravenous immunoglobu- mediated rejection, but that autoantibodies specific for the
lin, and rituximab.92 Therapy is certainly justified despite phospholipase A2 receptor are present in the majority of
the absence of results from well powered randomized clini- patients with recurrent membranous GN.100
cal trials.92,96
Mesangiocapillary Glomerulonephritis. Type I, II, and
IgA Nephropathy. IgA GN is a common disease in most III mesangiocapillary GN are all uncommon diseases with
countries, accounting for a relatively high proportion of high recurrence rates after transplantation.94,101 Evidence
end-stage renal failure. Recurrence rates are high, especially that recurrence of type III—dense deposit disease—may be
if sought in renal biopsies after transplantation using specific treated by eculizumab offers hope for what is otherwise a
identification of IgA deposits in the glomeruli. IgA is thus disease conferring a high risk of graft failure.102
4 • The Recipient of a Renal Transplant 63

Anti–Glomerular Basement Membrane (GBM) Disease. bladder function based on careful history taking and inves-
There is very little recent experience of recurrence of Good- tigation with urodynamic studies. More subtle problems
pasture syndrome after transplantation because of the early that may be encountered include asymptomatic prostatic
and convincing reports of recurrence in the presence of enlargement in an anuric dialysis dependent patient and
circulating antibody and advice to await clearance before the very small capacity bladder that will be encountered in
transplantation.103 It is thus essential to ensure a negative long-term dialysis patients who have been anuric for many
anti-GBM antibody test before transplantation. years. Creation of alternative bladder conduits is less popu-
Patients with Alport syndrome have abnormal base- lar than in the past because of the morbidity of the surgical
ment membrane antigens and they have been reported to procedures required. Indwelling or suprapubic catheters
develop antibody to the normal basement membrane of a followed by prostatectomy or self-catheterization are the
transplanted kidney, mimicking anti–glomerular basement standard approaches today for the majority of patients with
membrane (anti-GBM) disease in a small percentage of bladder dysfunction.112
patients but without the same level of destructive glomeru-
lar disease.104 Reflux Nephropathy. Recurrent urinary tract infection
and reflux nephropathy seldom lead to life-threatening sep-
Recurrent Vasculitis. Antineutrophil cytoplasmic anti- ticemia before transplantation, but when the pretransplant
bodies (ANCA) were discounted as a cause of recurrent experience of an individual demonstrates otherwise, bilat-
crescentic GN in 127 patients in whom the incidence of eral nephrectomy can be justified if antibiotic prophylaxis
recurrence was 17%, but with no association with presence fails to ameliorate the risk. Recurrent urinary sepsis is much
of ANCA.105 Current data support transplantation of this more common after transplantation despite prophylactic
group of patients without the need to resolve circulating measures and may threaten both the graft and the patient.
antibody levels, but most will delay transplantation until Bilateral native nephrectomy thus becomes the lesser risk
vasculitis is quiescent. Recurrence is possibly more frequent in a few patients after transplantation.
with proteinase 3 (PR3) ANCA than with mycloperoxidase
(MPO) ANCA and while treated traditionally with cyclo- Polycystic Kidney Disease. The size of polycystic kidneys
phosphamide, intensification of immunosuppression with must be evaluated before transplant surgery, preferably by
or without rituximab is generally advised.106,107 the surgeon who will be implanting the new kidney. CT
provides an excellent view of the anatomic challenge that
Hereditary Metabolic Disorders. Primary oxalosis has a will face the surgeon when the patient is horizontal on the
high recurrence rate after transplantation and is now best operating table, but will underestimate the space avail-
treated by combined kidney and liver transplantation, cor- able for the transplant when the patient stands up. Uni-
recting the metabolic abnormality simultaneously. Crystal lateral nephrectomy may be needed between the onset of
nephropathy also occurs after long-term high-dose vita- dialysis therapy and a renal transplant, precluding preemp-
min C administration in a dialysis dependent patient and in tive transplantation, though concurrent transplant and
the inherited disorder adenine phosphoribosyl transferase nephrectomy is also advocated.113
(APRT) deficiency, which can be treated successfully with
lifelong allopurinol.108 Coagulation Disorders
Fabry disease and cystinosis are both inherited enzyme Hemorrhage during the transplant and coagulation of the
deficiencies that cause renal disease through accumulated graft or other vital vascular conduit after the operation
glycosphingolipid and cystine, respectively. The former require careful prediction and management. Coagulation
leads to recurrent disease in the transplant but the latter disorders and the risk of thrombosis are much more predict-
only to extrarenal deposition of cystine. Both are, to a cer- able today through screening tests (Box 4.2). Use of hepa-
tain extent, treatable and recurrent diseases that should rin starting soon after transplantation in those identified as
be preventable with recombinant alpha-galactosidase having a possible thrombotic tendency seems to reduce the
A enzyme replacement and oral analogs of cysteamine, risk of thrombosis.89
respectively.109,110 The risk of hemorrhage is usually easily identified from
Tuberous sclerosis, although not leading to recurrent the medical history and from a careful review of the medi-
disease, deserves special consideration because of the high cation list. Iatrogenic hemorrhage is much more common
lifetime risk of developing renal cell carcinoma in the native than inherited disorder such as hemophilia, especially with
kidneys. The risk of tumor can be managed either by bilat- the widespread use of anticoagulation for atrial fibrilla-
eral nephrectomy or through regular screening by CT. The tion and after vascular stenting. Each transplant unit thus
known responsiveness of the angiomyolipomata of tuberous requires a protocol for the rapid reversal of anticoagula-
sclerosis to the mammalian target of rapamycin (mTOR) tion, usually involving small doses of vitamin K, together
inhibitors is significant and warrants selection of one or with fresh frozen plasma replacement (Box 4.3). Double
other of these agents in the immunosuppression protocol.111 antiplatelet anticoagulation for patients with cardiac or
dialysis fistula stents has led to substantial postoperative
Urogenital Tract Abnormalities bleeding problems and most transplant units regard this
Bladder. Pretransplant recognition of the patient with combination as a contraindication to listing. The advent of
bladder dysfunction is important to avoid immediate prob- new direct oral anticoagulants has complicated the situa-
lems during and after surgery. Patients with the triad syn- tion considerably. Because of the renal clearance of these
drome or other congenital obstructive uropathy, spina agents (dabigatran, rivaroxaban, apixaban, and edoxaban)
bifida, and diabetes are at easily recognizable risk of poor their use in dialysis dependent patients and those suitable
64 Kidney Transplantation: Principles and Practice

and other problems. The data now suggest that low BMI is
BOX 4.2 Risks of Thrombosis and the greater concern, with malnourished patients having
Coagulation Disorder higher mortality rates. The problem that both physician
Medication
and patient face is the task of reducing weight in the very
obese and increasing weight in the malnourished before
Dual antiplatelet therapy transplantation. In patients treated by peritoneal dialysis it
Aspirin
is especially hard to change the body habitus derived from
Warfarin
Heparin
the high carbohydrate intake from peritoneal dialysis fluids,
Low molecular weight heparins such that a switch to hemodialysis may be the only option.
Lifestyle changes are sometimes achievable when renal
Coagulation transplantation is the goal but bariatric surgery in a deter-
Medical history of thromboses mined individual provides the best chance of weight loss.89
Coagulation tests: prothrombin time, activated partial thrombo-
plastin time Psychosocial Factors
Factor V Leiden, protein C, protein S, antithrombin III deficiency Smoking provides serious cardiovascular and pulmonary
Antiphospholipid antibodies risks before, during, and after transplant surgery and is
Full blood count: polycythemia
heavily discouraged by all programs.118 The unanswered
Hemostasis question remains whether or not it is appropriate to trans-
Medication history (warfarin, aspirin, clopidogrel, dipyridamole) plant patients who continue to smoke. Many in both the lay
Medical history of bleeding and professional communities argue that it is not appro-
Medical history of liver disease priate for the community to provide access to the scarce
Coagulation tests: skin bleeding time, activated partial thrombo- resource of a donated kidney if the patient continues to self
plastin time, and congenital factor deficiencies harm through smoking.
Recreational drug and alcohol abuse require careful eval-
uation.119 It is important to wean patients from drug depen-
dency, testing compliance and assessing the possibility of
BOX 4.3 Anticoagulant Reversal Protocol recent hepatitis or HIV infection before activating them
Before Transplant Surgery on the transplant waiting list. Psychiatric evaluation and
treatment are often essential components of preparation for
A patient on warfarin who requires surgery within the next 8 transplantation in drug dependency, but may be rejected or
hours should receive: unsuccessful. Families may be the harshest critics of such
1. 1 unit of fresh frozen plasma (FFP) individuals and thus not offer living kidney donation, leav-
2. 5 mg intravenous vitamin K ing transplant programs with the decision of whether or
3. Prothrombinex (Human Prothrombin Complex)—dose not it is appropriate to provide access to a deceased donor
adjusted for International Normalized Ratio (INR) and patient kidney. Documented abstinence for 6 months and determi-
weight nation of likely compliance after transplantation provide a
□ if INR 2–3.9: 25 units/kg
nonjudgmental approach to resolving this dilemma but are
□ 4–5.9: 35 units/kg

□ >6.0: 50 units/kg
in themselves complex assessments.
□ Prothrombinex (1000 unit/vial)—calculate to the nearest
Alcoholism may be well hidden and needs an enquir-
1000 units ing and suspicious clinical evaluation including an under-
4. Check prothrombin time (PT)/activated partial thromboplastin standing of the effect on the liver and the psychological
time (APTT) before surgery and 4 to 8 hours later if surgery state of the patient. Compliance and reliability for follow-up
delayed. after transplantation are important factors that will influ-
5. If surgery is to occur >8 hours from reversal, FFP is not required ence patient and graft outcomes.
but PT/APTT needs to be rechecked before surgery to confirm Mental illness requires formal evaluation and treat-
adequate reversal. ment, with the important additional need to determine the
patient’s ability to understand and consent to renal trans-
plantation. There is no substitute for an independent psy-
for kidney transplantation is rare. Only dabigatran has a chiatric evaluation of fitness to consent and ensure optimal
method for rapid though incomplete reversal, using the pre- and postoperative psychiatric treatment.120
expensive monoclonal antibody fragment idarucizumab.
Experience with this agent before transplantation is cur- Sensitization and Transfusion Status
rently very limited and not recommended.114 Good knowledge of the anti–human leukocyte antigen
(HLA) antibody status and both the patient and donor’s
Obesity HLA type is critical to success but outside the scope of this
Increasing BMI is not associated with increased risks of chapter (see Chapter 22).
death or graft loss but is associated with new onset post-
transplant diabetes mellitus and in obese children is asso- Previous Transplantation
ciated with graft failure.115–117 The depth of abdominal Previous renal transplants provide both visible and invis-
fat certainly challenges the surgeon and without careful ible barriers to the next transplant, both of which need to be
management can lead to increased risk of wound infection considered carefully. Retransplantation is usually even less
4 • The Recipient of a Renal Transplant 65

successful than the first transplant procedure and requires consent to receive offers for donors outside of the standard
careful assessment of the patient’s immunologic reactivity. donor criteria. It is unreasonable to expect the patient
The total number of individuals with chronic graft loss is to consent meaningfully in the middle of the night to a
increasing in most countries, as is the number of patients complex offer of an older donor kidney or one potentially
with nonrenal organ transplants requiring a renal trans- infected with hepatitis B or C. Thus many programs have
plant as a result of nephrotoxicity or other cause of chronic special categories of prior consent, which permit individu-
renal failure.121 als to choose to receive a kidney from an extended criteria
The clinician’s decision to offer retransplantation is donor,124 an old donor,125 or one infected with the same
sometimes hard. Should a patient who has lost their first virus that they carry—be it hepatitis B or C, or HIV126 with
graft because of noncompliance with medication be offered realistic options for posttransplant treatment of hepatitis C
the chance to destroy another priceless donation in the now available.58
same way? Perhaps the older, wiser, and now experienced
individual will be a model of compliance the second time TO UNDERGO ELECTIVE LIVING DONOR
around? Assessment of the medical suitability for trans- TRANSPLANTATION
plantation needs to be just as rigorous the second time as
it was the first time, noting especially that infective, malig- The assessment of the recipient of a living donor graft is,
nant, and cardiovascular diseases are all more common in by contrast to the deceased donor recipient, a more orderly
the previously transplanted than in the dialysis patient. and planned affair. Despite this the focus is often more on
Opinion and practices have varied with respect to the the suitability of the donor and less on the recipient. Ensur-
management of a failed graft,122 but many units favor early ing that the donor and recipient are assessed by different
graft nephrectomy.123 Transplant nephrectomy is a reason- nephrologists and different surgeons brings attention back
ably low-risk procedure that removes an ongoing source of to the recipient. Providing that there is good communica-
foreign antigenic stimulation and allows for discontinu- tion between the two teams, it is possible to manage the
ation of immunosuppression without risk of incurring a interface between donor and recipient issues smoothly and
debilitating response. Nephrectomy is always sensible in effectively. It is just as important for the donor to under-
cases of early acute graft failure from whatever cause. stand the risks of a poor outcome in the recipient as it is
for the recipient to understand them. A donor unaware of
the possibility of recurrent disease in a patient with FSGS,
Preparation for Transplantation for example, will reasonably ask why he or she was not
informed before the donation. The risk of death at opera-
TO JOIN AND REMAIN ON THE DECEASED tion for a particular recipient may be acceptable to them,
but not to the donor who may be unprepared for that pos-
DONOR WAITING LIST
sibility. The opposite situation may also occur. A donor may
The majority of this chapter has defined the issues of impor- undertake risky behavior, such as intravenous drug use or
tance for assessment, selection, and preparation of candi- unprotected high-risk intercourse, which the recipient may
dates for the transplant waiting lists. Acceptance should know more about than either the donor or recipient’s medi-
then lead to histocompatibility testing and entry on to the cal teams. Understanding the level and nature of the risk is
transplant waiting list. The initial evaluation may be per- paramount for the recipient.
formed many years before the kidney becomes available
and thus repeated reassessment is also required over the TO UNDERGO DECEASED DONOR
years that a patient may be on the waiting list. Compliance TRANSPLANTATION
with the needs of the transplant waiting list and, in partic-
ular, providing a current blood sample for crossmatching A transplant team will receive the news that a kidney is
may sort out the willing and motivated patients from the available for a particular patient only a few hours before
noncompliant. Most programs maintain serum screening the operation needs to be performed. All too often, the allo-
protocols to identify patients who are sensitized, both to cation takes place in the middle of the night and the news
predict the chances of receiving a transplant and to better is passed through a transplant coordinator and junior doc-
evaluate donor T and B cell crossmatch results obtained tors. The patient and their family will probably not be in
after working hours. contact with the individuals who have assessed them and
Maintaining a current record of clinical events and rel- who care for their dialysis. Their questions and uncertain-
evant serology for infectious disease (HIV, hepatitis B, and ties will be carried away in a rush of investigations includ-
hepatitis C especially) should be the province of the dialy- ing a chest x-ray, an electrocardiogram, routine blood tests,
sis unit responsible for the patient’s treatment. Ensuring bowel preparation, shower, anesthetic evaluation, immu-
that this data is available to the transplant program in the nosuppressive medication, and perhaps also preoperative
middle of the night remains challenging and is likely to fail hemodialysis. Despite this rush of activity, the pressure to
without a good information system. In the final analysis reduce cold ischemia time for the kidney and to meet the
there is little alternative but to ensure that those who are deadlines and timetables of operating suites tend to over-
managing the patient on a daily basis are always contacted shadow the needs for discussion and informed consent.
when a kidney offer is made. This emphasizes the need for full education and informa-
An additional issue that has been raised by the dearth tion during the workup for acceptance onto the transplant
of donors and the burgeoning waiting lists has been prior waiting list.
66 Kidney Transplantation: Principles and Practice

References 24. Kasiske BL, Zeier MG, Chapman JR, et al. Best practice guidelines
KDIGO clinical practice guideline for the care of kidney transplant
1. Cosio FG, Alamir A, Yim S, et al. Patient survival after renal recipients: a summary. Kidney Int 2010;77(4):299–311.
transplantation: I. The impact of dialysis pre-transplant. Kid Int 25. US Department of Health & Human Services. Organ Procurement
1998;53:767. and Transplantation Network (OPTN): Kidney Donor Profile Index
2. Ashby VB, Leichtman AB, Rees MA, et al. A kidney graft survival calculator. Available online at: https://optn.transplant.hrsa.gov/re
calculator that accounts for mismatches in age, sex, HLA, and body sources/allocation-calculators.
size. Clin J Am Soc Nephrol 2017;12(7):1148–60. 26. Kennedy SE, Mackie FE, Rosenberg AR, et al. Waiting time and
3. Grace BS, Clayton PA, Cass A, McDonald SP. Transplantation rates outcome of kidney transplantation in adolescents. Transplantation
for living-but not deceased-donor kidneys vary with socioeconomic 2006;82(8):1046.
status in Australia. Kidney Int 2012;15:336–41. 27. Williams RC, Opelz G, Weil EJ, McGarvey CJ, Chakkera HA. The
4. World Health Organization. http://www.who.int/transplantation/ risk of transplant failure with HLA mismatch in first adult kidney
knowledgebase/en/ [accessed 06.12.17]. allografts 2: living donors, summary, guide. Transplant Direct
5. Howell M, Wong G, Rose J, Tong A, Craig JC, Howard K. Patient pref- 2017;3(5):e152.
erences for outcomes after kidney transplantation: a best-worst scal- 28. Delmonico F, Council of the Transplantation Society. A report of the
ing survey. Transplantation 2017;101(11):2765–73. Amsterdam Forum on the care of the live kidney donor: data and
6. Rosenberger J, van Dijk JP, Nagyova I, et al. Predictors of perceived medical guidelines. Transplantation 2005;79(Suppl. 6):S53.
health status in patients after kidney transplantation. Transplanta- 29. Rodriguez JR, Cornell DL, Lin JK, et al. Increasing live donor kidney
tion 2006;81(9):1306. transplantation: a randomized controlled trial of a home-based edu-
7. Wolfe RA, Ashby VB, Milford EL, et al. Comparison of mortality in cational intervention. Am J Transplant 2007;7(2):394.
all patients on dialysis, patients on dialysis awaiting transplanta- 30. Lentine KL, Costa SP, Weir MR, et al. Cardiac disease evaluation and
tion and recipients of a first cadaveric transplant. New Engl J Med management among kidney and liver transplantation candidates.
1999;341:1725. Circulation 2012;126:617–63.
8. Merion RM, Ashby VB, Wolfe RA, et al. Deceased-donor charac- 31. Abramowicz D, Cochat P, Claas F, et al. The European Renal Best
teristics and the survival benefit of kidney transplantation. JAMA Practice (ERBP) transplantation guideline. Nephrol Dial Transplant
2005;294(21):2726. 2013;28(2):ii1–71.
9. Mathes T, Großpietsch K, Neugebauer EAM, Pieper D. Interventions 32. Cosio FG, Falkenhain MF, Pesavento TE, et al. Patient survival after
to increase adherence in patients taking immunosuppressive drugs renal transplantation II: the impact of smoking. Clin Transplant
after kidney transplantation: a systematic review of controlled trials. 1999;13:336.
Syst Rev 2017;6(1):236. 33. Feringa HH, Bax JJ, Schouten O, et al. Ischemic heart disease in renal
10. Pérez-Sáez MJ, Arcos E, Comas J, Crespo M, Lloveras J, Pascual J. transplant candidates: towards non-invasive approaches for preop-
Catalan Renal Registry Committee: survival benefit from kidney erative risk stratification. Eur J Echocardiogr 2005;6(5):313.
transplantation using kidneys from deceased donors aged ≥75 34. Manske CL, Wang Y, Rector T, et al. Coronary revascularisation in
years: a time-dependent analysis. Am J Transplant 2016;16(9): insulin-dependent diabetic patients with chronic renal failure. Lan-
2724–33. cet 1992;340(8826):998.
11. Clayton PA, McDonald SP, Snyder JJ, Salkowski N, Chadban SJ. 35. Sharma R, Pellerin D, Gaze DC, et al. Dobutamine stress echocar-
External validation of the estimated posttransplant survival score diography and cardiac troponin T for the detection of significant
for allocation of deceased donor kidneys in the United States. Am J coronary artery disease and predicting outcome in renal transplant
Transplant 2014;14(8):1922–6. candidates. Eur J Echocardiogr 2005;6(5):327.
12. Meier-Kriesche HU, Cibrik DM, Ojo AO, et al. Interaction between 36. Cortigiani L, Desideri A, Gigli G, et al. Clinical, resting echo and
donor and recipient age in determining the risk of chronic renal dipyridamole stress echocardiography findings for the screening of
allograft failure. J Am Geriatr Soc 2002;50(1):14. renal transplant candidates. Int J Cardiol 2005;103(2):168.
13. Ojo AO. Expanded criteria donors: process and outcomes. Semin Dial 37. Norby GE, Günther A, Mjøen G, et al. Prevalence and risk factors
2005;18(6):463. for coronary artery calcification following kidney transplanta-
14. Lim WH, Russ GR, McDonald SP. Comparable transplant outcomes tion for systemic lupus erythematosus. Rheumatology (Oxford)
between local and shipped deceased-donor kidneys in Austra- 2011;50(9):1659–64.
lia: analysis of Australia and New Zealand Dialysis and Trans- 38. Wang LW, Fahim MA, Hayen A, et al. Cardiac testing for coronary
plant Registry 1992–2007. Nephrology (Carlton) 2010;15(1): artery disease in potential kidney transplant recipients: a systematic
124–32. review of test accuracy studies. Am J Kidney Dis 2011;57:476–87.
15. Chapman JR, O’Connell PJ, Nankivell BJ. Chronic renal allograft dys- 39. Borentain M, Le Feuvre C, Helft G, et al. Long-term outcome after
function. J Amer Soc Nephrol 2005;16(10):3015. coronary angioplasty in renal transplant and hemodialysis patients.
16. http://www.anzdata.org.au/anzdata/AnzdataReport/39thReport/c J Interv Cardiol 2005;18(5):331.
03_deaths_v2.1_20170418.pdf [accessed 06.12.17]. 40. Groetzner J, Kaczmarek I, Mueller M, et al. Freedom from graft ves-
17. h ttp://www.transplant-observatory.org/data-charts-and- sel disease in heart and combined heart- and kidney-transplanted
tables/chart/ [accessed 06.12.17]. patients treated with tacrolimus-based immunosuppression. J Heart
18. Muralidharan A, White S. The need for kidney transplantation in Lung Transplant 2005;24(11):1787.
low- and middle-income countries in 2012: an epidemiological per- 41. Nankivell BJ, Lau S-G, Chapman JR, et al. Progression of macrovas-
spective. Transplantation 2015;99(3):476–81. cular disease after transplantation. Transplantation 2000;69:574.
19. Batabyal P, Chapman JR, Wong G, Craig JC, Tong A. Clinical prac- 42. Wong G, Howard K, Chapman JR, et al. Comparative survival and
tice guidelines on wait-listing for kidney transplantation: consistent economic benefits of deceased donor kidney transplantation and
and equitable? Transplantation 2012;94(7):703–13. dialysis in people with varying ages and co-morbidities. PLoS One
20. McAdams-DeMarco MA, Olorundare IO, Ying H, et al. Frailty and 2012;7(1):e29591. Epub 2012 Jan 18.
postkidney transplant health-related quality of life. Transplantation 43. Eggers PW, Gohdes D, Pugh J. Nontraumatic lower limb extremity
2018;102(2):291–9. amputations in the Medicare end-stage renal disease population.
21. Rao PS, Schaubel DE, Guidinger MK, et al. A comprehensive risk Kidney Int 1999;56:1524.
quantification score for deceased donor kidneys: the Kidney Donor 44. de Mattos AM, Prather J, Olyaei AJ, et al. Cardiovascular events
Risk Index. Transplantation 2009;88:231–6. following renal transplantation: role of traditional and transplant-
22. Israni AK, Salkowski N, Gustafson S, et al. New national alloca- specific risk factors. Kidney Int 2006;70(4):757.
tion policy for deceased donor kidneys in the United States and 45. Hughes PD, Becker GJ. Screening for intracranial aneurysms in
possible effect on patient outcomes. J Am Soc Nephrol 2014;25(8): autosomal dominant polycystic kidney disease. Nephrology (Carl-
1842–8. ton) 2003;8(4):163.
23. Hart A, Gustafson SK, Skeans MA, et al. OPTN/SRTR 2015 Annual 46. Sarin Kapoor H, Kaur R, Kaur H. Anaesthesia for renal transplant
Data Report: early effects of the new kidney allocation system. Am J surgery. Acta Anaesthesiol Scand 2007;51(10):1354–67.
Transplant 2017;17(Suppl. 1):543–64.
4 • The Recipient of a Renal Transplant 67

47. Al-Efraij K, Mota L, Lunny C, Schachter M, Cook V, Johnston J. Risk tious Diseases Society of America. Clin Infect Dis 2014;59(9):
of active tuberculosis in chronic kidney disease: a systematic review e96–e138.
and meta-analysis. Int J Tuberc Lung Dis 2015;19(12):1493–9. 70. Avettand-Fenoël V, Rouzioux C, Legendre C, Canaud G. HIV
48. Ferguson TW, Tangri N, Macdonald K, et al. The diagnostic accu- infection in the native and allograft kidney: implications for
racy of tests for latent tuberculosis infection in hemodialysis management, diagnosis, and transplantation. Transplantation
patients: a systematic review and meta-analysis. Transplantation 2017;101(9):2003–8.
2015;99(5):1084–91. 71. Kotton CN, Kumar D, Caliendo AM, et al. International consensus
49. Al-Mukhaini SM, Al-Eid H, Alduraibi F, et al. Mycobacterium guidelines on the management of cytomegalovirus in solid organ
tuberculosis in solid organ transplantation: incidence before and transplantation: Transplantation Society International CMV Con-
after expanded isoniazid prophylaxis. Ann Saudi Med 2017;37(2): sensus Group. Transplantation 2010;89(7):779–95.
138–43. 72. Rodríguez-Romo R, Morales-Buenrostro LE, Reyes PA, et al. Sero-
50. Kasiske BL, Vazquez MA, Harmon WE, et al. Recommendations for prevalence of Trypanosoma cruzi in kidney transplant donors
the outpatient surveillance of renal transplant recipients: American and recipients in Mexico City. Transpl Infect Dis 2013;15(6):
Society of Transplantation. J Am Soc Nephrol 2000;4:S1. 639–44.
51. Lee J, Cho J-H, Lee JS, et al. Pretransplant hepatitis B viral infection 73. Martín-Dávila P, Fortún J, López-Vélez R, et al. Transmission of tropi-
increases risk of death after kidney transplantation: a multicenter cal and geographically restricted infections during solid-organ trans-
cohort study in Korea. Medicine (Baltimore) 2016;95(21):e3671. plantation. Clin Microbiol Rev 2008;21(1):60–96.
52. David-Neto E, Americo da Fonseca J, Jota de Paula F, et al. The 74. Vajdic CM, McDonald SP, McCredie MR, et al. Cancer incidence
impact of azathioprine on chronic viral hepatitis in renal transplan- before and after kidney transplantation. JAMA 2006;296(23):2823.
tation: a long-term single-centre, prospective study on azathioprine 75. van Leeuwen MT, Webster AC, McCredie MRE, et al. Effect of reduced
withdrawal. Transplantation 1999;68:976. immunosuppression after kidney transplant failure on risk of cancer:
53. Nadim MK, Sung RS, Davis CL, et al. Simultaneous liver–kid- population based retrospective cohort study. BMJ 2010;340:c570.
ney transplantation summit: current state and future directions. 76. Rosenberg SA. IL-2: the first effective immunotherapy for human
Am J Transplant 2012;12(11):2901–8. cancer. J Immunol 2014;192(12):5451–8.
54. Habib S, Khan K, Chiu-Hsieh H, Meister E, Rana A, Boyer T. Differen- 77. Johnson DB, Sullivan RJ, Menzies AM. Immune checkpoint inhibi-
tial simultaneous liver and kidney transplant benefit based on sever- tors in challenging populations. Cancer 2017;123(11):1904–11.
ity of liver damage at the time of transplantation. Gastroenterology 78. Williams NC, Tong A, Howard K, Chapman JR, Craig JC, Wong G.
Res 2017;10(2):106–15. Knowledge, beliefs and attitudes of kidney transplant recipients
55. Lauer GM, Walker BD. Hepatitis C virus infection. N Engl J Med regarding their risk of cancer. Nephrology (Carlton) 2012;17(3):
2001;345:41. 300–6.
56. Lubetzky M, Chun S, Joelson A, et al. Safety and efficacy of treatment 79. Chapman JR. Compliance: the patient, the doctor, and the medica-
of hepatitis C in kidney transplant recipients with directly acting tion? Transplantation 2004;77(5):782–6.
antiviral agents. Transplantation 2017;101(7):1704–10. 80. Ong SC, Gaston RS. Medical management of chronic kidney dis-
57. Saxena V, Khungar V, Verna EC, et al. Safety and efficacy of current ease in the renal transplant recipient. Curr Opin Nephrol Hypertens
direct-acting antiviral regimens in kidney and liver transplant recipi- 2015;24(6):587–93.
ents with hepatitis C: results from the HCV-TARGET study. Hepatol- 81. Ingulli EG, Mak RH. Growth in children with chronic kidney disease:
ogy 2017;66(4):1090–101. role of nutrition, growth hormone, dialysis, and steroids. Curr Opin
58. Heo NY, Mannalithara A, Kim D, Udompap P, Tan JC, Kim WR. Pediatr 2014;26(2):187–92.
Long-term patient and graft survival of kidney transplant recipi- 82. Gabardi S, Olyaei A. Evaluation of potential interactions between
ents with hepatitis C virus infection in the United States. Trans- mycophenolic acid derivatives and proton pump inhibitors. Ann
plantation 2018;102(3):454–60. Available online at: https://doi. Pharmacother 2012;46(7-8):1054–64.
org/10.1097/TP.0000000000001953. 83. White S, Chadban S. Diabetic kidney disease in Australia: cur-
59. Gane E, Lowitz E, Pugateh D, et al. Glecaprevir and pibrentasvir rent burden and future projections. Nephrology (Carlton)
in patients with HCV and severe renal impairment. N Engl J Med 2014;19(8):450–8.
2017;377:1448–55. 84. Young BY, Gill J, Huang E, et al. Living donor kidney versus simulta-
60. Sarkio S, Salmela K, Kyllönen L, Rosliakova M, Honkanen E, Halme neous pancreas-kidney transplant in type I diabetics: an analysis of
L. Complications of gallstone disease in kidney transplantation the OPTN/UNOS database. Clin J Am Soc Nephrol 2009;4(4):845–
patients. Nephrol Dial Transplant 2007;22(3):886–90. 52.
61. Genc G, Ozkaya O, Aygun C, Yakupoglu YK, Nalcacioglu H. Vac- 85. Gruessner AC, Sutherland DE, Gruessner RW. Long-term out-
cination status of children considered for renal transplants: missed come after pancreas transplantation. Curr Opin Organ Transplant
opportunities for vaccine preventable diseases. Exp Clin Transplant 2012;17(1):100–5.
2012;10(4):314–8. 86. Holmes-Walker DJ, Gunton JE, Hawthorne W, et al. Islet transplan-
62. Prelog M, Pohl M, Ermisch B, et al. Demand for evaluation of vac- tation provides superior glycemic control with less hypoglycemia
cination antibody titers in children considered for renal transplanta- compared with continuous subcutaneous insulin infusion or multi-
tion. Pediatr Transplant 2007;11(1):73–6. ple daily insulin injections. Transplantation 2017;101(6):1268–75.
63. Kruger S, Seyfarth M, Sack K, et al. Defective immune response to 87. Grace BS, Clayton P, McDonald SP. Increases in renal replacement
tetanus toxoid in haemodialysis patients and its association with therapy in Australia and New Zealand: understanding trends in dia-
diphtheria vaccination. Vaccine 1999;17:1145. betic nephropathy. Nephrology (Carlton) 2012;17(1):76–84.
64. h ttps://www.cdc.gov/meningococcal/clinical/eculizumab.html 88. Lim WH, Johnson DW, Hawley CM, Pascoe E, Wong G. Impact of
[accessed 06.12.17]. diabetes mellitus on the association of vascular disease before trans-
65. Smith KG, Isbel NM, Catton MG, et al. Suppression of the humoral plantation with long-term transplant and patient outcomes after
immune response by mycophenolate mofetil. Nephrol Dial Trans- kidney transplantation: a population cohort study. Am J Kidney Dis
plant 1998;13(1):160. 2018;71(1):102–11.
66. Pelletier SJ, Norman SP, Christensen LL, et al. Review of trans- 89. Scalea JR, Cooper M. Surgical strategies for type II diabetes. Trans-
plantation of HIV patients during the HAART era. Clin Transpl plant Rev (Orlando) 2012;26(3):177–82.
2004;63–82. 90. Gruessner AC, Laftavi MR, Pankewycz O, Gruessner RWG. Simulta-
67. Stock PG, Barin B, Murphy B, et al. Outcomes of kidney transplanta- neous pancreas and kidney transplantation: is it a treatment option
tion in HIV-infected recipients. N Engl J Med 2010;363(21):2004– for patients with type 2 diabetes mellitus? An analysis of the Interna-
14. tional Pancreas Transplant Registry. Curr Diab Rep 2017;17(6):44.
68. Muller E, Barday Z, Mendelson M, Kahn D. Renal transplantation 91. Briganti EM, Russ GR, McNeil JJ, et al. Risk of renal allograft loss from
between HIV-positive donors and recipients justified. S Afr Med J recurrent glomerulonephritis. N Engl J Med 2002;347(2):103.
2012;102(6):497–8. 92. Canaud G, Audard V, Kofman T, Lang P, Legendre C, Grimbert P.
69. Lucas GM, Ross MJ, Stock PG, et al. Clinical practice guideline for Recurrence from primary and secondary glomerulopathy after renal
the management of chronic kidney disease in patients infected with transplant. Transpl Int 2012;25(8):812–24.
HIV: 2014 update by the HIV Medicine Association of the Infec-
68 Kidney Transplantation: Principles and Practice

93. McDonald SP, Russ GR. Recurrence of IgA nephropathy among 111. Haidinger M, Werzowa J, Weichhart T, Säemann MD. Target-
renal allograft recipients from living donors is greater among those ing the dysregulated mammalian target of rapamycin pathway in
with zero HLA mismatches. Transplantation 2006;82(6):759. organ transplantation: killing 2 birds with 1 stone. Transplant Rev
94. Allen PJ, Chadban SJ, Craig JC, et al. Recurrent glomerulonephritis (Orlando) 2011;25(4):145–53.
after kidney transplantation: risk factors and allograft outcomes. 112. Cabello BR, Quicios DC, López ML, Simón RC, Charry GP, González
Kidney Int 2017;92(2):461–9. EC. The candidate for renal transplantation work up: medical, uro-
95. Doublier S, Zennaro C, Musante L, et al. Soluble CD40 ligand directly logical and oncological evaluation. Arch Esp Urol 2011;64(5):441–
alters glomerular permeability and may act as a circulating perme- 60.
ability factor in FSGS. PLoS One 2017;12(11):e0188045. 113. Tyson MD, Wisenbaugh ES, Andrews PE, Castle EP, Humphreys MR.
96. Alasfar S, Matar D, Montgomery RA, et al. Rituximab and thera- Simultaneous kidney transplantation and bilateral native nephrec-
peutic plasma exchange in recurrent focal segmental glomerulo- tomy for polycystic kidney disease. J Urol 2013;190(6):2170–4.
sclerosis postkidney transplantation. Transplantation 2018;102(3): 114. Salerno DM, Tsapepas D, Papachristos A, et al. Direct oral antico-
e115–e120. agulant considerations in solid organ transplantation: a review. Clin
97. Clayton P, McDonald S, Chadban S. Steroids and recurrent IgA Transplant 2017;31(1):1–13.
nephropathy after kidney transplantation. Am J Transplant 115. Strejar E, Molnar MK, Kovesdy CP, et al. Associations of pretrans-
2011;11(8):1645–9. plant weight and muscle mass with mortality in renal transplant
98. Rafat C, Coppo P, Fakhouri F, et al. Hemolytic and uremic syndrome recipients. Clin J Am Soc Nephrol 2011;6(6):1463–73.
and related thrombotic microangiopathies: treatment and progno- 116. Chang SH, Coates PT, McDonald SP. Effects of body mass index at
sis. Rev Med Intern 2017;38(12):833–9. transplant on outcomes of kidney transplantation. Transplantation
99. Legendre C, Sberro-Soussan R, Zuber J, et al. Eculizumab in renal 2007;84(8):981–7.
transplantation. Transplant Rev (Orlando) 2013;27(3):90–2. 117. Ladhani M, Lade S, Alexander SI, et al. Obesity in pediatric kidney
100. Debiec H, Martin L, Jouanneau C, et al. Autoantibodies specific for transplant recipients and the risks of acute rejection, graft loss and
the phospholipase A2 receptor in recurrent and de novo membra- death. Pediatr Nephrol 2017;32(8):1443–50.
nous nephropathy. Am J Transplant 2011;11(10):2144–52. 118. Bluman LG, Mosca L, Newman N, et al. Preoperative smoking habits
101. Andresdottir MB, Assmann KJM, Hoitsma AJ, et al. Renal transplan- and postoperative pulmonary complications. Chest 1998;113:883.
tation in patients with dense deposit disease: morphological char- 119. Parker R, Armstrong MJ, Corbett C, Day EJ, Neuberger JM. Alcohol
acteristics of recurrent disease and clinical outcome. Nephrol Dial and substance abuse in solid-organ transplant recipients. Trans-
Transplant 1999;14:1723. plantation 2013;96(12):1015–24.
102. McCaughan JA, O’Rourke DM, Courtney AE. Recurrent dense 120. DiMartini A, Crone C, Fireman M, Dew MA. Psychiatric
deposit disease after renal transplantation: an emerging role for aspects of organ transplantation in critical care. Crit Care Clin
complementary therapies. Am J Transplant 2012;12(4):1046–51. 2008;24(4):949–53.
103. Wilson CB, Dixon FJ. Antiglomerular basement membrane antibody 121. Srinivas TR, Stephany BR, Budev M, et al. An emerging population:
induced glomerulonephritis. Kidney Int 1973;3:74. kidney transplant candidates who are placed on the waiting list
104. Gobel J, Olbricht CJ, Offner G, et al. Kidney transplantation in Alport’s after liver, heart, and lung transplantation. Clin J Am Soc Nephrol
syndrome: long-term outcome and allograft anti-GBM nephritis. 2010;5(10):1881–6.
Clin Nephrol 1992;38:299. 122. Bennett WM. The failed renal transplant: in or out? Semin Dial
105. Nachman PH, Segelmark M, Westman K, et al. Recurrent ANCA- 2005;18(3):188.
associated small vessel vasculitis after transplantation: a pooled 123. Ayus JC, Achinger SG, Lee S, Sayegh MH, Go AS. Transplant
analysis. Kidney Int 1999;56:1544. nephrectomy improves survival following a failed renal allograft.
106. Geetha D, Eirin A, True K, et al. Renal transplantation in antineutro- J Am Soc Nephrol 2010;21(2):374–80.
phil cytoplasmic antibody-associated vasculitis: a multicenter expe- 124. Grams ME, Womer KL, Ugarte RM, Desai NM, Montgomery RA,
rience. Transplantation 2011;91(12):1370–5. Segev DL. Listing for expanded criteria donor kidneys in older adults
107. Geetha D, Lee SM, Shah S, Rahman HM. Relevance of ANCA positiv- and those with predicted benefit. Am J Transplant 2010;10(4):
ity at the time of renal transplantation in ANCA associated vasculi- 802–9.
tis. J Nephrol 2017;30(1):147–53. 125. Giessing M, Fuller TF, Friedersdorff F, et al. Outcomes of transplant-
108. Bollée G, Cochat P, Daudon M. Recurrence of crystalline nephropa- ing deceased-donor kidneys between elderly donors and recipients.
thy after kidney transplantation in APRT deficiency and primary J Am Soc Nephrol 2009;20(1):37–40.
hyperoxaluria. Can J Kidney Health Dis 2015;2:31. 126. Carbone M, Cockwell P, Neuberger J. Hepatitis C and kidney trans-
109. Lidove O, Joly D, Barbey F, et al. Clinical results of enzyme replace- plantation. Int J Nephrol 2011;2011:593291.
ment therapy in Fabry disease: a comprehensive review of literature.
Int J Clin Pract 2007;61(2):293.
110. Markello TC, Bernardini IM, Gahi WA. Improved renal function
in children with cystinosis treated with cysteamine. N Engl J Med
1993;328:1157.
5 Access for Renal
Replacement Therapy
JAMES P. HUNTER and JAMES A. GILBERT

CHAPTER OUTLINE Introduction Arteriovenous Fistula Surveillance and


Vascular Access Catheters Maintenance
Temporary Dialysis Catheters Advances in Vascular Access
Permanent Dialysis Catheters HeRO (Hemodialysis Reliable Outflow) Graft
Complications of Hemodialysis Catheters Surfacer Inside-Out Catheter System
Catheter Dysfunction Endovascular Arteriovenous Fistula
Catheter-Related Central Vein Stenosis Peritoneal Dialysis
Central Vein Occlusion Acute Peritoneal Dialysis
Infection Peritoneal Dialysis Delivery Systems and
Fistulas and Synthetic Grafts Catheters
Planning and Timing of Vascular Access Catheter Selection
Preoperative Assessment Catheter Insertion
Anesthesia Complications Associated With Peritoneal
Dialysis Catheters
Autogenous Arteriovenous Fistulas
Bleeding
Wrist Fistula
Pain
Elbow Fistulas
Cuff Extrusion
Arteriovenous Grafts
Catheter Obstruction
Complications of Arteriovenous Fistulas and
Grafts Pericatheter Leak
Hemorrhage Hernias
Stenosis Exit-Site and Tunnel Infections
Thrombosis Peritoneal Dialysis Peritonitis
Infection Encapsulating Peritoneal Sclerosis
Aneurysm Formation Renal Transplant Issues With Peritoneal
Dialysis
Arteriovenous Access Ischemic Steal
Syndrome Conclusion

Introduction in recent years and the leading cause of kidney disease is


now diabetes, which accounts for 30% of all cases.1 This is
Worldwide the population of patients with chronic kidney important to recognize, because many of these patients are
disease (CKD) is expanding, leading to increasing numbers high-risk surgical candidates and are particularly challeng-
of patients hitting end-stage renal failure (ESRF) and requir- ing with regard to creating dialysis access because of small,
ing renal replacement therapy (RRT). In the UK the inci- diseased vessels, a paucity of good-caliber veins, and higher
dence of new dialysis starters increased from 115 per million infection rates.
population (pmp) in 2014 to 120 pmp in 2015, resulting in Long-term access for RRT through the creation of an
7814 new patients initiating RRT.1 The median age of these arteriovenous fistula (AVF), placement of an arteriovenous
new dialysis starters was 64.4 years, highlighting the fact graft (AVG), or peritoneal dialysis (PD) catheter requires
that a significant proportion of the UK dialysis population the input of multiple dedicated access specialists. Over the
are elderly patients, many of whom have multiple comor- past decade this has expanded to include nephrologists, sur-
bid conditions, such as diabetes, hypertension, ischemic geons, radiologists, and nurse specialists who work closely
heart disease, and obesity. There has been a paradigm shift as a multidisciplinary team to provide timely access creation
69
70 Kidney Transplantation: Principles and Practice

and ongoing access maintenance for each individual renal venous catheter insertion are displayed in Box 5.1. Per-
patient. There is a growing recognition that the approach manent catheters are always tunneled, dual-lumen,
to access creation should be increasingly bespoke for each large-diameter (14 to 16 French) polyurethane catheters
individual patient. The historical tradition of starting dial- that have an annular fibrous cuff that holds that cath-
ysis via a tunneled dialysis catheter while waiting for a eter in place within the tunnel tract.3 CVCs are composed
wrist fistula to mature has evolved to a “fistula first,” “line of biologically neutral material that should not induce
last” strategy. This is employed with the aim of preserving catheter lumen thrombosis or a perivascular reaction
venous real estate and ensuring patients start dialysis using and subsequent venous thrombosis. The catheter should
an access that is patient-specific based on comorbidities and be soft and compliant, easy to insert, durable, and should
needs. be coated with an agent that reduces bacterial prolif-
Technologic advancements of products specifically for eration and biofilm formation. Furthermore it should be
dialysis access have provided an array of options for patients inexpensive and permit blood flow of >350 mL/min to
on RRT, particularly when access options have been facilitate efficient dialysis. Numerous catheters are avail-
exhausted. The improved long-term outcomes with newly able for use, and they can be differentiated by the design
developed grafts along with increasing armamentarium of the distal tip and the presence of side holes and flow
such as drug-eluting balloons and stents for the mainte- dividers.3 The hemodynamic effects of these design varia-
nance of access or technologies to recanalize occluded cen- tions are regularly debated and remain largely unknown
tral veins are providing renal failure patients with ongoing and are influenced by numerous factors such as insertion
reliable access and improved life expectancy rates on dialy- technique, site of insertion, and position of the catheter
sis. This chapter will explore much of the current modern- tip.3 Although interesting, catheter design and inser-
day practices associated with establishing and maintaining tion principles are less important than providing efficient
RRT for patients with ESRF. dialysis. This includes the ability of the catheter to be able
to process at least 50 L of fluid during a standard 4-hour
VASCULAR ACCESS CATHETERS dialysis session with favorable arterial and venous pump
pressures and minimal recirculation.3–5
Temporary Dialysis Catheters The right IJV is the most favored site of placement
Central venous catheters (CVCs) are an option in patients of a permanent catheter, which is then tunneled to an
with renal failure to provide dialysis. Catheters are either exit site on the anterior chest wall. Care should always
cuffed or uncuffed. Uncuffed CVCs are used as a temporiz- be taken to ensure that the cuff is at least 4 cm from the
ing measure in patients who require immediate or emer- exit site to minimize the risk of cuff extrusion and mini-
gent hemodialysis. About 40% of patients with renal failure mize infection rates. Internal jugular vein catheter place-
present acutely and require short-term vascular access and ment can be performed surgically using a cut down at the
dialysis. These patients include the “acute presenter” and medial border of sternocleidomastoid. However, the Seld-
the “crash lander.” Acute presenters require short-term inger technique using ultrasound to guide the needle into
RRT to allow renal recovery, and crash landers, those who the vessel is more frequently performed. It is essential to
present suddenly with new ESRF, require short-term access place catheters under fluoroscopic control, to ensure that
during planning for a long-term option. In both instances a the tip of the catheter is placed in the superior vena cava
temporary uncuffed CVC may be placed to facilitate dialysis. (SVC).6 The malposition rate of catheters when placed
Temporary catheters can be inserted at the bedside and can without fluoroscopic control has been shown to be as
remain in situ for up to 3 weeks depending on the site of high as 29%.7,8 The tip of the catheter should be at the
insertion. junction of the SVC and right atrium as this affords the
The three most common locations for temporary vascu- most optimal blood flow through the catheter. A list of
lar access are the internal jugular vein (IJV), subclavian complications associated with central venous catheter
vein (SCV), and common femoral vein (CFV). Upper limb insertion is displayed in Box 5.2.
catheters can remain in situ for up to 3 weeks, patients can Tunneled catheters can provide access for months and
remain fully ambulatory, infection rates are lowest, and even years, particularly in those patients in whom all
they are therefore preferred to groin CVCs. Currently the native venous conduits for AVF or AVG formation have
favored site for temporary venous access is the right IJV, been exhausted or where arteriovenous strategies are
although some patients find the visibility of the catheter in likely to induce risks of limb loss from steal syndrome or
the neck above the collar unsightly. The advantages of IJV precipitating heart failure from the high volume venous
placement are lower risk of infection, higher patency rates, return. Indeed, in such patients who become dependent
and lower risk of insertion-related complications. on a tunneled catheter, placement is becoming more
adventurous with reports of transhepatic, translumbar
Permanent Dialysis Catheters and even transmediastinal approaches being used as last
Patients requiring long-term dialysis and in whom there resort procedures.9–11
has not been an opportunity to create a fistula, AVG,
or peritoneal dialysis catheter in a timely fashion will COMPLICATIONS OF HEMODIALYSIS
require a permanent dialysis catheter. In the US the inci- CATHETERS
dence of patients starting dialysis on a tunneled catheter
remains high at around 70%.2 The UK incidence is not Catheter Dysfunction
much better with 48% of new starters initiating hemo- Catheter dysfunction was historically defined as “failure to
dialysis via a CVC.1 The current indications for central attain and maintain adequate extracorporeal blood flow
5 • Access for Renal Replacement Therapy 71

contrast via the arterial port can help elucidate the pres-
BOX 5.1 Indications for Tunneled ence of a fibrin sheath. Treatment of a fibrin sheath usu-
Hemodialysis Catheters ally involves replacement of the catheter at a new location
During maturation of autogenous arteriovenous fistula
although there is some evidence to support mechanical
During maturation of peritoneal dialysis catheter stripping, which is carried out using a snare around the
Patients awaiting a living donor transplant catheter.14
Dialysis bridge after failure of current access to permit planning
and imaging for long-term access
Catheter-Related Central Vein Stenosis
Permanent access—all other sites exhausted, severe cardiac dys- The long-term presence of any device within the central
function or patient choice. venous system is associated with the development of venous
stenosis. The exact mechanism by which this happens is
unclear; however, catheter-related thrombosis or infection
along with progressive trauma to the endothelium is the
BOX 5.2 Complications of Central Venous most likely cause. The incidence of CVC-induced stenosis is
Catheter Insertion reportedly as high as 50% although there are values as low
as 18% in the literature. The rate of stenosis after subcla-
Arterial puncture vian vein cannulation is higher than that seen when the
Bleeding catheter is placed in the IJV, and catheters placed on the left
Pneumothorax side of the neck have a higher reported incidence of associ-
Hemothorax ated stenosis compared with those placed on the right.15,16
Hemomediastinum Fig. 5.2 shows an example of a right arm venogram in a
Atrial perforation patient with a central venous stenosis as a result of a CVC.
Air embolus
Arrhythmias Central Vein Occlusion
Primary failure
The presence of occlusive thrombus in the central veins,
in particular the SVC, is asymptomatic in about 30% of
cases. Although it can manifest as catheter dysfunction, the
sufficient to perform dialysis in a timely fashion.” A recent presence of arm and facial edema or prominent superficial
collaboration of transplant surgeons and physicians, the vessels should raise suspicion of central venous occlusion
North American Vascular Access Consortium (NAVAC), or stenosis. Thrombus is usually detected by angiography;
introduced a new definition in an attempt to create a uni- however, transesophageal Doppler and contrast-enhanced
form standard. Dysfunction was defined as the first occur- cross sectional imaging are also useful diagnostic tools. The
rence of either (1) peak blood flow of 200 mL ⁄ min or less treatment of central vein thrombus depends on the chro-
for 30 minutes during a dialysis treatment, (2) mean blood nicity of the thrombus. Acute thrombus may respond to
flow of 250 mL ⁄ min or less during two consecutive dialysis fibrinolytic therapy. However, fibrinolytic therapy will not
treatments, or (3) inability to initiate dialysis owing to inad- have any effect on older, organized thrombus. Percutane-
equate blood flow, after attempts to restore patency have ous angioplasty and stenting have an increasing role in
been attempted. maintaining central venous catheter patency and recana-
Dysfunction accounts for 17% to 33% of all catheter lization of stenosed or occluded vessels.17 In addition, the
removals and can be either early or late. Early dysfunction advent of novel technology such as the Surfacer Inside-Out
is due to technical error such as kinking of the catheter device (Merit Medical) has provided a potential means to
in the subcutaneous tunnel or catheter malpositioning. persist with upper limb access in the presence of SVC occlu-
Late dysfunction is due to central vein occlusion, cath- sion. The details of the device are covered in the Advances
eter thrombosis, and fibrin sheath formation (Fig. 5.1).7 in Access section later in the chapter.
Catheter thrombosis has an estimated frequency of 0.5 to
3 episodes per 1000 days and an incidence of 46% and is Infection
the major cause of catheter dysfunction.7,12 Antifibrinol- Septic complications from CVCs are well documented, and
ytic therapy introduced via the catheter is the treatment the longevity of modern cuffed and tunneled catheters is
of choice and first-line therapy is usually 5000 IU/mL of still limited by infection. Catheter-related bacteremia rates
urokinase of sufficient volume to fill the lumen. This can for cuffed, tunneled catheters ranges from 1.5 to 5.5 per
be repeated immediately and if bolus dosing fails can be 1000 days.18 Catheter-related infections include exit site
followed by a systemic infusion of 20,000 IU/mL/hr over 6 infection, tunnel infections, and catheter-associated bac-
hours or a continuous infusion during dialysis of 250,000 teremia. Sepsis is the most significant cause of catheter-
IU. An RCT of tenecteplase versus placebo in patients with associated morbidity and mortality (Fig. 5.3). Catheter
catheter dysfunction demonstrated that patients treated infection is responsible for between 6% and 28% of cathe-
with tenecteplase had significantly increased flow rates ter failure.19 Gram-positive skin dwelling bacteria, in par-
(>300 mL/min). Patients within the study with <1 day ticular Staphylococcus species, account for 40% to 80% of
of catheter dysfunction had the greatest benefit with 35% infections with gram-negative organisms accounting for
of tenecteplase-treated patients gaining adequate dialy- 20% to 40%. Polymicrobial (10%) and rare causes such
sis flow rates compared with 8% in the placebo group.13 as fungal infection (<5%) make up the remainder.19 The
If thrombolysis fails then imaging of the catheter to route of infection is twofold, either tracking along the
obtain further information is indicated. Direct injection of external surface of the catheter or via the lumen. Exit-site
72 Kidney Transplantation: Principles and Practice

Fibrin
Sheath

Intraluminal
Clot

Mural
Thrombosis

Venous
Thrombosis
Fig. 5.1 Illustration of the causes of central venous catheter dysfunction. Determining the cause of dysfunction will direct immediate and future
management.

TCC in situ 6/12

Subsequent Right B/C


Failing to mature

Fig. 5.2 Venographic image illustrating a right sided central


venous stenosis resulting from a central venous catheter. Note the
tapering of the contrast around the catheter and the presence of tortu-
ous collateral vessels visible in the upper arm and axilla as a result of
the stenosis.

infections present with localized erythema with or with-


out discharge and often respond to topical or oral anti-
Fig. 5.3 Photograph of a patient with a tunnel infection. There is
biotics. Unresponsive infections or those with discharge erythema along the course of the subcutaneous catheter and at the
and no signs of systemic sepsis and no bacterial growth exit site.
on blood culture should be treated with parenteral anti-
biotic therapy. The Kidney Disease Outcomes Quality Ini-
tiative (KDOQI) guidelines recommend treatment for 3 Fistulas and Synthetic Grafts
weeks.20,21 However, if the patient is hemodynamically
unstable or fails to improve after 36 hours of parenteral In the past decade there has been a concerted effort to
antibiotics, with bactericidal serum levels of an appropri- improve the outcomes for patients requiring arteriovenous
ate antibiotic, the catheter should be removed. Serious access for dialysis. This has led to an expansion in the vol-
complications from catheter-associated infections occur ume and quality of evidence in vascular access. There has
in 3% to 44% of cases and include endocarditis, osteomy- been an increase in the number and variety of randomized
elitis, thrombophlebitis, septic arthritis, spinal epidural controlled trials (RCTs), prospective cohort studies, and
abscess, and large atrial thrombi. Mortality from cathe- guidelines to inform practice. This has coincided with the
ter-associated infections is greatest when Staphylococcus concept of a personalized access plan for each patient and
aureus is the culprit with a reported rate of up to 30% in has challenged clinicians to consider all possible options,
some units. Methicillin-resistant Staphylococcus aureus including AVGs as an initial access procedure. “Fistula first”
(MRSA) has three to five times higher mortality compared should still be considered the foundation for the majority of
with methicillin-sensitive strains and is considerably more patients; however, decision making on access should be on
costly to treat and manage.19 an individual basis.
5 • Access for Renal Replacement Therapy 73

AVFs using native veins are associated with the high- 85% of all prevalent hemodialysis patients should be dia-
□ 

est long-term patency and lowest complications rates,22–24 lyzed using a native AVF.
although they suffer from high initial failure rates.25,26 In No patient requiring dialysis should wait more than 4
□ 

patients with enough time for fistula planning, creation, weeks for fistula construction.
and maturation, they should be the vascular access of Permanent access should be constructed 16 weeks and
□ 

choice for patients requiring long-term hemodialysis.1,27 preferably 6 months before anticipated need for dialysis.
Synthetic grafts have lower rates of primary failure but can Preservation of arm veins is critical and unnecessary ven-
□ 

be used for dialysis earlier; indeed many synthetic grafts can ipuncture must be avoided.
be needled immediately.28 However, the complication and   

intervention rates are higher, and the use of an AVG may The Vascular Access Society recommendations are simi-
reduce the amount of native vein available for future use. lar, and as with those of NKF-KDOQI and the Renal Asso-
Furthermore the National Kidney Foundation-Kidney Dis- ciation, state that arm veins must be preserved and that the
ease Outcomes and Quality Initiatives (NKF-KDOQI) guide- planning of access should commence when a patient’s GFR
lines state that all suitable patients should have a native reaches 20 to 25 mL/min. Patients should see an access
AVF for dialysis access. surgeon when the GFR is 15 mL/min so that access can
be created 6 to 12 months in advance of dialysis (or even
sooner if progression is rapid).
PLANNING AND TIMING OF VASCULAR ACCESS
Emerging data suggest that AVF outcomes in terms of
Construction of an AVF involves the conversion of an patency are significantly improved if access is constructed
accessible vein to a high-flow vessel from which blood can earlier rather than when the patient is on the brink of dialy-
be rapidly removed and returned via two needles during sis.31 It is unclear why this may be but it has been suggested
a dialysis session. Veins are low-pressured, low-resistance that worsening uremia affects vascular biology especially at
capacitance vessels. Their relative lack of elastic recoil the level of the AVF anastomosis.32
compared with that seen in arteries allows them to progres- The historical focus in vascular access has been AVF loca-
sively increase in size if high-pressure flow passes through tion-specific, rather than patient-specific. Vascular access
them. After AVF formation, high-pressure arterial flow has become much more patient centered and bespoke even
passes into the low-pressure capacitance vein. The sub- if that conflicts the view of wrist fistula first. However, the
sequent increased turbulent flow gives rise to a palpable importance of carefully planning access procedures to pro-
thrill and an audible bruit within the AVF. In addition, the tect venous real estate still remains. There are a few general
fistula vein increases in size over time. It is this second fea- principles that are important in the planning of vascular
ture that is crucial for needling and subsequent hemodi- access. The upper limb should be used in preference to the
alysis to take place. As a general rule, the fistula vein needs lower limb and distal sites should be used before proximal
to be a minimum of 6 mm diameter 6 weeks after creation sites. This permits the maximum use from a particular ves-
of the fistula if needling is to be attempted and hemodialy- sel and preserves proximal sites for future use. It is also
sis successful. preferential to use the nondominant arm ahead of the domi-
Given that there is a minimum of 6 weeks maturation nant arm, and this is particularly important in patients who
time of a fistula from the point of its creation, the timing of needle their own fistula at home. The formation of preemp-
fistula creation in relation to the predicted start of dialysis tive AVF requires liaison between nephrologist and surgeon
is critical. Permanent vascular access in the form of a fis- and flexibility in theater list planning to ensure patients
tula should be created well in advance to allow not only for with rapidly deteriorating renal function can be accommo-
maturation but also for further procedures should there be dated before reaching end-stage disease. However, patients
failure of the initial access procedure. Many guidelines exist who are referred late, who are already undergoing RRT,
regarding the timing of vascular access. In the US the NKF- or who present acutely with an emergent need to dialyze,
KDOQI guidelines recommend:29 should have planning of vascular access adapted from the
   wrist fistula first model, to suit the patient. Such examples
Patients with glomerular filtration rate (GFR) <30 mL/
□  include the use of early cannulation AVG (ecAVG), which
min should be educated on all modalities of RRT includ- can be needled immediately and used in preference to a
ing transplantation. tunneled dialysis catheter. A recent RCT randomized 121
A fistula should be placed at least 6 months before the an-
□  patients who needed dialysis within 48 hours to ecAVG or
ticipated start of dialysis. tunelled central venous catheter (TCVC). They demon-
A new AVF should be allowed to mature for at least 1
□  strated a reduction in bacteremia rates at 3 months in the
month and preferably 3 to 4 months before cannulation. ecAVG group and lower mortality. The higher upfront cost
Prosthetic grafts should be placed 3 to 6 weeks before an-
□  of ecAVG was offset at 6 months by the increase in compli-
ticipated need for hemodialysis. cations of the TCVC, and those in the ecAVG had a shorter
□ Never insert hemodialysis catheters until actually needed. hospital stay and reduction in use of temporary CVCs.33
  

In the UK the Renal Association and the joint working PREOPERATIVE ASSESSMENT
party of the Renal Association, the Vascular Society of Great
Britain and Ireland, and the British Society of Interventional The three requirements for a successful AVF are as follows:
  
Radiology have produced the following standards1,30:
   1. Good arterial supply—inflow
65% of all patients presenting within 3 months of dialysis
□  2. Adequate vein—conduit
should start hemodialysis on a useable AVF. 3. Patent central veins—outflow
  
74 Kidney Transplantation: Principles and Practice

Adequacy of the inflow to the arm can usually be not. The disadvantages of radiocephalic AVFs are the high
assessed clinically with palpation of the brachial, ulnar, primary failure rate and failure to mature. Mean primary
and radial pulses and Allen’s test can be used to determine patency rates at 1 year are 55% (46%–63%) and secondary
patency of the palmar arch and the dominance of radial or patency rates are 71% (65%–77%). At 1 year, 23% (17%–
ulnar artery in supplying the hand. Suitability of arm veins 29%) of fistulas will have been abandoned without use.40
can usually be determined clinically using a tourniquet The surgical technique for formation of radiocephalic
placed proximally on the arm. However, vein diameter and fistulas follows. The cephalic vein and radial artery are
patency can be confirmed by duplex ultrasound, which is exposed via a longitudinal, oblique, or S-shaped incision
cheap, noninvasive, and improves patency rates in those depending on the proximity of the vessels and the prefer-
patients with veins that are difficult to assess clinically.34,35 ence of the surgeon. The cephalic vein is mobilized for 3 cm
It is generally accepted that to stand a good chance of suc- from beneath the lateral skin flap ensuring preservation of
cess the vein needs to be greater than 2 to 2.5 mm in diam- the sensory dorsal branch of the radial nerve. The artery is
eter although the evidence is conflicting.34,36,37 Assessment located lateral to the tendon of the flexor carpi radialis and
of the basilic vein is usually performed using duplex ultra- lies underneath the fibers of the deep fascia, which must be
sound. In patients with previous CVCs or suspected central divided. Between 2 to 3 cm of artery should be mobilized
venous pathology, venography should be performed before and branches of the vessel can be ligated and divided. The
surgery. vein and artery are controlled proximally and distally with
vascular slings and microvascular clamps. In an end-to-
side anastomosis the cephalic vein is ligated distally and
ANESTHESIA
divided obliquely to leave a spatulated end for anastomo-
The majority of upper limb AVF can be performed under sis. An arteriotomy is then performed on the anterolateral
local anesthetic (LA), in particular wrist and elbow fistu- surface of the radial artery. In a side-to-side anastomosis an
las. For more extensive procedures such as transposition of arteriotomy (1–1.5 cm) is performed on the lateral aspect
the basilic vein and forearm loop grafts, a regional block of the vessel, and afterwards a venotomy of equal length is
with local infiltration may suffice. Regional anesthesia performed on the medial side of the cephalic vein. A 6-0 or
(RA) has the added advantage of blocking sympathetic 7-0 nonabsorbable, monofilament suture with two needles
nerves as well as sensory nerves, which causes vasodila- is then used to perform a continuous anastomosis. Once
tion. In prolonged operations or if the patient is intolerant the anastomosis has been completed and in the presence
of the procedure, the anesthetist can administer a short- of an acceptable thrill within the vein, the distal cephalic
acting sedative to avoid a general anesthetic (GA). This is artery can be ligated and divided. In a successful procedure
advantageous because many patients with renal failure there should be a thrill present once the clamps have been
have significant cardiovascular disease and other comor- released and the slings loosened. In the absence of a thrill
bid conditions making a GA high risk. Surgical procedures one must ensure the patient is not hypotensive, there are
in the axilla and lower limb usually require GA. Compari- no adventitial bands constricting the vein, and there are no
son of LA and RA on patency rates of AVF has been the errors, such as an intimal flap, twisting of the vein, or pres-
subject of a recent RCT and meta-analysis. An observer ence of thrombus.
blinded RCT of 126 patients allocated to brachial plexus
block (BPB) or LA demonstrated greater patency in the BPB Elbow Fistulas
group at 3 months. However, functional patency rates, In the presence of failed radiocephalic fistulas or inadequate
which are a more clinically relevant outcome, did not dif- forearm vessels, an autogenous elbow fistula should be the
fer between the groups.38 A meta-analysis of four RCTs next procedure of choice. A brachiocephalic AVF is usually
comparing AVF failure rates in those performed using RA the first procedure of choice at the elbow and is a straight-
with LA concluded that failure rates were lower in the RA forward procedure performed under LA.
group. Follow-up in the studies varied from 40 to 100 days,
and the definition of failure was slightly different in each Brachiocephalic Arteriovenous Fistulas. The original
paper; however, a sensitivity analysis still demonstrated brachiocephalic AVF (BCAVF) was described as an end-to-
that failure rates were lower in RA.39 There are no longer- side anastomosis of the cephalic vein to the brachial artery
term data on patency rates of AVFs formed under RA or (Fig. 5.4). As with radiocephalic AVF, if the elbow vessels
LA, which would be interesting, because this information are small then a side-to-side configuration can also be used.
would better inform future practice. The venous anatomy in the cubital fossa is variable, and it
is important to establish how the veins are related before
AUTOGENOUS ARTERIOVENOUS FISTULAS deciding on which vessel to use; the cephalic vein at the
elbow is often used for venipuncture and can be sclerosed
Wrist Fistula and unsuitable. The median cubital vein often drains into
The initial procedure of choice, in patients with suitable the cephalic vein and can also be used for anastomosis to the
vessels, is a radiocephalic AVF at the wrist. The operation is brachial artery. The surgical technique for the brachioce-
performed under LA as a day case procedure. The original phalic fistula is the same in principle as for the radiocephalic
Brescia-Cimino AVF involved a side-to-side radial artery to at the wrist. Patency rates of BCAVFs are similar to radioce-
cephalic vein anastomosis although more recently an end phalic AVFs at 1 year, with mean primary patency of 52%
of cephalic vein to side of radial artery has come into favor. (41%–61%) and mean secondary patency rates of 74%
The original Brescia-Cimino AVF often led to venous hyper- (63%–83%). They also have excellent longer-term patency
tension in the hand, whereas the end-to-side variation does of up to 70% at 3 years.40,41 Unlike with radiocephalic
5 • Access for Renal Replacement Therapy 75

Fig. 5.4 Intraoperative photograph of a brachiocephalic arteriove-


nous fistula. The brachial artery is controlled by vascular slings (yellow)
and bulldog vascular clamps. The cephalic vein has been divided, spat- Fig. 5.5 Photograph of a brachiobasilic transposition fistula. The
ulated, and has been anastomosed to the side of the brachial artery. fistula course is visible from the cubital fossa along the arm toward the
axilla with a healed scar identifying the original anatomic position of
the vein.
fistulas, there can be a significant increase in flow rates in
BCAVFs, which can lead to high-output cardiac failure and study yielding patency rates of 70% at 3 years.40,41 A recent
steal syndrome. Such complications can be minimized by systematic review and meta-analysis concluded that there
ensuring the arteriotomy does not exceed 75% of the diam- is no statistical difference in outcomes between one- and
eter of the artery. two-stage procedures although individual studies tended to
favor the two-stage procedure.43
Brachiobasilic Arteriovenous Fistula. The basilic vein
originates on the medial aspect of the forearm at the wrist
ARTERIOVENOUS GRAFTS
and runs superficially in the forearm. It only remains
superficial for a short distance in the arm before coursing Historically, in the UK and Europe, prosthetic grafts were
beneath the deep fascia to run up the medial aspect of the reserved for patients who had exhausted native venous
arm alongside the medial cutaneous nerve of the forearm. access, and although that is still the most common indica-
Brachiobasilic fistula formation can be split into one-stage tion, practice has changed. This is in an attempt to avoid
and two-stage procedures. A one-stage procedure is usually the complications associated with the use of temporary
performed under general anesthesia and requires an exten- “bridging” central venous catheters. The three categories of
sive incision from the cubital fossa running longitudinally patients who require urgent access for dialysis in whom this
along the medial aspect of the arm toward the axilla. The is relevant are as follows:
  
basilic vein is mobilized from beneath the deep fascia and
Patients with an acute presentation of chronic renal
□ 
all tributaries are ligated and divided. Once an appropri-
failure
ate length has been exposed the vein is divided and placed □ Patients who have lost their current hemodialysis access
superficial to the medial cutaneous nerve of the forearm.
Patients requiring a long-term modality switch from peri-
□ 
The vein can then either be transposed via a subcutaneous
toneal dialysis to hemodialysis
tunnel, or superficialized, where the lateral skin flap can be   

undermined and the vein placed in a suitable position for The first group includes patients who require dialysis
needling, above the deep fascia (Fig. 5.5). The advantages urgently and those who will need dialysis before an AVF
of a one-stage procedure are that it only requires one opera- can be performed and will mature. As a result there has
tion and a single hospital stay, and the fistula can be used been an increase in the use of prosthetic grafts, in particu-
more quickly. The disadvantage is that if the fistula fails lar, ecAVGs. Conventional AVGs are composed of expanded
the patient has undergone a significant procedure, with a polytetrafluoroethylene (ePTFE), which is durable, easy to
substantial incision, usually under general anesthesia. A handle, and produces reproducible results. It requires about
two-stage procedure comprises a first stage, during which 14 days to become incorporated into the surrounding tissue
the basilic vein is anastomosed to the brachial artery under before use, which limits its use as a rescue procedure. Early
local anesthetic, through a small cubital fossa incision. The cannulation grafts such as Flixene (Maquet) or Acuseal
fistula is then assessed for maturation 4 to 6 weeks after (W.L. Gore) can be used within 24 hours of formation and
formation, and if deemed adequate, the second stage can can therefore avoid bridging dialysis catheters. This reduces
be performed. Innovative minimally invasive techniques the infection risk associated with catheters and minimizes
have also been described using videoscopic assistance to damage to central veins thereby reducing risk of central
mobilize the vein and ligate the tributaries although such venous stenosis.
techniques are not in widespread use.42 The mean 1-year Data on patency rates and complications in AVFs and
primary patency rate is 55% (47%–63%) and mean second- AVGs generally favor AVFs.23,24,44 In a systematic review of
ary patency rate is 75% (67%–82%) with one randomized more than 200 studies with 875,269 access events, 2-year
76 Kidney Transplantation: Principles and Practice

pooled primary patency rate for AVFs was 0.55 (95% confi-
dence interval [CI], 0.52–0.58) and secondary patency was
0.63 (95% CI, 0.59–0.67). In AVGs the values were inferior
with primary patency of 0.40 (95% CI, 0.35–0.44) and sec-
ondary patency of 0.60 (95% CI, 0.55–0.65).27 In another
review primary patency rates ranged from 0.18 to 0.70 at
1 year for AVGs compared with 0.53 to 0.74 for AVFs. Sec-
ondary patency rates at 2 and 3 years were 0.47 to 0.73
and 0.39 to 0.70 for AVGs compared with 0.26 to 0.94 and
0.64 to 0.84 for AVGs.45
Complication rates are higher in AVGs than AVFs
although AVFs had greater rates of stenosis (51.4% vs.
40.6%, P = 0.0182), whereas AVG had greater thrombosis
rates (14.6% vs. 31.9%, P < 0.001) and overall increased
risk of death compared with an AVF (relative risk = 1.18,
95% CI = 1.09–1.27).45,46 AVGs also have a higher rate of
interventions to retain patency with data in the range of 1.7
to 3.5 per graft per year.46,47
A prosthetic graft can be anastomosed to any suitable
artery and vein, in an end-to-side disposition and then
tunneled subcutaneously. It can be performed under LA
but usually requires general anesthesia. Brachioaxillary
grafts in a straight configuration are an excellent option A B
in patients with unsuitable cephalic and basilic veins at
the elbow. Grafts can also be configured as a loop and they Fig. 5.6 (A) Photograph of left upper arm graft (brachial artery to axil-
lary vein) with evidence of needling along the length of the graft. (B)
are commonly located in the forearm, arm, and thigh (Fig. Photograph of forearm loop PTFE graft. The cubital fossa incision and
5.6). Forearm loop grafts are an excellent option in obese forearm incision are clearly visible and the loop configuration of the
patients with deep upper arm veins because the procedure graft can be seen.
can be performed using RA or LA. It also preserves upper
arm veins for future use. US data demonstrates that fore-
arm loop grafts have similar patency rates to upper arm
straight grafts.48 However, this may not be transferable to are anticoagulated. Direct pressure on the site of veni-
a European population because up to 90% of the patients puncture will usually stop the bleeding and reversal of
had a graft as their initial access procedure. AVGs can be anticoagulation is not usually necessary. Bleeding from
constructed in many anatomic locations once conventional aneurysms or at needling sites as a result of infection can
access has been lost and such examples include forearm, be catastrophic and frightening for the patient. Although
arm, on the chest wall, in the thigh, and as a necklace— direct pressure may be sufficient to control hemorrhage,
from one axilla to the other. More elaborate procedures surgical exploration is often required and may result in
have also been undertaken including brachiojugular grafts, ligation of the fistula, for which the patient must be con-
and a straight graft has successfully been fashioned from sented. Exploration of a late bleed from a fistula should be
the axillary artery to the external iliac vein and even the performed under general anesthesia because an exten-
subclavian artery to the right atrial appendage.49–51 Once sive incision is often required to gain proximal control
venous access is lost, arterioarterial grafts have been used of the vessels and it can be extremely unsettling for the
with impressive success. A systematic review showed pri- patient.
mary patency rates ranged from 67% to 94.5% at 6 months
to 54% to 61% at 36 months, with secondary patency Stenosis
rates from 83% to 93% at 6 months to 72% to 87% at 36 The most common cause of AVF and AVG thrombosis
months.52 is due to the presence of an underlying stenosis.54 In
native AVFs stenosis can be present at any point from the
arterial anastomosis to the central veins. The common
COMPLICATIONS OF ARTERIOVENOUS
locations are juxta-anastomotic, at the swing point, at
FISTULAS AND GRAFTS needling sites and the cephalic arch (BCAVF). In AVGs
Hemorrhage the most common location is at the venous anastomosis.
Bleeding can be categorized as early, which is within the The pathophysiology of stenosis is as a result of neointi-
first 24 hours, or late, which is anytime thereafter. Early mal hyperplasia, thought to be due to turbulent blood
bleeding may either be due to technical error within the flow and shear stress, although a proportion of patients
anastomosis, slipping of a ligature, or due to generalized have intimal hyperplasia evident before AVF formation.
oozing as a result of uremic platelet53 dysfunction. Clini- There is conflicting evidence on the effect this has on AVF
cians should have a low threshold to reexplore the fistula maturation.55–57 Early detection of stenosis increases
to resolve any technical cause of bleeding. Late hemor- the number of fistulas that mature and prolongs fistula
rhage from an AVF is usually from a needling site imme- patency.58–60 Fistula and graft surveillance is discussed
diately after dialysis and often occurs in patients who in more detail later.
5 • Access for Renal Replacement Therapy 77

Fig. 5.7 Intraoperative photograph of a patch-plasty repair of the bra- Fig. 5.8 Photograph of a pseudoaneurysm of an upper arm PTFE
chial artery just proximal to the origin of the cephalic vein anastomosis. graft. The patient had also had a previous brachiobasilic arteriovenous
The same technique of suturing a prosthetic patch to a fistula can be fistula shown by the longitudinal scar on the inferior aspect of the arm.
used to treat an underlying stenosis.
relative immunocompromised state uremia produces.
Uremia affects the immune system by inhibiting the bac-
Thrombosis tericidal, phagocytic, and chemotactic action of neutro-
Immediate thrombosis in the presence of adequate quality phils and by suppressing both B cell and T cell64 responses.
and appropriate sized vessels may be due to technical error or Furthermore renal patients are more readily colonized by
a platelet plug and merits reexploration. Thrombectomy and S. aureus compared with the general population, with an
refashioning of the anastomosis should salvage the fistula. incidence of up to 70% upper respiratory tract colonization
Early thrombosis, that occurs after 24 hours but before the compared with 15%,65 respectively. S. aureus is the leading
fistula maturing, may be as a result of patient factors such as culprit in infective complications of vascular access con-
hypotension, either as a result of fluid depletion after dialysis duits and is often resistant to first-line antibiotics. Routine
or cardiac failure, or it may be due to inadequate vessel size use of antibiotics for autologous AVF formation is not uni-
and/or quality. Attempted salvage of an early thrombosis is versal. However, any procedure in which prosthetic mate-
often unsuccessful and a pragmatic approach should be taken rial is used, a second-line intravenous antibiotic such as
to avoid unnecessary and costly interventions that are often vancomycin or teicoplanin should be given. Wound infec-
futile. Late thrombosis is usually due to the presence of a grad- tion rates are as low as 2% to 3% after autologous AVF for-
ually progressive stenosis secondary to neointimal hyper- mation although they are higher in AVGs, ranging between
plasia, and these account for about 85% of all stenoses.61 5% and 35%.27,46,66 Drainage of any collections either by
Treatment of fistula thrombosis can be radiologic or surgical liberal suture removal or formal evacuation and irrigation
and is dependent on local expertise. Radiologic intervention under anesthesia alongside antibiotic therapy resolves the
has the combined advantage of thrombectomy followed by majority of infections. If there is concern about the severity
venography and treatment of any underlying stenosis with of the infection or if MRSA is isolated and the infection is
balloon angioplasty. Surgical intervention can also provide not responding to antibiotic therapy, then surgical debride-
definitive treatment of stenoses in the form of patch plasty ment and inspection of the anastomosis should be under-
(Fig. 5.7) or interposition graft, as long as the stenosis is acces- taken. There is a small but potentially life-threatening risk
sible to the surgeon. There is some evidence that stenoses of hemorrhage from an infected fistula, and ligation of the
that are treated surgically have better long-term patency.62 fistula may be required. Superficial wound infection in a
A recent comprehensive review of percutaneous interven- patient with underlying prosthetic material should always
tion for thrombosed vascular access demonstrated an 80% be treated seriously. Aggressive, early antibiotic therapy
success rate in retaining patency and avoiding temporary should be employed to treat the infection and reduce the
access63 catheters. Fistulas that are unsuitable for percuta- risk of graft infection. Superficial infection can often be
neous intervention often require revision because of anas- treated successfully, but if there is a purulent infection or
tomotic or perianastomotic strictures that cannot be treated the graft is proven to be infected it must be removed.
by angioplasty. Standard Fogarty vascular thrombectomy
catheters are usually sufficient to clear early, soft thrombus Aneurysm Formation
but are not designed to penetrate mature thrombus. Fistulas Vascular access conduits can develop true and pseudoan-
that require surgical revision must retain enough length to eurysms. Unlike a true aneurysm, a pseudoaneurysm does
accommodate two dialysis needles after revision. not contain all layers of the blood vessel wall. It is a hema-
toma in direct communication with the lumen (Fig. 5.8).
Infection The incidence of pseudoaneurysm is about 10% in pros-
Vascular access procedures are “clean surgery” although thetic grafts and 2% in autologous AVF. Duplex ultrasound
infection rates in renal patients are higher because of the is usually diagnostic but venography or cross sectional
78 Kidney Transplantation: Principles and Practice

A B
Fig. 5.9 (A) Radiograph of a fistulogram showing needle-site true aneurysms of a brachiocephalic arteriovenous fistula. The upper black arrow shows
the brachial artery, lower black arrow the anastomosis, and the white arrows show the aneurysms. The adjacent photograph (B) is the same fistula.

imaging may be warranted if central venous pathology is


suspected. Treatment can be radiologic or surgical, with
the latter being the conventional method. Surgical repair
usually involves oversewing the defect or removing the
damaged section of graft and restoring the AVF either by
direct end-to-end anastomosis or by using an interposition
graft. Increasingly interventional radiologic methods are
employed first; direct injection of thrombin into the defect
is usually sufficient if the defect is small, and percutaneous
deployment of a covered stent can also be used to exclude
the aneurysm from the circulation. A true aneurysm can be
defined as a threefold or greater increase in diameter com-
pared with the rest of the access. True aneurysms are rela-
tively commonly in upper limb fistulas, and older fistulas
are more likely to become aneurysmal (Fig. 5.9). Aside from
the fistula being unsightly, most aneurysms are uncompli-
cated and at extremely low risk of rupture. However, aneu-
rysms that are rapidly increasing in size, have thin skin, or Fig. 5.10 Photograph of arteriovenous access ischemic steal syn-
infection present should be surgically corrected or ligated. drome. The left hand is discolored, and on the index and middle fingers
Investigation of the rest of the access is essential before there is evidence of early ulceration.
intervention, because up to 50% will have a clinically rele-
vant stenosis. Many different surgical procedures have been affected, causing severe pain and neurologic defect imme-
detailed, and the choice is surgeon-dependent.67 diately after AVF formation. The treatment is urgent AVF
ligation to prevent long-term neurologic deficit. Mild steal
Arteriovenous Access Ischemic Steal Syndrome syndrome usually improves with conservative manage-
Arteriovenous access ischemic steal (AVAIS) syndrome is ment and the improvement is due to increase in collat-
diagnosed when there is hypoperfusion of the limb distal to eral flow around the elbow. Duplex imaging will establish
the arteriovenous anastomosis. If flow dynamics are inves- whether there is reversal of flow in the artery distal to the
tigated, it is present in many patients but only 1% to 8% of anastomosis, which is characteristic of AVAIS. AVF flow
patients are symptomatic. AVAIS is most common in the should also be measured as in the presence of low flow, and
upper limb in procedures involving the brachial artery, and underlying arterial disease surgical management may be
patients with arteriosclerosis and diabetes are particularly limited to ligation of the access and treatment of underlying
at risk with an incidence of up to 25%. The presence of a arterial disease. Angiography will determine the severity of
small brachial artery (less than 5 mm) at the time of surgery arterial disease, and any proximal stenosis should be treated
should alert the surgeon to the possibility of steal syndrome, with angioplasty. High flow through the AVF permits more
and limiting the arteriotomy to 75% of the vessel diameter management options, and if symptoms are severe or there
may reduce the overall risk. Symptoms of steal syndrome is critical ischemia due to the steal syndrome, then surgical
range from mild, such as a cold hand, to severe ischemia intervention is indicated. The simplest intervention is liga-
with rest pain, neurologic deficit, and tissue loss (Fig. 5.10). tion of the fistula, which almost invariably ameliorates the
Ischemic monomelic neuropathy (IMN) is a severe form symptoms, but the fistula is then lost. There are many surgi-
of AVAIS that occurs when a nutrient artery to a nerve is cal procedures described to treat AVAIS, including banding
5 • Access for Renal Replacement Therapy 79

of the access to limit the flow, which can be performed with


a ligature; proximalization of access inflow (PAI); distal
revascularisation interval ligation (DRIL); revision using
distal inflow (RUDI); and DRAL.

ARTERIOVENOUS FISTULA SURVEILLANCE AND


MAINTENANCE
Fistula surveillance aims to detect fistula dysfunction so that
timely intervention can be performed to prevent loss of patency.
Fistula surveillance is recommended in vascular access guide-
lines, and there is now good evidence that it improves patency,
reduces thrombosis, and is cost effective.68,69 Surveillance can
be carried out clinically by assessing for the presence of promi-
nent collateral veins, arm swelling, and prolonged bleeding.
More sophisticated methods such as monitoring venous pres-
sures during dialysis, measuring recirculation, and calculat-
ing dialysis adequacy (Kt/V) appear to improve AVF patency
but are not sensitive, and early meta-analysis data showed
a nonsignificant benefit.58,59,70 However, more advanced
methods of surveillance that measure access flow such as
ultrasound dilution, (e.g., transonic and Doppler ultrasound)
have been shown in an RCT to reduce thrombosis, improve
primary and secondary patency, and reduce cost compared
with conventional surveillance.71 Fig. 5.11 Illustration of the anatomy and location of the HeRO
There is no compelling evidence to support the addi- device. The venous outflow is depicted in the right atrium through
tion of any medical therapies to improve AVF patency or insertion via the internal jugular vein. The outflow is tunneled subcu-
prevent failure. There have been a number of trials com- taneously and connected to the arterial graft, which is anastomosed to
paring placebo with different therapies including aspirin, the brachial artery.
clopidogrel, dipyridamole, fish oil, human type-I pancreatic
elastase, ticlopidine, sulfinpyrazone, and warfarin. A meta- Conventional options for such patients would be a CVC
analysis of these interventions failed to demonstrate any through the stenosis or a lower limb procedure. The HeRO
advantage compared with placebo, except with ticlopidine graft was developed to extend the use of upper limb access
(an antiplatelet), but ticlopidine has been taken off the mar- while avoiding the complications of a central catheter. The
ket.72 The only large trial included in this meta-analysis HeRO device consists of a 6 mm diameter, nitinol reinforced
recruited 887 patients who were randomized to placebo silicon “outflow” that is placed through a central venous
or clopidogrel. The clopidogrel group had fewer episodes of lesion into the upper third of the right atrium. It is tunneled
thrombosis, but there was no difference in AVF suitability from its entry point to the clavipectoral space where it con-
for dialysis.25 Three trials have compared aspirin with pla- nects onto a PTFE graft via a unique titanium connector.
cebo and they all have small numbers and variable method- The PTFE graft is tunneled subcutaneously and anasto-
ology. Two of the trials suggest aspirin is beneficial in terms mosed onto an inflow artery, usually the brachial artery
of patency; however, this important question has yet to be (Fig. 5.11). The result is blood flow directly from the artery,
answered and would be an excellent topic for a large-scale through the PTFE graft, and into the right atrium via the
multicenter RCT. outflow component. Although there have been more than
10,000 insertions worldwide, only about 250 have been
ADVANCES IN VASCULAR ACCESS outside the US. A recent meta-analysis demonstrated
24-month mean secondary patency of 59.4% (39.4%–
Over the past few years there have been a number of excit- 78%), 2.8 interventions per graft per year to retain patency,
ing technologic advancements that have occurred to pro- and a mean infection rate of 10.1% (2.5%–21%).47
vide patients with definitive vascular access in clinical
situations where, historically, access may have been impos- Surfacer Inside-Out Catheter System
sible to achieve. Two such technologies include the HeRO The Surfacer Inside-Out Catheter System is a recent tech-
graft (Merit Medical) and the Surfacer Inside-Out Catheter nologic innovation that provides a safe and reliable way
System (Merit Medical). Endovascular techniques to create to reestablish central venous access (CVA) in patients who
AV fistulas have also been introduced such as the everlinQ have central venous occlusions.
endoAVF (TVA Medical). CVA is vital for many patients who require life-sustain-
ing therapies such as hemodialysis, chemotherapy, and
HeRO (Hemodialysis Reliable Outflow) Graft parenteral nutrition. At present it is estimated that 3.5
Patients with problematic upper limb access due to cen- million people in Europe are dependent on a CVA device
tral venous stenosis or occlusion will either require access (CVAD). CVA is typically obtained by puncturing one of
ligation to alleviate symptoms of venous hypertension or the four large upper body veins, including the right or
need angioplasty and/or stent placement to retain access. left internal jugular veins or the right or left subclavian
80 Kidney Transplantation: Principles and Practice

veins. Long-term and repeated access to these veins car- Peritoneal Dialysis
ries increased risk of central vein occlusions. Chronic
venous occlusions occur when thrombus forms around In the UK in 2015, approximately 13% of dialysis patients
a long-term CVAD and organizes into a dense thrombus were using peritoneal dialysis (PD).1 Over recent years there
or fibrous tissue that obliterates the lumen. When this has been a consistent decrease in the number of patients in
occurs there is no effective, low-risk procedure to address the UK with renal failure commenced on PD as their first
the problem. Patients end up with lines in the groin region treatment modality. This is in part due to an increase in
that are associated with higher infective complications or preemptive renal transplantation, but other reasons impli-
require complicated and time-consuming radiologic pro- cated in the decline are the structural organization of dialy-
cedures to recanalize the occluded veins or to place lines sis facilities and arrangements for PD catheter insertion. In
via nonconventional routes such as translumbar or trans­ the US less than 10% of dialysis patients are on PD (United
hepatic vessels. States Renal Data System [USRDS]), despite most nephrol-
In conventional CVA procedures, a needle is directed ogists believing this figure should be at least 40%.75
inward through the skin under ultrasound guidance toward The concept behind PD is straightforward. The peri-
a central vein. In patients with normal central venous anat- toneum, with a total surface area of 2 m2, is composed of
omy, this carries risks even in experienced hands, and com- endothelium, interstitium, and mesothelium and can act
plication rates have been reported as high as 10% including as an efficient semipermeable membrane. Infusing a hyper-
arterial puncture, nerve injury, pneumothorax, and cath- tonic dialysate fluid into the peritoneal cavity allows ultra-
eter malposition. An inability to cannulate the vessel may filtration of solutes and electrolytes.
also occur in >15% of procedures. When the central veins PD may have certain advantages over hemodialysis. Some
are narrowed or occluded, this risk rises even further and studies have described improved survival in PD patients, par-
often the procedure has to be abandoned and access is not ticularly in the first 1 to 2 years after commencing RRT.76,77
possible. Radiologic techniques are available but require However, after 1.5 to 2 years undergoing dialysis, the risk of
significant time (4 hours) and highly skilled clinicians to death in PD patients becomes equivalent to that in hemodial-
reestablish access; however, success rates are variable with ysis patients, and when comparing patients suitable for both
up to 50% failure rates reported. modalities, mortality is comparable.78 The reasons for this
The Surfacer Inside-Out Access Catheter System pro- improved survival have yet to be clarified, although mainte-
vides a novel approach to establish CVA for patients who nance of residual renal function may be a factor, because this
have occluded. The Surfacer system provides a safe and is better preserved in PD patients compared with those on
repeatedly reliable means of achieving CVA and uses the hemodialysis. PD patients usually report greater satisfaction
same principle technique as conventional radiologic pro- than those on hemodialysis, which may relate to the greater
cedures of tunelling or bypassing the occlusion; however, autonomy afforded by PD. Furthermore, the cost of PD per
it does so in a reverse direction. A guidewire is inserted patient per year is approximately one-third less than that of
through the femoral vein and navigated up the body to hemodialysis.79 In addition, there is some evidence that sur-
the point of the venous occlusion. The device is passed vival after transplant is superior and rates of delayed graft
over the wire and establishes a transient passage across function (DGF) are lower in patients undergoing PD com-
the occlusion to the supraclavicular region. An exit target pared with hemodialysis.80
on the skin provides the means for fluoroscopic guidance However, the major drawback of PD is technique fail-
of a needle wire that passes from within the device that ure, which remains high. A recent multicenter prospective
is inside the vessel to the marked exit point on the skin. study found that 25% of PD patients transferred to hemodi-
The wire essentially passes from “inside to outside” and alysis, 70% within the first 2 years after commencing PD.81
“surfaces” through the skin enabling a peelable introducer Despite reductions in peritonitis, infection remains the pri-
to be placed over the wire and pulled back into the central mary cause of technique failure.
veins to provide access.73 This section of the chapter will focus on specific clinical
aspects of PD; however, comprehensive guidelines covering
Endovascular Arteriovenous Fistula PD initiation, maintenance, management of complications,
The everlinQ endoAVF (TVA Medical) is an endovascular and governance are published by the Renal Association for
technique that creates an AVF by aligning two magne- those areas not covered.82
tized catheters in an adjacent artery and vein and using
thermal energy to create the anastomosis. The theory ACUTE PERITONEAL DIALYSIS
is that unlike during surgical AVF, endovascular tech-
niques can avoid manipulation of vessels, which may There has been a recent increase in the use of PD in patients
lead to neointimal hyperplasia and stenosis. Novel endo- requiring RRT as a result of acute kidney injury (AKI),
vascular access trial (NEAT) study recruited 80 patients although the evidence base for this is not new. In the devel-
who underwent the procedure using the ulnar artery and oping world it is a more cost effective and straightforward
ulnar vein. The maturation rate of 87%, 1 year primary option than hemodialysis and can be successfully per-
patency of 67%, and 1 year functional patency of 64% formed on patients presenting with AKI due to a breadth of
were comparable to that of surgically created fistulas.74 causes. In the developed world the majority of studies have
Such technology may have a place in certain circum- recruited patients from intensive care, and in a recent meta-
stances, but potential disadvantages such as equipment analysis of 24 studies (n = 1556 patients) there was no
cost, device failures, and complications associated with difference in mortality or outcomes between the two modal-
arterial and venous puncture should be considered. ities.83 In two RCTs PD showed a reduction in time to renal
5 • Access for Renal Replacement Therapy 81

recovery (5.5 vs. 7.5 days) and better correction of acidosis TABLE 5.1 Contraindications to Peritoneal Dialysis
and a significant cost saving whereas venovenous filtration
showed improved fluid balance.84,85 Catheter insertion in Absolute Contraindications Relative Contraindications
acutely unwell patients with fluid overload and hyperkale- Encapsulating peritoneal sclerosis Severe obesity
mia may not be possible under general anesthesia, which Inflammatory bowel disease Severe peritoneal adhesions
may exclude those unwilling to undergo a LA procedure Large irreparable abdominal Large abdominal wall hernias
hernia Abdominal stomas
or those in whom anatomy or body habitus precludes a LA Chronic obstructive airway disease
approach. Psychosocial factors likely to result
in poor compliance
Physical disability
PERITONEAL DIALYSIS DELIVERY SYSTEMS AND Learning disability
CATHETERS
PD is a closed-loop system comprising dialysate fluid, a
delivery system, and an indwelling peritoneal catheter. gram-negative PD peritonitis. Although PD can be per-
Fluid is infused under gravity from a reservoir of dialysate. formed in patients with stomas, there is a predisposition to
Luer-Lok or rotating self-lock devices have been devised to infection. Abdominal wall hernias may enlarge in patients
connect the dialysate bag with the delivery system for ease receiving PD and should, if possible, be repaired before or at
of connection and sterility. The Y delivery systems are the the time of catheter insertion. Table 5.1 lists the relative and
most commonly used. The single branch of the Y is con- absolute contraindications to PD catheter insertion.
nected to the indwelling peritoneal catheter via an inert tita- A variety of techniques for catheter insertion have been
nium connector and the upper two branches are connected described including open surgical, percutaneous, perito-
to the dialysate reservoir and an empty bag. This configura- neoscopic, and laparoscopic.
tion allows complete drainage of any contaminating dialy- In the open technique, the catheter is introduced via a
sate fluid before infusion of sterile, fresh fluid through the small vertical infraumbilical incision placed in the mid-
indwelling catheter. Several RCTs have shown the superi- line or paramedian position. Before positioning, the cath-
ority of various Y systems over conventional PD systems in eter should be flushed and immersed in saline because wet
reducing the incidence of infective complications.86 cuffs stimulate more rapid tissue ingrowth. A small incision
is made in the peritoneum, and the tube is inserted using
blunt forceps with or without a metal stylet within the
CATHETER SELECTION catheter lumen. The tube tip must be placed in the recto-
PD catheters should be soft, flexible, atraumatic, radi- vesical pouch in men and the rectovaginal pouch of Doug-
opaque, and relatively inert. Several different catheters las in women (Fig. 5.12). The peritoneum is closed with
are available but the Tenckhoff catheter is the most popu- an absorbable suture around the cuff to create a water-
lar. The original Silastic Tenckhoff design was a straight, 5 tight seal, and the linea alba or rectus sheath closed with a
mm external diameter tube, with two Dacron cuffs and a nonabsorbable suture. The extraperitoneal segment of the
perforated intraperitoneal segment. Many variations of the catheter is tunneled subcutaneously and brought out at a
Tenckhoff device exist, including catheters with single cuffs conveniently placed lateral exit site. At the end of the pro-
and coiled intraperitoneal ends. A number of randomized cedure the catheter should be flushed to ensure free inward
trials have compared straight versus coiled PD catheters and outward flow of dialysate fluid. Two RCTs have com-
with a meta-analysis demonstrating no significant differ- pared a midline versus lateral insertion site for PD catheters,
ence in the risk of peritonitis, exit site or tunnel infection, with a meta-analysis showing no significant difference in
or catheter removal.87 A further meta-analysis did demon- risk of peritonitis, exit or tunnel site infection, or mortal-
strate an increased risk of catheter tip migration with coiled ity.87 Catheter removal was reported in one trial and shown
catheters, but this was not associated with an increased to be significantly reduced with midline insertion.92,93
risk of catheter failure.88,89 A single randomized trial has Over recent years, laparoscopic insertion of PD catheters
compared single versus double-cuffed catheters and again has superseded the open surgical technique in a number
shown no significant difference in the risk of peritonitis, exit of centers. Laparoscopy provides the ability to assess and
site or tunnel infection, or catheter removal.90 However, a address anatomic problems that may result in mechani-
large comparative study based on Canadian registry data cal obstruction such as adhesions, and allows placement
did show a reduced risk of S. aureus peritonitis in patients of the catheter in the correct position in the pelvis under
with double-cuffed catheters.91 direct vision. There are a number of techniques for lapa-
roscopic PD catheter insertion described in the literature.
The principles are as follows. A 5- or 10-mm camera port is
CATHETER INSERTION placed in either the left or right upper quadrant so as not to
Most patients are suitable for PD; the only absolute con- interfere with the catheter insertion site. The tunnel for the
traindications are severe peritoneal adhesions, inflamma- catheter can be created either using a further 5-mm port or
tory bowel disease, and previous encapsulating peritoneal specific kits containing a needle with an expandable plastic
sclerosis. Obesity, advanced age, abdominal hernias, sto- sheath and serial dilators (Fig. 5.13). A further port can be
mas, and chronic obstructive pulmonary disease are rela- inserted to allow manipulation of the catheter into the pel-
tive contraindications. Severe colonic diverticulosis may vis if required. Once the catheter is in a suitable position the
increase the translocation of gut organisms, and there pneumoperitoneum is released and the subcutaneous tun-
is a strong association between diverticular disease and nel created.
82 Kidney Transplantation: Principles and Practice

Peritoneum

Dialysis fluid

Drainage fluid Dialysis fluid

Fig. 5.12 Sagittal section demonstrating the optimal position for a peritoneal dialysis tube located in the pouch of Douglas. The diagram also
illustrates a Y-delivery system for PD fluid.

contrast under fluoroscopic control. A guidewire is then


placed and serial dilators passed to create a tunnel. A peel-
away sheath is then introduced into the peritoneal cavity,
through which the catheter is passed, before the sheath is
removed and the catheter tunneled subcutaneously. In the
peritoneoscopic technique, rather than using fluoroscopic
guidance, the position within the peritoneal cavity is con-
firmed using a 2.2-mm telescope introduced down the peel-
away sheath.
Three randomized controlled trials and eight cohort
studies have compared laparoscopic with open insertion
of PD catheters,94–96 and one RCT has compared perito-
neoscopic with open insertion.97 A meta-analysis of these
trials showed no significant differences in risk of mortal-
ity, peritonitis, hernia, or exit site or tunnel infection.
However, catheter migration and catheter failure were
significantly reduced in the laparoscopic group although
bleeding was more frequent.98 The LOCI-1 trial compar-
Fig. 5.13 YTec insertion kit photographic image showing the compo-
ing open versus laparoscopic PD catheter insertion has
nents of the kit, which include from left to right: insertion trocar with been submitted for publication but struggled to recruit
plastic sheath, medium and large sized tract dilators, cuff insertion because of a reduction in number of PD patients and lack
device, and subcutaneous tunelling device. The single-cuff PD tube of equipoise among surgeons.99 LOCI-2 is a national,
with metal insert is at the top of the photograph. prospective registry study that follows from LOCI-1, but
surgeons can select the catheter insertion technique (per-
The percutaneous and peritoneoscopic techniques are sonal correspondence).
both variations on the Seldinger technique and use a simi- No RCTs have been conducted to compare percutane-
lar kit to that described earlier for use in the laparoscopic ous techniques with other techniques. Two comparative
technique. In the percutaneous technique, ultrasound studies of percutaneous versus open surgical insertion of
guidance can be used to determine a safe puncture site PD catheters have been reported; one demonstrated no
where there is maximum separation between bowel and difference between the techniques,100 the other reported
the anterior abdominal wall. After puncture, the posi- a significantly higher early leak rate in the percutaneous
tion within the peritoneum can be confirmed by injecting group.101
5 • Access for Renal Replacement Therapy 83

COMPLICATIONS ASSOCIATED WITH of guidewire manipulation to restore catheter function is an


PERITONEAL DIALYSIS CATHETERS indication for laparoscopic intervention. This allows direct
visualization of the cause of catheter obstruction and defini-
Bleeding tive treatment including catheter repositioning, adhesioly-
Bloody fluid is a common finding, occurring in 30% of sis, omentectomy, or omentopexy.
patients for the first few catheter exchanges after insertion.
The bleeding most often arises from small vessels on the peri-
Pericatheter Leak
toneal surface at the catheter entry site and usually stops Any variable that predisposes to poor wound healing (e.g.,
within 24 hours. If bleeding does occur, frequent flushing of steroids, obesity, malnutrition, diabetes) may culminate
the catheter or, if possible, low-volume exchanges reduces in pericatheter leakage, rates of which have been reported
the rate of catheter obstruction by clots.102 Bleeding long between 2% and 25%.109 Choice of surgical technique may
after insertion may occur with encapsulating peritoneal determine leak rates; leaks are said to be more common
sclerosis. with midline catheter insertion compared with lateral inser-
tion,110 and open compared with laparoscopic, although
Pain that is not supported by meta-analysis data.109 Pericatheter
The first attempts at dialysate infusion can produce discom- leakage allows fluid extravasation around the catheter or
fort. This is more common with straight catheters when accumulation in the abdominal wall.
infusion pressure is greatest. With coiled catheters pain is When an early postoperative leak develops, dialysate
less likely because dialysate flows through the side perfora- exchange should be stopped for 2 to 4 weeks, which may
tions. The pain is usually temporary and resolves within a necessitate a temporary switch to hemodialysis. Late-onset
few weeks. Slower infusion rates and incomplete drainage leaks usually require catheter replacement. Pericatheter
may alleviate symptoms. leaks associated with herniation should be treated by cath-
eter removal and hernia repair.
Cuff Extrusion
The most important factor for cuff extrusion is the depth
Hernias
at which the subcutaneous cuff is implanted; at least 2 cm The increased intraabdominal pressures after infusion of
below the skin is required. Tension on the extraperitoneal dialysate can enlarge preexisting hernias, so it is best to
portion of the catheter, such as during bag exchange, can repair these preemptively before commencement of PD.
bring a poorly implanted cuff to the surface, or it may relate However, simultaneous catheter insertion and hernia
to an exit site or tunnel infection. In the absence of infec- repair is also safe and feasible.111 The reported prevalence
tion, if a cuff is exposed or very superficial it can be excised of de novo abdominal hernias in PD patients is 2.5% to
to aid healing (Fig. 5.14A). 25%.111–114 One study showed 40% are umbilical, 33%
are inguinal, and 19% are incisional. After surgical repair
Catheter Obstruction 86% of patients continued PD successfully and only 7 of 73
Catheter obstruction is usually due to outflow obstruction patients required temporary hemodialysis.113 The pressure
that can be extrinsic or related to catheter positioning. of dialysate can cause recanalization of a patent processus
Clotted blood may collect in the distal portion of the cath- vaginalis, which manifests as scrotal or rarely, labial edema.
eter shortly after surgery; this can be treated effectively Surgical ligation is necessary, with a postoperative regimen
with a per-catheter infusion of heparin, streptokinase, or of low-volume, high-frequency dialysate exchanges until
urokinase.103,104 healing has occurred. The use of a prosthetic mesh is advis-
Extrinsic compression causing obstruction can be due to able and does not necessitate withholding PD unless there is
bladder distension, an impacted sigmoid colon, or uterine clear leak using low-volume, frequent exchanges.114
fibroids. Constipation is a very common cause of catheter
malfunction and laxatives should always be used for initial
Exit-Site and Tunnel Infections
management of poor drainage even if there is no history As lone entities, exit site and tunnel infections pose little
or radiologic evidence of constipation. Omental wrapping risk, but the possibility of developing PD peritonitis demands
remains the most common cause of refractory catheter careful attention to the infections; PD peritonitis occurs
obstruction responsible in 35% to 80% of cases. All of these in approximately 12% of patients with exit-site or tunnel
causes may result in catheter tip migration out of the pel- infections.
vis. However, not all migrated catheters become obstructed, An exit-site infection is defined by the presence of puru-
and conversely, catheters well positioned in the pelvis may lent drainage, with or without erythema of the skin (Fig.
still become obstructed. Approximately half of all catheters 5.14B). Reported rates of exit-site infections range from
migrate to some extent over time, but only 20% of migrated 0.05 to 1.02 episodes per patient per year.89 Erythema at
catheters malfunction.105 the exit site without purulent drainage can be an early indi-
Persisting catheter obstruction after treatment for consti- cation of infection but can also be a simple skin reaction,
pation is an indication for manipulation, either fluoroscopi- particularly in newly inserted catheters. A positive culture
cally using a stiff guidewire or surgically. Initial success rates from an exit site in the absence of purulent drainage repre-
for guidewire manipulation have been reported between sents colonization rather than infection and is not an indi-
78% and 85%. However, this improvement in catheter cation for treatment. A tunnel infection may present with
patency is temporary, and patency rates after 30 days are erythema, edema, or tenderness over the subcutaneous
in the range of 29% and 85%; this is probably because the portion of the catheter; however, it is often clinically occult
underlying problem has not been addressed.106–108 Failure and only detectable on ultrasound scanning (Fig. 5.15).
84 Kidney Transplantation: Principles and Practice

A B
Fig. 5.14 (A) Photographic image of an extruded superficial cuff which may be due to infection or positioning of the cuff too close to the exit site and
failure of healing. (B) Photographic image of an exit site infection with surrounding erythema and a purulent exudate.

TABLE 5.2 Treatment of Exit Site Problems and


Infections
Problem Treatment
Erythema alone, no discharge Topical chlorhexidine, mupirocin,
hydrogen peroxide
Overgranulation Silver nitrate cauterization
Gram-positive infection Flucloxacillin, co-amoxiclav,
vancomycin, rifampicin, cepha-
Fig. 5.15 Photographic image of an early tunnel infection. There is losporins
erythema tracking along the course of the catheter from the exit site, Gram-negative infection Ciprofloxacin, gentamicin
without a purulent discharge. Pseudomonas infection Ciprofloxacin plus another agent
(e.g., cephalosporin) and cath-
eter removal
Fungal infection Catheter removal with systemic
The majority of exit-site and tunnel infections are caused antifungal agent
by S. aureus and Pseudomonas aeruginosa; nasal carriage of
S. aureus increases the risk of exit-site infection four-fold.
Table 5.2 summarizes the recommended management of diarrhea. Localized pain or tenderness should raise suspi-
exit-site problems and infections. cions of surgical pathology such as appendicitis, cholecys-
titis, or diverticulitis.
Peritoneal Dialysis Peritonitis The causative organisms in PD peritonitis generally dif-
Peritonitis is the most significant complication of PD and is fer from the organisms causing “surgical” peritonitis. In the
the second most common cause of mortality in PD patients. latter case, infections are usually polymicrobial, consisting
Incidence from large audits range from 0.16 to 0.6 episodes of aerobic and anaerobic bacteria, whereas a single micro-
per patient per year.115,116 The International Society for organism is usually isolated in primary PD peritonitis. S.
Peritoneal Dialysis (ISPD) guidelines give an acceptable aureus, Staphylococcus epidermidis, and Streptococcus species
peritonitis rate of 0.5 episodes per year.89,117 Although less account for 60% to 80% of cases with coagulase-negative
than 5% of peritonitis episodes result in death, peritonitis is staphylococci constituting 30% to 40% and streptococci
a contributory factor in 16% of deaths in PD patients and constituting 10% to 15%. Yeast, such as Candida spp. are
can lead to failure of the peritoneal membrane.89 At least a the most common cause of fungal peritonitis, entering via
quarter of episodes culminate in catheter removal.116 The the catheter or commonly per vaginam.
most common portal of entry for infection is the exit site Dialysate samples for microbiologic examination should
before wound healing. be taken from the first cloudy bag for culture and antibiotic
The first indication of peritonitis is the presence of cloudy sensitivities. Negative culture rates vary significantly with
effluent containing >100×106/L leukocytes. Abdominal historical values as high as 50% although a large registry
pain usually follows, and abdominal tenderness is typically series of more than 6000 patients showed a rate of 13%.89,118
generalized and often associated with rebound tenderness. ISPD guidelines state that if more than 15% of cases are
Other signs include pyrexia, nausea and vomiting, and culture negative, the process of specimen testing should
5 • Access for Renal Replacement Therapy 85

be reevaluated.89 Other investigations include abdominal


and chest radiographs to check for catheter position and air
under the diaphragm, although pneumoperitoneum is not
attributable to perforation in all patients on PD.
To prevent delay in treatment, antibiotic therapy should
be commenced as soon as cloudy effluent is seen and dialy-
sate has been sent for culture. Empiric therapy must cover
both gram-negative and gram-positive organisms. Regi-
mens tend to be center-specific dependent on local antibi-
otic sensitivities, but should include gram-negative cover
by a third generation cephalosporin or aminoglycoside and
gram-positive cover by vancomycin or a cephalosporin.
Intraperitoneal administration is superior to intravenous
dosing.82 Once culture and sensitivity results are available,
antibiotic therapy can be adjusted to narrow-spectrum
agents as appropriate. In polymicrobial peritonitis, particu-
larly in association with anaerobic bacteria, the risk of death Fig. 5.16 Computed tomography image illustrating the characteris-
is increased, and an urgent surgical evaluation should be tic radiologic finding in encapsulating peritoneal sclerosis. Note the
sought because early laparotomy may reduce mortality.119 cocoon of small bowel in the left iliac fossa.
The majority of episodes of PD peritonitis will respond to
antibiotic therapy and in mild cases, patients do not require
hospital admission. However, there are circumstances The prevalence of EPS is between 0.5% and 4.4%, and
where catheter removal is required, as outlined in Table its incidence increases with the duration of PD. Reported
5.2. The majority of patients undergo laparotomy and cath- mortality rates are high (25%–55%), especially during the
eter removal, with concomitant peritoneal washout. How- first year after diagnosis, and increase with the duration of
ever, laparoscopic removal and washout has been shown PD. Patients may present with abdominal pain, a decline
to be as effective as the open technique, but associated with in net ultrafiltration, ascites, bloody effluent (7%–50%),
less postoperative pain and bowel dysfunction.120 bowel obstruction, vomiting, malnutrition, or an abdomi-
Refractory peritonitis is defined as failure of the efflu- nal mass.125
ent to clear after 5 days of appropriate antibiotic therapy. The pathogenesis involves loss of the mesothelial layer of
Prolonged attempts to medically treat refractory peritoni- the peritoneum with submesothelial thickening as a result
tis are associated with extended hospital stay, peritoneal of sclerosis and fibrosis. A number of factors have been
membrane damage, increased risk of fungal peritonitis, implicated in the development of EPS, including duration of
and death.121 Relapsing peritonitis is defined as an epi- PD, composition of dialysate solutions, peritonitis episodes,
sode with the same organism that occurs within 4 weeks and genetic predisposition.125,126
of completion of therapy and is more likely to require cath- The diagnosis is confirmed by radiologic studies. Ultraso-
eter removal than uncomplicated PD peritonitis.122 Fungal nography may reveal bowel wall thickening, a thick-walled
peritonitis is often serious with a high mortality and cath- mass containing bowel loops, loculated ascites, and fibrous
eter removal is indicated immediately after fungi are iden- adhesions. Computed tomography demonstrates perito-
tified by microscopy or culture. Mycobacterium infection is neal enhancement, thickening and calcification associated
a particular problem in at-risk cohorts. Diagnosis can be with adhesions, bowel obstruction, fluid loculations, and a
difficult, but should be suspected in the presence of per- cocoon of bowel (Fig. 5.16).127
sistently elevated mononuclear cell counts with negative The mainstay of treatment for patients with EPS should
cultures. Acid-fast bacilli smears of dialysate fluid may be be supportive care with enteral or parenteral nutrition.
negative in 90% of cases, but formal cultures are usually Surgery to remove all fibrotic tissue and free the bowel is a
positive. Treatment consists of long-term antituberculous major undertaking, with a mean operative time of 7 hours
therapy. Catheter removal is usually undertaken, although and a mortality of around 7%. However, bowel function
it is not considered necessary for cure. Mortality attributed is restored in 96% of patients, although 25% of patients
to tuberculous peritonitis is around 15%, and much of this will require repeat surgery. To try and prevent recurrence
may relate to treatment delay. patients may be treated with immunosuppressive agent
After a severe episode of PD peritonitis necessitating cath- postoperatively, most commonly corticosteroids. Other
eter removal, less than half of patients are able to return to drug treatments used in the treatment of EPS include
PD, and only a third will remain on PD for more than 1 tamoxifen and ACE inhibitors, although strong evidence for
year. Reasons for technique failure include adhesions and their benefit is lacking.
permanent membrane failure.123
Encapsulating Peritoneal Sclerosis
RENAL TRANSPLANT ISSUES WITH
Encapsulating peritoneal sclerosis (EPS) is a rare, but PERITONEAL DIALYSIS
potentially life-threatening complication of PD. The defini-
tion of EPS is signs of intermittent and persistent or recur- Active PD peritonitis is an absolute contraindication to
rent gastrointestinal obstruction, with macroscopic and/ transplantation, although it may be safe to proceed if the
or radiologic evidence of sclerosis, calcification, peritoneal patient has had recent infections but several days of intra-
thickening, or encapsulation of the intestines.124 peritoneal antibiotics with clear effluent. Previous studies
86 Kidney Transplantation: Principles and Practice

have highlighted the risks of infection after renal transplan- 14. Janne d’Othee B, Tham JC, Sheiman RG. Restoration of patency
tation although the values are low.128 in failing tunneled hemodialysis catheters: a comparison of cath-
eter exchange, exchange and balloon disruption of the fibrin
Some studies have demonstrated a high incidence of sheath, and femoral stripping. J Vasc Interv Radiol 2006;17(6):
posttransplantation EPS, typically developing within 1 year 1011–5.
of the procedure.129 Whether this is related to discontinu- 15. Hernandez D, Diaz F, Rufino M, et al. Subclavian vascular access ste-
ing PD or to the transplantation process remains unclear. nosis in dialysis patients: natural history and risk factors. J Am Soc
Nephrol 1998;9(8):1507–10.
A few studies have shown less delayed graft function 16. Schillinger F, Schillinger D, Montagnac R, Milcent T. Post-catheter-
after renal transplant in patients on PD compared with ization venous stenosis in hemodialysis: comparative angiographic
those on hemodialysis, which may relate to volume status study of 50 subclavian and 50 internal jugular accesses. Nephrolo-
at the time of transplantation or residual native renal func- gie 1992;13(3):127–33.
tion.130 A further study has also demonstrated significantly 17. Mansour M, Kamper L, Altenburg A, Haage P. Radiological cen-
tral vein treatment in vascular access. J Vasc Access 2008;9(2):
lower all-cause mortality in transplanted patients on PD 85–101.
compared with those on hemodialysis.131 18. Miller LM, Clark E, Dipchand C, et al. Hemodialysis tunneled
Catheter removal in a PD patient undergoing kidney catheter-related infections. Can J Kidney Health Dis 2016;3:
transplant can be at the time of the procedure for live donor 2054358116669129.
19. Lok CE, Mokrzycki MH. Prevention and management of catheter-
recipients as the risk of DGF is very low. In deceased donor related infection in hemodialysis patients. Kidney Int 2011;79(6):
transplant recipients catheter removal should be as early as 587–98.
is feasible. Routine removal at 4 weeks in patients with a 20. Vascular Access Work Group. Clinical practice guidelines for vascu-
functioning transplant minimizes potential infections. lar access. Am J Kidney Dis 2006;48(Suppl. 1):S248–73.
21. Vascular Access 2006 Work Group. Clinical practice guidelines for
vascular access. Am J Kidney Dis 2006;48(Suppl. 1):S176–247.
22. Jadlowiec CC, Lavallee M, Mannion EM, Brown MG. An out-
Conclusion comes comparison of native arteriovenous fistulae, polytetrafluo-
rethylene grafts, and cryopreserved vein allografts. Ann Vasc Surg
Optimal physical and psychological health for the dialy- 2015;29(8):1642–7.
23. Murad MH, Elamin MB, Sidawy AN, et al. Autogenous versus pros-
sis patient can be achieved by multidisciplinary care, and thetic vascular access for hemodialysis: a systematic review and
central to this is effective dialysis access. Early planning of meta-analysis. J Vasc Surg 2008;48(Suppl. 5):34S–47S.
access procedures permits a smooth transition from predi- 24. Maya ID, O’Neal JC, Young CJ, Barker-Finkel J, Allon M. Outcomes
alysis to dialysis and minimizes the use of temporary venous of brachiocephalic fistulas, transposed brachiobasilic fistulas, and
catheters. Furthermore, effective dialysis access provides a upper arm grafts. Clin J Am Soc Nephrol 2009;4(1):86–92.
25. Dember LM, Beck GJ, Allon M, et al. Effect of clopidogrel on early
lifeline for an ever-increasing group of patients in whom failure of arteriovenous fistulas for hemodialysis: a randomized con-
transplantation is deemed unsuitable. trolled trial. JAMA 2008;299(18):2164–71.
26. Huijbregts HJ, Bots ML, Wittens CH, et al. Hemodialysis arteriove-
References nous fistula patency revisited: results of a prospective, multicenter
initiative. Clin J Am Soc Nephrol 2008;3(3):714–9.
1. The Renal Association. 19th Annual Report. Nephron 2017;137
27. Almasri J, Alsawas M, Mainou M, et al. Outcomes of vascular access
(Suppl. 1):45–72.
for hemodialysis: a systematic review and meta-analysis. J Vasc Surg
2. Feest TG, Rajamahesh J, Byrne C, et al. Trends in adult renal replace-
2016;64(1):236–43.
ment therapy in the UK: 1982–2002. QJM 2005;98(1):21–8.
28. Schild AF, Schuman ES, Noicely K, et al. Early cannulation pros-
3. Vesely TM, Ravenscroft A. Hemodialysis catheter tip design: observa-
thetic graft (Flixene) for arteriovenous access. J Vasc Access
tions on fluid flow and recirculation. J Vasc Access 2016;17(1):29–39.
2011;12(3):248–52.
4. Moossavi S, Vachharajani TJ, Jordan J, Russell GB, Kaufman T, Moos-
29. National Kidney Foundation. NKF-DOQI clinical practice guidelines
savi S. Retrospective analysis of catheter recirculation in prevalent
for vascular access. National Kidney Foundation - Dialysis Outcomes
dialysis patients. Semin Dial 2008;21(3):289–92.
Quality Initiative 2006; 2006. Available online at: https://www.ki
5. Moist LM, Hemmelgarn BR, Lok CE. Relationship between blood flow
dney.org/sites/default/files/docs/12-50-0210_jag_dcp_guidelines-
in central venous catheters and hemodialysis adequacy. Clin J Am Soc
hd_oct06_sectiona_ofc.pdf.
Nephrol 2006;1(5):965–71.
30. Winearls CG, Fluck R, Mitchell DC. The organisation and deliv-
6. Gallieni M, Martina V, Rizzo MA, et al. Central venous catheters: legal
ery of the vascular access service for maintenance haemodialysis
issues. J Vasc Access 2011;12(4):273–9.
patients. Report of a joint working party. 2006. Available online at:
7. Niyyar VD, Chan MR. Interventional nephrology: catheter dys-
https://renal.org/wp-content/uploads/2017/06/Report_of_a_
function—prevention and troubleshooting. Clin J Am Soc Nephrol
Joint_Working_Party_on_Vascular_Access_September_2006.pdf.
2013;8(7):1234–43.
Accessed February 25, 2019.
8. Deitel M, McIntyre JA. Radiographic confirmation of site of central
31. Roy-Chaudhury P, Arend L, Zhang J, et al. Neointimal hyperplasia in
venous pressure catheters. Can J Surg 1971;14(1):42–52.
early arteriovenous fistula failure. Am J Kidney Dis 2007;50(5):782–
9. Lund GB, Trerotola SO, Scheel Jr PJ. Percutaneous translumbar
90.
inferior vena cava cannulation for hemodialysis. Am J Kidney Dis
32. Kokubo T, Ishikawa N, Uchida H, et al. CKD accelerates develop-
1995;25(5):732–7.
ment of neointimal hyperplasia in arteriovenous fistulas. J Am Soc
10. Matsuura J, Dietrich A, Steuben S, Ricker J, Barkema K, Kuhl T. Medi-
Nephrol 2009;20(6):1236–45.
astinal approach to the placement of tunneled hemodialysis catheters
33. Aitken E, Thomson P, Bainbridge L, Kasthuri R, Mohr B, Kingsmore
in patients with central vein occlusion in an outpatient access center.
D. A randomized controlled trial and cost-effectiveness analysis
J Vasc Access 2011;12(3):258–61.
of early cannulation arteriovenous grafts versus tunneled central
11. Po CL, Koolpe HA, Allen S, Alvez LD, Raja RM. Transhepatic Perm-
venous catheters in patients requiring urgent vascular access for
Cath for hemodialysis. Am J Kidney Dis 1994;24(4):590–1.
hemodialysis. J Vasc Surg 2017;65(3):766–74.
12. Kakkos SK, Topalidis D, Haddad R, Haddad GK, Shepard AD. Long-
34. Silva Jr MB, Hobson 2nd RW, Pappas PJ, et al. A strategy for increas-
term complication and patency rates of Vectra and IMPRA Carboflo
ing use of autogenous hemodialysis access procedures: impact of pre-
vascular access grafts with aggressive monitoring, surveillance and
operative noninvasive evaluation. J Vasc Surg 1998;27(2):302–7;
endovascular management. Vascular 2011;19(1):21–8.
discussion 307–8.
13. Tumlin J, Goldman J, Spiegel DM, et al. A phase III, randomized, dou-
35. Allon M, Lockhart ME, Lilly RZ, et al. Effect of preoperative sono-
ble-blind, placebo-controlled study of tenecteplase for improvement of
graphic mapping on vascular access outcomes in hemodialysis
hemodialysis catheter function: TROPICS 3. Clin J Am Soc Nephrol
patients. Kidney Int 2001;60(5):2013–20.
2010;5(4):631–6.
5 • Access for Renal Replacement Therapy 87

36. Lee KG, Chong TT, Goh N, et al. Outcomes of arteriovenous fistula 60. Nassar GM, Nguyen B, Rhee E, Achkar K. Endovascular treatment of
creation, effect of preoperative vein mapping and predictors of fistula the “failing to mature” arteriovenous fistula. Clin J Am Soc Nephrol
success in incident haemodialysis patients: a single-centre experi- 2006;1(2):275–80.
ence. Nephrology (Carlton). 2017;22(5):382–7. 61. Bent CL, Sahni VA, Matson MB. The radiological management of the
37. Lauvao LS, Ihnat DM, Goshima KR, Chavez L, Gruessner AC, Mills thrombosed arteriovenous dialysis fistula. Clin Radiol 2011;66(1):
JLS. Vein diameter is the major predictor of fistula maturation. J Vasc 1–12.
Surg 2009;49(6):1499–504. 62. Argyriou C, Schoretsanitis N, Georgakarakos EI, Georgiadis GS,
38. Aitken E, Jackson A, Kearns R, et al. Effect of regional versus local Lazarides MK. Preemptive open surgical vs. endovascular repair for
anaesthesia on outcome after arteriovenous fistula creation: a ran- juxta-anastomotic stenoses of autogenous AV fistulae: a meta-anal-
domised controlled trial. Lancet 2016;388(10049):1067–74. ysis. J Vasc Access 2015;16(6):454–8.
39. Cerneviciute R, Sahebally SM, Ahmed K, Murphy M, Mahmood W, 63. Bittl JA. Catheter interventions for hemodialysis fistulas and grafts.
Walsh SR. Regional versus local anaesthesia for haemodialysis arte- JACC Cardiovasc Interv 2010;3(1):1–11.
riovenous fistula formation: a systematic review and meta-analysis. 64. Haag-Weber M, Horl WH. Uremia and infection: mechanisms of
Eur J Vasc Endovasc Surg 2017;53(5):734–42. impaired cellular host defense. Nephron 1993;63(2):125–31.
40. Bylsma LC, Gage SM, Reichert H, Dahl SLM, Lawson JH. Arterio- 65. Yu VL, Goetz A, Wagener M, et al. Staphylococcus aureus nasal car-
venous fistulae for haemodialysis: a systematic review and meta- riage and infection in patients on hemodialysis. Efficacy of antibiotic
analysis of efficacy and safety outcomes. Eur J Vasc Endovasc Surg prophylaxis. N Engl J Med 1986;315(2):91–6.
2017;54(4):513–22. 66. Bachleda P, Utikal P, Kalinova L, et al. Infectious complications of
41. Koksoy C, Demirci RK, Balci D, Solak T, Kose SK. Brachiobasilic ver- arteriovenous ePTFE grafts for hemodialysis. Biomed Pap Med Fac
sus brachiocephalic arteriovenous fistula: a prospective randomized Univ Palacky Olomouc Czech Repub 2010;154(1):13–9.
study. J Vasc Surg 2009;49(1):171–7. e5. 67. Inston N, Mistry H, Gilbert J, et al. Aneurysms in vascular access: state
42. Leone JP, Glaser AD, Hufstetler R, Illig KA. Videoscopic basilic vein of the art and future developments. J Vasc Access 2017;18(6):464–
harvest for creation of transposed brachiobasilic arteriovenous fistu- 72.
lae. Vasc Endovascular Surg 2014;48(5–6):421–4. 68. Tordoir J, Canaud B, Haage P, et al. EBPG on vascular access.
43. Bashar K, Healy DA, Elsheikh S, et al. One-stage vs. two-stage bra- Nephrol Dial Transplant 2007;22(Suppl. 2):ii88–117.
chio-basilic arteriovenous fistula for dialysis access: a systematic 69. Sidawy AN, Spergel LM, Besarab A, et al. The Society for Vascular
review and a meta-analysis. PLoS ONE 2015;10(3):e0120154. Surgery: clinical practice guidelines for the surgical placement and
44. Huber TS, Carter JW, Carter RL, Seeger JM. Patency of autogenous and maintenance of arteriovenous hemodialysis access. J Vasc Surg
polytetrafluoroethylene upper extremity arteriovenous hemodialysis 2008;48(Suppl. 5):2S–25S.
accesses: a systematic review. J Vasc Surg 2003;38(5):1005–11. 70. Tessitore N, Lipari G, Poli A, et al. Can blood flow surveillance and
45. Ravani P, Palmer SC, Oliver MJ, et al. Associations between hemodi- pre-emptive repair of subclinical stenosis prolong the useful life of
alysis access type and clinical outcomes: a systematic review. J Am arteriovenous fistulae? A randomized controlled study. Nephrol Dial
Soc Nephrol 2013;24(3):465–73. Transplant 2004;19(9):2325–33.
46. Greenberg J, Jayarajan S, Reddy S, et al. Long-term outcomes of 71. Aragoncillo I, Abad S, Caldes S, et al. Adding access blood flow sur-
fistula first initiative in an urban university hospital—is it still rel- veillance reduces thrombosis and improves arteriovenous fistula
evant? Vasc Endovascular Surg 2017;51(3):125–30. patency: a randomized controlled trial. J Vasc Access 2017;18(4):
47. Al Shakarchi J, Houston JG, Jones RG, Inston N. A review on the 352–8.
Hemodialysis Reliable Outflow (HeRO) graft for haemodialysis vas- 72. Tanner NC, da Silva AF. Medical adjuvant treatment to improve the
cular access. Eur J Vasc Endovasc Surg 2015;50(1):108–13. patency of arteriovenous fistulae and grafts: systematic review and
48. Farber A, Tan TW, Hu B, et al. The effect of location and configura- meta-analysis. Eur J Vasc Endovasc Surg 2016;52(2):243–52.
tion on forearm and upper arm hemodialysis arteriovenous grafts. J 73. Ebner A, Gallo S, Cetraro C, Gurley J, Minarsch L. Inside-out upper body
Vasc Surg 2015;62(5):1258–64. venous access: the first-in-human experiences with a novel approach
49. Polo JR, Sanabia J, Garcia-Sabrido JL, Luno J, Menarguez C, Echena- using the Surfacer inside-out access catheter system. Endovasc Today;
gusia A. Brachial-jugular polytetrafluoroethylene fistulas for hemo- 2013. Available online at: https://evtoday.com/2013/06/inside-out-
dialysis. Am J Kidney Dis 1990;16(5):465–8. upper-body-venous-access/ [accessed 25.02.19].
50. Evans DC, Upton EC, Lawson JH. Axillary to common iliac arteriove- 74. Lok CE, Rajan DK, Clement J, et al. Endovascular proximal forearm
nous graft for hemodialysis access: case report and review of ‘exotic’ arteriovenous fistula for hemodialysis access: results of the prospec-
axillary-based grafts. J Vasc Access 2005;6(4):192–5. tive: multicenter Novel Endovascular Access Trial (NEAT). Am J Kid-
51. Glass C, Maevsky V, Massey T, Illig K. Subclavian vein to right atrial ney Dis 2017;70(4):486–97.
appendage bypass without sternotomy to maintain arteriovenous 75. Ledebo I, Ronco C. The best dialysis therapy? Results from an
access in patients with complete central vein occlusion, a new international survey among nephrology professionals. NDT Plus
approach. Ann Vasc Surg 2009;23(4):465–8. 2008;1(6):403–8.
52. Grima MJ, Vriens B, Holt PJ, Chemla E. An arterioarterial prosthetic 76. Heaf JG, Lokkegaard H, Madsen M. Initial survival advantage of peri-
graft as an alternative option for haemodialysis access: a system- toneal dialysis relative to haemodialysis. Nephrol Dial Transplant
atic review. J Vasc Access 2018;19(1):45–51. Available online at: 2002;17(1):112–7.
https://doi.org/10.5301/jva.5000808. 77. Weinhandl ED, Foley RN, Gilbertson DT, Arneson TJ, Snyder JJ,
53. Remuzzi G. Bleeding in renal failure. Lancet 1988;1(8596):1205–8. Collins AJ. Propensity-matched mortality comparison of incident
54. Allon M. Current management of vascular access. Clin J Am Soc hemodialysis and peritoneal dialysis patients. J Am Soc Nephrol
Nephrol 2007;2(4):786–800. 2010;21(3):499–506.
55. Allon M, Litovsky S, Young CJ, et al. Medial fibrosis, vascular calci- 78. Wong B, Ravani P, Oliver MJ, et al. Comparison of patient survival
fication, intimal hyperplasia, and arteriovenous fistula maturation. between hemodialysis and peritoneal dialysis among patients eli-
Am J Kidney Dis 2011;58(3):437–43. gible for both modalities. Am J Kidney Dis 2018;71(3):344–51.
56. Lee T, Chauhan V, Krishnamoorthy M, et al. Severe venous neointi- Available online at: https://doi.org/10.1053/j.ajkd.2017.08.028.
mal hyperplasia prior to dialysis access surgery. Nephrol Dial Trans- 79. Pike E, Hamidi V, Ringerike T, Wisloff T, Klemp M. More use of peri-
plant 2011;26(7):2264–70. toneal dialysis gives significant savings: a systematic review and
57. Tabbara M, Duque JC, Martinez L, et al. Pre-existing and postopera- health economic decision model. J Clin Med Res 2017;9(2):104–16.
tive intimal hyperplasia and arteriovenous fistula outcomes. Am J 80. Joachim E, Gardezi AI, Chan MR, Shin JI, Astor BC, Waheed S. Asso-
Kidney Dis 2016;68(3):455–64. ciation of pre-transplant dialysis modality and post-transplant out-
58. Tessitore N, Bedogna V, Poli A, et al. Should current criteria for comes: a meta-analysis. Perit Dial Int 2017;37(3):259–65.
detecting and repairing arteriovenous fistula stenosis be reconsid- 81. Jaar BG, Plantinga LC, Crews DC, et al. Timing, causes, predictors
ered? Interim analysis of a randomized controlled trial. Nephrol Dial and prognosis of switching from peritoneal dialysis to hemodialysis:
Transplant 2014;29(1):179–87. a prospective study. BMC Nephrol 2009;10. 3-2369-10-3.
59. Tessitore N, Bedogna V, Poli A, et al. Adding access blood flow sur- 82. Woodrow G, Fan SL, Reid C, Denning J, Pyrah AN. Renal Associa-
veillance to clinical monitoring reduces thrombosis rates and costs, tion clinical practice guideline on peritoneal dialysis in adults and
and improves fistula patency in the short term: a controlled cohort children. BMC Nephrol 2017;18(1). 333-017-0687-2.
study. Nephrol Dial Transplant 2008;23(11):3578–84.
88 Kidney Transplantation: Principles and Practice

83. Chionh CY, Soni SS, Finkelstein FO, Ronco C, Cruz DN. Use of peri- 104. Zorzanello MM, Fleming WJ, Prowant BE. Use of tissue plasminogen
toneal dialysis in AKI: a systematic review. Clin J Am Soc Nephrol activator in peritoneal dialysis catheters: a literature review and one
2013;8(10):1649–60. center’s experience. Nephrol Nurs J 2004;31(5):534–7.
84. Gabriel DP, Caramori JT, Martim LC, Barretti P, Balbi AL. High vol- 105. Ersoy FF, Twardowski ZJ, Satalowich RJ, Ketchersid T. A retrospec-
ume peritoneal dialysis vs daily hemodialysis: a randomized, con- tive analysis of catheter position and function in 91 CAPD patients.
trolled trial in patients with acute kidney injury. Kidney Int Suppl Perit Dial Int 1994;14(4):409–10.
2008;108:S87–93. 106. Kwon YH, Kwon SH, Oh JH, Jeong KH, Lee TW. Fluoroscopic guide
85. George J, Varma S, Kumar S, Thomas J, Gopi S, Pisharody R. Com- wire manipulation of malfunctioning peritoneal dialysis catheters
paring continuous venovenous hemodiafiltration and peritoneal initially placed by interventional radiologists. J Vasc Interv Radiol
dialysis in critically ill patients with acute kidney injury: a pilot 2014 Jun;25(6):904–10.
study. Perit Dial Int 2011;31(4):422–9. 107. Plaza MM, Rivas MC, Dominguez-Viguera L. Fluoroscopic manipu-
86. Daly CD, Campbell MK, MacLeod AM, et al. Do the Y-set and double- lation is also useful for malfunctioning swan-neck peritoneal cath-
bag systems reduce the incidence of CAPD peritonitis? A systematic eters. Perit Dial Int 2001;21(2):193–6.
review of randomized controlled trials. Nephrol Dial Transplant 108. Moss JS, Minda SA, Newman GE, Dunnick NR, Vernon WB, Schwab
2001;16(2):341–7. SJ. Malpositioned peritoneal dialysis catheters: a critical reap-
87. Strippoli GF, Tong A, Johnson D, Schena FP, Craig JC. Catheter praisal of correction by stiff-wire manipulation. Am J Kidney Dis
type, placement and insertion techniques for preventing perito- 1990;15(4):305–8.
nitis in peritoneal dialysis patients. Cochrane Database Syst Rev 109. Hagen SM, Lafranca JA, IJzermans JN, Dor FJ. A systematic review
2004;4:CD004680. and meta-analysis of the influence of peritoneal dialysis cath-
88. Xie J, Kiryluk K, Ren H, et al. Coiled versus straight peritoneal dialy- eter type on complication rate and catheter survival. Kidney Int
sis catheters: a randomized controlled trial and meta-analysis. Am J 2014;85(4):920–32.
Kidney Dis 2011;58(6):946–55. 110. Eklund BH, Honkanen EO, Kala AR, Kyllonen LE. Catheter configu-
89. Li PK, Szeto CC, Piraino B, et al. ISPD peritonitis recommenda- ration and outcome in patients on continuous ambulatory perito-
tions: 2016 update on prevention and treatment. Perit Dial Int neal dialysis: a prospective comparison of two catheters. Perit Dial
2016;36(5):481–508. Int 1994;14(1):70–4.
90. Eklund B, Honkanen E, Kyllonen L, Salmela K, Kala AR. Peritoneal 111. Sodo M, Bracale U, Argentino G, et al. Simultaneous abdominal wall
dialysis access: prospective randomized comparison of single-cuff defect repair and Tenckhoff catheter placement in candidates for
and double-cuff straight Tenckhoff catheters. Nephrol Dial Trans- peritoneal dialysis. J Nephrol 2016;29(5):699–702.
plant 1997;12(12):2664–6. 112. Nelson H, Lindner M, Schuman ES, Gross GF, Hayes JF. Abdominal
91. Nielsen PK, Hemmingsen C, Friis SU, Ladefoged J, Olgaard K. Com- wall hernias as a complication of peritoneal dialysis. Surg Gynecol
parison of straight and curled Tenckhoff peritoneal dialysis catheters Obstet 1983;157(6):541–4.
implanted by percutaneous technique: a prospective randomized 113. Balda S, Power A, Papalois V, Brown E. Impact of hernias on peri-
study. Perit Dial Int 1995;15(1):18–21. toneal dialysis technique survival and residual renal function. Perit
92. Rubin J, Didlake R, Raju S, Hsu H. A prospective randomized evalu- Dial Int 2013;33(6):629–34.
ation of chronic peritoneal catheters. Insertion site and intraperito- 114. Guzman-Valdivia G, Zaga I. Abdominal wall hernia repair in
neal segment. ASAIO Trans 1990;36(3):M497–500. patients with chronic renal failure and a dialysis catheter. Hernia
93. Ejlersen E, Steven K, Lokkegaard H. Paramedian versus midline inci- 2001;5(1):9–11.
sion for the insertion of permanent peritoneal dialysis catheters. A 115. Ye H, Zhou Q, Fan L, et al. The impact of peritoneal dialysis-related
randomized clinical trial. Scand J Urol Nephrol 1990;24(2):151–4. peritonitis on mortality in peritoneal dialysis patients. BMC Nephrol
94. Jwo SC, Chen KS, Lee CC, Chen HY. Prospective randomized study for 2017;18(1). 186-017-0588-4.
comparison of open surgery with laparoscopic-assisted placement of 116. Ghali JR, Bannister KM, Brown FG, et al. Microbiology and outcomes
Tenckhoff peritoneal dialysis catheter—a single center experience of peritonitis in Australian peritoneal dialysis patients. Perit Dial Int
and literature review. J Surg Res 2010;159(1):489–96. 2011;31(6):651–62.
95. Wright MJ, Bel’eed K, Johnson BF, Eadington DW, Sellars L, Farr MJ. 117. Li W, Jin HF, Liu D, et al. Hydrogen sulfide induces apoptosis of pul-
Randomized prospective comparison of laparoscopic and open peri- monary artery smooth muscle cell in rats with pulmonary hyper-
toneal dialysis catheter insertion. Perit Dial Int 1999;19(4):372–5. tension induced by high pulmonary blood flow. Chin Med J (Engl).
96. Tsimoyiannis EC, Siakas P, Glantzounis G, et al. Laparoscopic place- 2009;122(24):3032–8.
ment of the Tenckhoff catheter for peritoneal dialysis. Surg Laparosc 118. von Graevenitz A, Amsterdam D. Microbiological aspects of peritoni-
Endosc Percutan Tech 2000;10(4):218–21. tis associated with continuous ambulatory peritoneal dialysis. Clin
97. Gadallah MF, Pervez A, el-Shahawy MA, et al. Peritoneoscopic ver- Microbiol Rev 1992;5(1):36–48.
sus surgical placement of peritoneal dialysis catheters: a prospective 119. Tzamaloukas AH, Obermiller LE, Gibel LJ, et al. Peritonitis associated
randomized study on outcome. Am J Kidney Dis 1999;33(1):118– with intra-abdominal pathology in continuous ambulatory perito-
22. neal dialysis patients. Perit Dial Int 1993;13(Suppl. 2):S335–7.
98. Qiao Q, Zhou L, Hu K, Xu D, Li L, Lu G. Laparoscopic versus tradi- 120. Barlow AD, Yates PJ, Hosgood SA, Nicholson ML. Case-control com-
tional peritoneal dialysis catheter insertion: a meta analysis. Ren Fail parison of laparoscopic and open washout for peritoneal dialysis-
2016;38(5):838–48. associated peritonitis. Br J Surg 2008;95(11):1416–9.
99. Hagen SM, van Alphen AM, Ijzermans JN, Dor FJ. Laparoscopic ver- 121. Choi P, Nemati E, Banerjee A, Preston E, Levy J, Brown E. Perito-
sus open peritoneal dialysis catheter insertion, the LOCI-trial: a study neal dialysis catheter removal for acute peritonitis: a retrospective
protocol. BMC Surg 2011;11. 35-2482-11-35. analysis of factors associated with catheter removal and prolonged
100. Perakis KE, Stylianou KG, Kyriazis JP, et al. Long-term complication postoperative hospitalization. Am J Kidney Dis 2004;43(1):103–11.
rates and survival of peritoneal dialysis catheters: the role of percu- 122. Burke M, Hawley CM, Badve SV, et al. Relapsing and recurrent peri-
taneous versus surgical placement. Semin Dial 2009;22(5):569–75. toneal dialysis-associated peritonitis: a multicenter registry study.
101. Rosenthal MA, Yang PS, Liu IL, et al. Comparison of outcomes of Am J Kidney Dis 2011;58(3):429–36.
peritoneal dialysis catheters placed by the fluoroscopically guided 123. Troidle L, Gorban-Brennan N, Finkelstein FO. Outcome of patients on
percutaneous method versus directly visualized surgical method. J chronic peritoneal dialysis undergoing peritoneal catheter removal
Vasc Interv Radiol 2008;19(8):1202–7. because of peritonitis. Adv Perit Dial 2005;21:98–101.
102. Gadallah MF, Torres-Rivera C, Ramdeen G, Myrick S, Habashi S, 124. Kawaguchi Y, Kawanishi H, Mujais S, Topley N, Oreopoulos DG.
Andrews G. Relationship between intraperitoneal bleeding, adhe- Encapsulating peritoneal sclerosis: definition, etiology, diagnosis,
sions, and peritoneal dialysis catheter failure: a method of preven- and treatment. International Society for Peritoneal Dialysis Ad Hoc
tion. Adv Perit Dial 2001;17:127–9. Committee on Ultrafiltration Management in Peritoneal Dialysis.
103. Shea M, Hmiel SP, Beck AM. Use of tissue plasminogen activator for Perit Dial Int 2000;20(Suppl. 4):S43–55.
thrombolysis in occluded peritoneal dialysis catheters in children. 125. Alston H, Fan S, Nakayama M. Encapsulating peritoneal sclerosis.
Adv Perit Dial 2001;17:249–52. Semin Nephrol 2017;37(1):93–102.
5 • Access for Renal Replacement Therapy 89

126. Korte MR, Sampimon DE, Betjes MG, Krediet RT. Encapsulating peri- 129. Fieren MW, Betjes MG, Korte MR, Boer WH. Posttransplant encap-
toneal sclerosis: the state of affairs. Nat Rev Nephrol 2011;7(9):528– sulating peritoneal sclerosis: a worrying new trend? Perit Dial Int
38. 2007;27(6):619–24.
127. Upponi S, Butler AJ, Watson CJ, Shaw AS. Encapsulating peritoneal 130. Joseph JT, Jindal RM. Influence of dialysis on post-transplant events.
sclerosis—correlation of radiological findings at CT with underlying Clin Transplant 2002;16(1):18–23.
pathogenesis. Clin Radiol 2014;69(1):103–9. 131. Schwenger V, Dohler B, Morath C, Zeier M, Opelz G. The role of pre-
128. Pampa-Saico S, Caravaca-Fontan F, Burguera-Vion V, et al. Out- transplant dialysis modality on renal allograft outcome. Nephrol
comes of peritoneal dialysis catheter left in place after kidney trans- Dial Transplant 2011;26(11):3761–6.
plantation. Perit Dial Int 2017;37(6):651–4.
6 Brain Death and Cardiac
Death: Donor Criteria and
Care of Deceased Donor
LAURA S. JOHNSON and RAM M. SUBRAMANIAN

CHAPTER OUTLINE Introduction Diagnosis


Death by Neurologic Criteria (Brain Death) Donor Management
Incidence and Causes Cardiac
Physiologic Response Respiratory
Cardiac Renal
Pulmonary Endocrine
Renal Infectious Disease
Hepatic Hematology
Endocrine Other Topics in Donor Management
Inflammatory Ethics
Diagnosis Future Technologies
Death by Cardiopulmonary Criteria Conclusion
(Cardiac Death)

Introduction despite adoption in some states of universal helmet laws.


Although implementation of helmet laws can decrease
As the number of people awaiting organ transplanta- motorcyclist mortality, the lack of a nationwide universal
tion grows yearly, the relative scarcity of available organs helmet law limits the effectiveness of this protective mea-
increasingly requires a standardized, evidence-based sure.1 However, in the overall population of people who
approach to the management of each donor. From the ini- would constitute donor candidates, the death rate has
tial diagnosis of death by neurologic criteria or imminent decreased significantly, attributable primarily to increased
death from cardiorespiratory failure to the optimization of safety measures aimed at motor vehicle drivers.
donor physiology before removal of organs, the intensivist
plays an integral role in this first portion of the transplanta-
PHYSIOLOGIC RESPONSE
tion process.
Brain tissue ischemia, the root source of brain death, trig-
gers a series of well-defined effects as the damage progresses
Death by Neurologic Criteria from the cerebral cortex through the brainstem. These
effects proceed in a consistent sequence. Ischemia of the
(Brain Death) cerebral cortex and upper brainstem (the midbrain) results
in a predominance of parasympathetic activity. Clinically,
INCIDENCE AND CAUSES
this manifests as hypotension and bradycardia. Subse-
In the US just over 53,000 people die of traumatic brain quently, brainstem ischemia at the level of the pons trig-
injuries (TBIs) each year.1 These occur primarily from gers elevation of norepinephrine and epinephrine to well
firearm-related events, motor vehicle-related events, and above normal physiologic levels, while leaving some func-
fall-related events. Firearm-related events remain the most tional parasympathetic nuclei. This results in the “Cush-
prevalent causes of TBI, with 75% of deaths from self- ing reflex,” characterized by significant hypertension due
inflicted injuries and 40% of deaths from assaults coming to an increase in systemic vascular resistance (SVR), and
from brain injury.1 Homicide-related deaths have increased a concomitant bradycardia as the still functional parasym-
over the past decade in people 20 to 24 years of age.1 Motor- pathetic reflex arc attempts to compensate. As the ischemia
cyclists are also at risk for TBI. Deaths from brain injury in progresses through the pontine region to the medulla, the
this population have actually doubled over the past decade parasympathetic nuclei can no longer function, leaving
90
6 • Brain Death and Cardiac Death: Donor Criteria and Care of Deceased Donor 91

unopposed sympathetic input throughout the body. This subsequently leads to additional noncardiogenic pulmo-
is the period referred to as the “autonomic storm.” Finally, nary edema. Lavage samples from donors have demon-
complete brainstem ischemia results in a decrease in sym- strated significant increases in inflammatory markers
pathetic output and complete cardiovascular collapse, relative to nonbrain-dead controls.6 In this setting, addi-
similar to the vasodilatory shock that occurs after a high tional damage to the lung from ventilator-induced injury
spinal cord injury. These changes have very specific effects represents the second insult in a “double-hit” model of pul-
throughout the body, which are best considered by organ monary injury.7
system.
Renal
Cardiac Kidneys also demonstrate decreased survival when
Multiple levels of cardiac dysfunction are seen after brain obtained from brain-dead donors. This has been attributed
death, ranging from histologic changes consistent with to both inflammatory infiltrates in renal grafts and isch-
patchy myocardiocyte ischemia and necrosis, to more emia-reperfusion injury that occurs as the period of auto-
structural changes associated with ventricular dysfunc- nomic storm waxes and wanes.8,9 The kidneys are also
tion. In addition, there are well-characterized electrocar- affected by posterior pituitary failure and the cessation in
diographic changes. Although the mechanisms associated production of arginine vasopressin (AVP). This results in
with these observations are not completely understood, the central diabetes insipidus, a common occurrence in brain-
physiologic effect of the autonomic storm can be tied to a dead patients, occurring in up to 78% of patients.10 Inap-
number of these changes. propriately dilute urine output increases rapidly, leading
Hemodynamic changes follow the level of brainstem to hypovolemia and hypernatremia. The hypovolemia can
injury, with an initial catecholamine surge resulting in worsen the already precarious hemodynamic status of the
significant elevations in SVR.2 Because cardiac output is brain-dead donor, whereas the resulting hypernatremia
influenced inversely by the resistance of the vascular beds it has a significant negative effect on renal and hepatic graft
pumps against, this elevation in SVR results in a decrease in function.
cardiac output. Pressure transmission results in an increase
in left atrial pressure, often above mean pulmonary artery Hepatic
pressure (see Pulmonary section). Subsequent decreases in Although the liver is tolerant of prolonged periods of isch-
sympathetic input lead to a decrease in SVR, often below emia, it nevertheless is affected by both brain death-related
baseline values, a decrease in myocardial contractility, and inflammation and prolonged hypernatremia. The direct
venodilation. Intravascular volume is thus relatively low, effects of inflammation have yet to be elucidated, but biop-
and this decrease in preload with accompanying hypoten- sies after brain death demonstrate increases in inflamma-
sion results in decreased coronary perfusion. Myocardial tory cells, which can potentially increase the risk of primary
contractility is then further affected by ischemia. Despite nonfunction and acute rejection.11,12
these hemodynamic changes, gross echocardiographic Hypernatremia, defined as a plasma sodium level >155
changes vary, with just under 50% of brain-injured mmol/L, has also been associated with poor outcomes after
patients having left ventricular systolic dysfunction, most transplant. Presumably, the hyperosmolar cellular milieu
with evidence of segmental wall motion abnormality.3 The established in hepatocytes while the donor is hypernatremic
electrocardiographic changes progress in a similar fashion results in osmotic injury when the liver is transplanted into
from parasympathetic overload (sinus bradycardia and a nonhypernatremic recipient.13
occasional complete heart block) to sympathetic overload
(sinus tachycardia, progressing to ventricular tachycar- Endocrine
dia). Eventually, there is a return to normal sinus rhythm, Pituitary failure is the primary source of endocrine abnor-
with eventual resolution of the acute ischemic changes that malities associated with brain death. However, not all
initially appear.2 hormones decrease to the same amount. Novitzky et al.
demonstrated in animal studies that, whereas AVP drops
Pulmonary to undetectable levels by 6 hours, and free triiodothy-
The cardiac dysfunction during the autonomic storm ronine (T3) concentrations dropped to 50% of baseline
directly contributes to pulmonary dysfunction in brain- within an hour of injury and were undetectable by 9
dead patients. The intense increase in SVR results in left hours after injury, adrenocorticotropic hormone (ACTH)
atrial pressures often in excess of pulmonary artery pres- and thyroid-stimulating hormone (TSH) were not signifi-
sures. This significantly alters intravascular hydrostatic cantly lower when measured at 16 hours after injury.14
pressure. In addition, increased blood return to the right Although other animal studies have shown less dra-
atrium with systemic shunting increases pulmonary blood matic thyroid hormone responses, they have neverthe-
flow. Both of these changes result in destruction of pulmo- less demonstrated differential responses of other pituitary
nary capillary integrity and cause pulmonary edema and hormones to brain death.10 Human studies suggest that
alveolar and interstitial hemorrhage.4 Also, evidence sug- these differences can be attributed to anatomic differences
gests that the catecholamine storm can directly stimulate between the anterior and posterior pituitary: whereas
pulmonary capillary permeability due to interactions with posterior pituitary hormones (antidiuretic hormone, or
the alpha-adrenoceptor.5 AVP) decrease rapidly after brain death, anterior pitu-
Inflammation-mediated acute lung injury also con- itary hormone (ACTH, TSH) changes are less predictable.
tributes to pulmonary dysfunction after brain injury. The exact mechanisms for this difference are yet to be
Brain death initiates an inflammatory response that fully elucidated.
92 Kidney Transplantation: Principles and Practice

Fig. 6.1 The distribution and pathophysiologic correlation of the rostral–caudal progression of cerebral-spinal ischemia termed coning, which eventu-
ates in herniation and brain death. (Courtesy Kenneth E. Wood, DO.)

Inflammatory
BOX 6.1 Confounding Conditions and
There are two triggers of inflammatory response to brain Exclusions in the Diagnosis of Brain Death
death. The first is direct neural tissue damage, which results
in inflammation of the central nervous system. The second Hypothermia
is in response to ischemia-reperfusion injury that occurs Diagnosis of brain death requires core temperature >32°C
during the period of supranormal SVR in response to pon- Absence of brainstem reflexes when core temperature <28°C
tine ischemia. Release of inflammatory cytokines has been Drug intoxications
demonstrated locally in response to brain injury.15 Both Barbiturates
Tricyclics
cytokine profiles and complement levels have been studied
Alcohol
specifically in organ donation, with effect on delayed graft Narcotics
function16,17; however, future work is necessary to iden- Benzodiazepines
tify ways to blunt these responses and improve donor graft Antipsychotics
function (Fig. 6.1). Antiepileptics
Antihistamines
Acute metabolic endocrine derangements
DIAGNOSIS Electrolyte, acid–base derangements
Death by neurologic criteria, or brain death, is a clinical diag- Uremia
nosis based on the presence or absence of a set of responses Hepatic coma
Hypoglycemia
to neurologic stimuli. In the half century since the Harvard
Hypothyroid
Committee first reported on “irreversible coma” as a new Neurologic diseases
criterion for death, numerous variations on the determina- Persistent vegetative state
tion of brain death have been proposed.18 However, after Locked-in syndrome
the President’s Commission for the Study of Ethical Prob- Akinetic mutism
lems in Medicine equated cardiac death and brain death in
1981,19 the modern criteria for brain death determination
were set. These were then expounded upon by the Ameri-
can Academy of Neurology in 1995,20 and more recently confirmatory for brain death and may be misleading. Com-
reviewed and revalidated by Wijdicks and colleagues in plicating medical conditions that may interfere with clini-
2010.21 Ranging from electroencephalograms to neuro- cal assessment have to be addressed and resolved (Box 6.1).
pathologic examination,22 none of the ancillary tests have Note that severe facial and ocular trauma will interfere with
proven as consistently reliable as a clinical examination many of the brain death tests, making it difficult to form a
done by a physician experienced in performing these tests. definitive clinical diagnosis if they are present. However,
Several prerequisites have to be met before initiation of a in addition to traumatic injuries, severe electrolyte dis-
brain death examination. First, there needs to be evidence turbances (hyper- or hyponatremia, hyper- or hypoglyce-
of a catastrophic brain injury that is compatible with a pos- mia), severe acid–base disturbances (profound acidosis),
sible diagnosis of brain death. This can be established either and endocrine dysfunctions (profound cortisol depletion or
clinically (evidence of gross head trauma) or using basic hypothyroidism) must be identified and corrected. No drug
neuroimaging (computed tomography [CT]). However, it is intoxication or evidence of poisoning can be present. This
important to remember that CT findings are not themselves requires performing a drug screen and waiting for clearance
6 • Brain Death and Cardiac Death: Donor Criteria and Care of Deceased Donor 93

of any alcohol to below the legal limit for driving (0.08%). most difficult part of the examination for practitioners,
In addition, it is necessary to identify any medications given as a wide range of spontaneous or reflex movements
in the hospital that could result in central nervous system have been described in the literature. These include
depression; these need to have cleared the patient’s system everything from isolated jerks of the upper extremi-
before proceeding with testing. An appropriate time has been ties to cremasteric and abdominal muscle reflexes to
suggested at five times a drug’s half-life, assuming normal respiratory-like movements.23,24 More extreme reflexes
hepatic and renal function. Reversal agents are appropriate have elicited more fanciful descriptions, including the
where available (opiates and benzodiazepines). Finally, the “Lazarus sign,” where a combination of shoulder, neck,
patient must at least have a core body temperature >32°C and extremity movements makes patients appear to
before starting the brain death examination. Severe hypo- rise from the bed.24 Clinical expertise is necessary to
thermia, defined as core body temperature <32°C, affects differentiate between these movements, and central
papillary light response, with complete loss of brainstem and cerebral motor responses to pain. Ultimately, these
reflexes at core temperatures <28°C. Although rewarming movements do not invalidate the diagnosis of brain
techniques are beyond the scope of this chapter, for most death, but it is important for the clinician to be aware of
patients a warming blanket should be sufficient to obtain these responses so as to counsel family members appro-
appropriate temperatures before starting the examination. priately.
Once these prerequisites have been met, the brain death 2. Absence of brainstem reflexes20,21
examination can proceed (Fig. 6.2). Three major findings a. Pupillary reflex (cranial nerve [CN] II and III)
need to be identified and documented to confirm brain i. Pupils are round or oval, typically of midrange
death. These are: (1) coma or unresponsiveness, (2) absence size (4 mm), though some can be as dilated as
of brainstem reflexes, and (3) apnea. 9 mm.
   ii. Pupils show no response to light.
1. Coma or unresponsiveness. This component requires b. Ocular movement (CN III, VI, and VIII)
that there is no motor response or eye movement to i. Oculocephalic reflex. Otherwise known as the
noxious stimuli, typically described as nail bed pres- “doll’s eye” sign, in brain death the pupils will not
sure or supraorbital pressure. Often, this can be the show any movement as the head is turned rapidly

COMA

Prerequisites Exclusions/Confounding Conditions


• Cause of coma established AND • Hypothermia
• Supportive neuroimaging • Drug intoxications
• Endocrine crisis
• Severe electrolyte/acid–base
disturbances

Above defined/excluded
YES

Clinical Examination
• Absent motor response
• Absent brainstem reflexes
• Apnea with documented PCO2 ≥60 mmHg

Ability to perform all elements of clinical exam

YES NO

Diagnosis of Brain Death Confirmatory Studies

YES

Absence of cerebral blood flow

NO

Brain death diagnosis not supported


Fig. 6.2 General approach to the diagnosis of brain death.
94 Kidney Transplantation: Principles and Practice

to one side or the other. Caution: This test is not vi.  No prior history of CO2 retention (no history
to be utilized in patients who have a suspicion of of chronic obstructive pulmonary disease or
spine instability or fracture. obstructive sleep apnea)
ii. Vestibulo-ocular reflex. Also known as the b. Preparation
“caloric reflex test,” this test requires confir- i. Preoxygenate the patient with 100% O2 before
mation of a clear external auditory canal and the test; target is a Pao2 >200 mmHg.
elevation of the head to 30 degrees before ii. Reduce the ventilation frequency to 10 to 12
starting. Each external auditory canal is irri- breaths/min to achieve eucapnea.
gated (separately, with an interval of at least iii. Measure arterial Po2, Pco2, and pH after these
5 minutes) with approximately 50 mL of ice preparatory steps before starting the test.
water. In the presence of brain death, no eye c. Testing
movement will be seen during the 1-min- i. Disconnect the patient from the ventilator.
ute observation period, regardless of the ear ii. Continue to deliver 100% Fio2 at the level of the
irrigated. carina through a suction catheter or straight nasal
c. Facial sensation and facial motor response (CN V cannula placed through the endotracheal tube.
and VII) iii. Observe closely for respiratory movements
i. Absence of a corneal reflex (eyelid movement/“blink (abdominal, chest, neck) that could produce ade-
reflex”—CN V1 and VII). Although many texts quate tidal volumes.
describe performing this test with a cotton swab, iv. Continue the test as long as the patient remains
some centers have moved toward stimulation with stable. If, at the completion of 8 to 10 minutes, the
a puff of air from an empty 10-cc syringe. This patient remains stable, another 1 to 2 minutes
decreases the risk of corneal damage from direct can be taken before drawing an arterial blood gas.
contact, while still providing a sufficient stimulus If the patient becomes hypotensive (systolic blood
to potentially evoke a response. pressure <90 mmHg), hypoxic (O2 sat <85%), or
ii. Absence of a jaw reflex (masseter reflex—CN V3). develops cardiac arrhythmias, at any time before
For this test, the mandible is tapped at a down- a full 8 to 10 minutes, an arterial blood gas needs
ward angle just below the lips. A positive test to be drawn immediately and the ventilator needs
would result in the upward movement of the to be reconnected.
mandible in response. Usually, this reflex is very d. Result interpretation
slight. i. If respiratory movements are observed, the test is
iii. Absence of facial movement to noxious stimuli negative and the patient is not brain-dead.
(CN V3 and VII). Deep pressure on the supraorbital ii. If respiratory movements are not observed, and
ridge or mandibular condyles at the temporoman- the Paco2 is >60 mmHg or >20 mmHg above
dibular joint should not result in any facial muscle baseline normal Paco2, the test is positive and the
movement (grimacing). patient is clinically brain-dead.
d. Pharyngeal and tracheal reflexes (CN IX and X) iii. If respiratory movements are not observed, but
i. Pharyngeal reflex (“gag reflex”—CN IX and X). the test was halted early for hemodynamic insta-
Posterior pharyngeal stimulation with a tongue bility and the Paco2 parameters were not met, the
blade or hard suction catheter should not elicit a test is indeterminate and additional testing should
response. Note that this is separate from the tra- be considered.
cheal reflex. This test cannot be performed on every patient; approxi-
ii. Tracheal reflex (“cough reflex”—CN X). Tracheal mately 10% of patients will be hemodynamically unstable
stimulation, usually achieved by suctioning the at the time testing could occur, or before the conclusion
endotracheal tube, should not elicit a response of testing, requiring a premature stop.25 In these circum-
over multiple passes. stances, other tests may be utilized.  
3. Apnea. The absence of a drive to breathe is the final test Additional testing is not required by the American
in the clinical evaluation of brain death. Normally, an Association of Neurology guidelines, as brain death is a
elevation in CO2 above a critical level (in the US defined clinical examination. However, in patients for whom a
as >60 mmHg)19 will stimulate the respiratory center in complete examination cannot be performed, ancillary
the brainstem (medulla), which then signals the respi- testing can be useful.19,21 Certain hospital or state guide-
ratory muscles to breathe. The apnea test is designed to lines on the declaration of brain death may also require an
provoke this response in an effort to establish whether additional test, and therefore the practitioner is encour-
the medulla (the lowest anatomic segment of the brain- aged to review hospital and state-specific requirements
stem) is alive. (Box 6.2).
a. Prerequisites
i. Normotension (systolic blood pressure ≥90
mmHg)
Death by Cardiopulmonary
ii. Normothermia (core temperature >36°C) Criteria (Cardiac Death)
iii. Euvolemia
iv. Eucapnea (Paco2 35–45 mmHg) With less than 1% of deaths in the US occurring from
v. Absence of hypoxia brain death, it has become a priority in the transplantation
6 • Brain Death and Cardiac Death: Donor Criteria and Care of Deceased Donor 95

BOX 6.2 Confirmatory Studies BOX 6.3 Prediction of Death Within 60


Minutes of Withdrawal of Life-Sustaining
Cerebral angiography
Contrast agent injected under high pressure into anterior and
Treatment
posterior circulations UNOS Characteristics For Death within 60 minutes
Absence of cerebral filling at carotid and vertebral entrance into
skull Apnea
Potential for contrast-induced nephrotoxicity Respiratory rate <8 or >30 breaths/min
Rarely performed Dopamine ≥15 μg/kg/min
Cerebral scintigraphy (technetium 99mTc-HMPAO) Left or right ventricular assist device
Can be performed at bedside in brief time Venoarterial or venovenous extracorporeal membrane
Good correlation with conventional angiography Oxygenation
Isotope angiography Positive end-expiratory pressure ≥10 and Sao2 ≥92%
Albumin labeled with technetium 99m Fio2 ≥0.5 and Sao2 ≤92%
Can be performed at bedside Norepinephrine or phenylephrine ≥0.2 μg/kg/min
Delayed filling of sagittal and transverse sinuses Pacemaker unassisted heart rate <30
Posterior cerebral circulation not visualized Intraarterial Balloon Pump set at 1:1 or dobutamine or dopamine
Transcranial Doppler ultrasound ≥10 μg/kg/min and Cardiac Index ≤2.2 L/min/m2
Middle cerebral artery through temporal bone above zygomat- IABP 1:1 and CI ≤1.5 L/min/m2
ic arch and vertebral or basilar arteries through suboccipital
transcranial windows bilaterally
Lack of transcranial Doppler signals should not be interpreted
Controlled DCD takes place when a planned withdrawal
as confirmatory because 10% of patients may not have
temporal windows
of cardiorespiratory support has been determined to be the
May not be diagnostic with intratentorial lesions best course for a particular patient. Naturally, this neces-
Electroencephalogram sitates ongoing discussions with the family about the
No electrical activity for 30 minutes patient’s wishes and plans of care. After families express the
Complex technical requirements desire to proceed with withdrawal, a representative from
the organ procurement organization can present the option
of DCD. Determining who will be a good candidate, or in
other words, which patients will die within an acceptable
community to expand available sources for organs to trans- warm ischemia time, is difficult (Box 6.3). Various studies
plant. One way this has been done is to reevaluate donor have demonstrated that increased ventilatory and circula-
criteria and designate “expanded criteria” donors based on tory support correlate with death in under 60 minutes.32–34
age and comorbidities. Another way this has been achieved Decision support tools have been suggested to help with this
has been to redefine “standard” donor criteria based on sci- process, although the opinion of an intensivist has proven
entific evidence; this has been most successful in the arena similarly effective in the most recent large trial.35 With 20%
of lung transplantation.26 to 25% of patients not correctly identified, it is important to
Finally, the most recent method has been to revisit counsel families who request withdrawal of care that this
donation after circulatory death (DCD). Historically, the is an option, while at the same time preparing for ongoing
first transplants were done using organs from asystolic patient care through the dying process if they do not meet
donors, but with professional acceptance of brain death criteria.
following the 1968 Ad Hoc Committee of Harvard Medi- Once DCD has been authorized, every attempt is made
cal School review of the issue,18 and evidence of improved to keep the patient medically stable until treatment is
outcomes from donors whose hearts continue to beat, withdrawn. A short period of time is necessary to allow for
DCD faded into obscurity. However, with new evidence organ allocation, but this process should not be extended
that organs can tolerate short periods of warm ischemia over several days out of respect for the patient and family.
with successful outcomes, DCD is being revived by the Once arrangements have been made, the patient is trans-
transplant community.27–30 Often this option is possible ported to the operating room, at which point the family is
for patients who have suffered a significant head injury allowed to pay their respects. The medical team respon-
requiring full cardiopulmonary support, but who are not sible for the patient in the intensive care setting then
able to undergo brain death testing.31 Less frequently, proceeds with withdrawal of care, and after a period of 5
patients who have had cardiac arrests or suffer from ter- minutes of continuous asystole (monitoring with an elec-
minal respiratory diseases may be good candidates for trocardiograph and arterial line), the patient is declared
DCD.31 dead.30,36 Stiegler and colleagues have shown recently
in an animal model that there is no return of brainstem
function after 5 minutes of asystole, even if cardiopulmo-
DIAGNOSIS
nary resuscitation is started at 5 minutes.37 At this point,
Deceased circulatory death is categorized using the modi- the transplant team can proceed with organ recovery
fied Maastricht classification. Although the possibility for (Fig. 6.3).
acute retrieval from an uncontrolled DCD does exist, for Clearly, this process has significant ethical implica-
the purposes of this chapter only controlled DCD will be tions and has raised questions around the world regard-
discussed. ing the nature of death and organ donation. Until the
96 Kidney Transplantation: Principles and Practice

Patient with a devastating illness

Family expresses wish


to withdraw care

OPO notified & presents


option for DCD Donation

Family does not agree to DCD Family agrees to DCD

Proceed with withdrawal Maintenance of hemodynamic


stability while organ location performed

THEN

Procedure scheduled for


the OR

Primary team withdraws care


in the OR & monitors patient

Patient is asystolic for 5 minutes Patient is not asystolic for >60–90 minutes

Procede with procurement DCD NO LONGER POSSIBLE

Move patient to quiet room for remainder


of withdrawal period

Fig. 6.3 General approach to the diagnosis of brain death. OPO, organ procurement organization; DCD, donation after circulatory death; OR, operating
room.

ethicists decide otherwise, the most important role the physicians should be familiar with their hospital’s policy
medical team can play in a DCD is to enforce the “dead on DCD, as they are often the individuals declaring death
donor rule,” wherein patients can only become donors in these situations.
after they are dead, and recovery of organs cannot cause
a donor’s death. There is some thought that premortem
interventions, such as obtaining blood samples and main- Donor Management
taining life-sustaining therapy while organ allocation
processes take place, are acceptable, as the overall goal is The management of organ donor candidates falls under
to respect the patient’s final wish for organ donation.30,36 the purview of the critical care physician. As such, the
However, procedures that can cause serious harm, such same systematic approach employed when managing
as systemic heparinization, or cause pain, such as femo- other patients in the intensive care unit should be used in
ral cannulation, are not permitted until the patient has the management of potential organ donors. Approaches
been declared clinically dead.36 It is also important to note to organ donor management following a protocol con-
that the transplantation team can have absolutely no tinue to show improvement in the overall number of
involvement in patient management until after death has organs transplanted, with beneficial effects on graft func-
been declared, to avoid a conflict of interest. Critical care tion.38–40 As a result, various international groups have
6 • Brain Death and Cardiac Death: Donor Criteria and Care of Deceased Donor 97

endorsed standardized pathways for the management of


CARDIAC
potential organ donors.41 Donor management guidelines
are constantly undergoing reevaluation and updating as As described previously, the cardiovascular system is subject
the science of critical care advances; the guidelines laid to rapid changes over the course of brain death. A proactive
out in Box 6.4 represent the current recommendations in approach to management of these patients can decrease
the management of organ donation. For the context of this the percentage of donors who suffer a cardiac arrest in the
section, it is important to remember that the smooth inte- predonation period, typically reported as 5% to 10% of all
gration of organ donor management techniques within potential donors. Overall, the hemodynamic goals for a
the context of a preexisting systematic approach to criti- potential organ donor are the same as for any other patient
cally ill patients will result in the best outcomes for these in the intensive care unit: to optimize cardiac output so as to
patients, regardless of whether they proceed to organ achieve organ perfusion with minimal vasoactive support.
donation. In addition, strict attention to donor manage- Linking the physiology of brain death to effects on cardiac
ment guidelines will result in the highest-quality organs status can better direct therapy to this end.
for procurement, and ensure the best outcomes for organ The earliest effect of brain death on cardiac func-
donation recipients (see Box 6.4).42 tion is during the autonomic storm, during which time
catecholamine concentrations reach supraphysiologic
levels. This can result in one or several of the following:
increased heart rate, increased blood pressure, increased
BOX 6.4 Donor Management Guidelines cardiac output, and increased SVR. All of these result in a
deficit in myocardial oxygen availability in the setting of
Cardiac increased demand, and can result in anatomic heart dam-
□ Mean arterial pressure 70–100 mmHg age. Although this period is self-limited, and can be quite
□ Treat hypertension with short-acting beta-blocker short, the question of whether it should be treated has
□ Treat hypotension with volume, followed by vasoactive been raised. Audibert and colleagues demonstrated that
agents management of these initial cardiac responses to auto-
Limit vasoactive agents where possible to ≤1 nomic storm with short-acting beta-blockade can improve
□ Heart rate 60–120 beats/min cardiac function in hearts after transplantation.43 Given
□ Urine output 0.5–3 mL/kg/h
□ Hemoglobin 8 g/dL
these initial data, judicious utilization of a short-acting
□ Scvo2 >70% beta-blocker (esmolol 100–500 μg/kg bolus followed by
□ EF ≥50% 100–300 μg/kg/min, or nitroprusside 0.5–5.0 μg/kg/
min) should be considered in potential cardiac donors
Respiratory when systolic blood pressures rise above 160 mmHg or
□ Mechanical ventilation goals mean arterial pressures rise above 90 mmHg, keeping in
□ Fraction of inspired oxygen 0.40 mind that these medications should be stopped quickly
□ Normal arterial pH
once the period of autonomic storm is completed.
□ Tidal volumes 8–10 mL/kg
The first 48 hours of progressive brain ischemia and death
□ Plateau pressure <35 cm H O
2 are characterized by an increased incidence of arrhythmias.
□ Fluid management
□ Judicious fluid administration to avoid pulmonary edema,
Early arrhythmias range from tachyarrhythmias during
with diuresis as necessary after the early resuscitative phase the initial catecholamine surge to bradyarrhythmias as the
of care heart tries to accommodate the early period of severe hyper-
tension; later bradyarrhythmias will occur due to vagus
Renal nerve disruption and parasympathetic overstimulation of
□ E uvolemia with appropriate end-organ perfusion and oxygen the sinoatrial node or atrial ventricular node.13,44 As many
delivery (UOP 0.5–3 mL/kg/h) as one in five patients will manifest an arrhythmia.45
□ Early recognition and correction of DI (UOP >4 mL/kg/h,
Strict control of electrolyte imbalances, body tempera-
increasingly serum osm, inappropriately dilute urine, or ture, and acidosis can help mitigate these issues, which
hypernatremia Na >145) should otherwise be treated as per the Advanced Cardiopul-
□ DDAVP 8 ng/kg loading dose followed by 4 ng/kg/h titrated

to urine output <3 mL/kg/h


monary Life Support guidelines published by the American
□ Plasma sodium concentration 140–155 mmol/L College of Cardiology and the American Heart Association.
It is worth noting that the need for cardiopulmonary resus-
Endocrine citation (CPR) alone does not negatively affect graft func-
□ Thyroid hormone replacement tion if it is implemented in a timely, controlled fashion for a
□ Triiodothyronine 4 μg bolus followed by 3 μg/h infusion × 10 quickly correctable problem.46
hours After the initial catecholamine surge, loss of vasomo-
□ If only thyroxine is available, start 20 μg bolus followed by 10
tor centers with brainstem infarction results in reduction
μg/h infusion × 10 hours of SVR and profound vasodilatation. Often, this is a time
□ Euglycemia (glucose ≥180) when patients can become hypotensive due to the physiol-
Miscellaneous ogy of distributive shock. This can also further destabilize
□ Normothermia the myocardium, previously injured during the period of
autonomic storm. Standard physiologic parameters asso-
DI, diabetes insipidus; DDAVP, desmopressin; EF, ejection fraction; ciated with end organ perfusion should be targeted, and
Scvo2, central venous oxygen saturation; UOP, urine output. include a mean arterial pressure >70 mmHg, urine output
98 Kidney Transplantation: Principles and Practice

0.5 to 3 mL/kg/h, heart rate of 60 to 120 beats/min, and although this has yet to be borne out in the organ donor
a hemoglobin >8 g/dL. Acidosis needs to be corrected, management literature.49,50 Regardless of the modality
targeting a pH 7.40 to 7.45. Volume expansion is the used to evaluate cardiac function, the recommended tar-
initial therapy of choice, using isotonic fluids or colloids gets are a cardiac index >2.4 L/min/m2 and a mean arterial
(blood or albumin), with a target of euvolemia at the end pressure of >70 mmHg.
of resuscitation. Fluid choice should be tailored depending It is important to remember that donor management
on potential organs to be harvested, as crystalloid admin- techniques also increase the potential for the heart trans-
istration in excess can decrease the chances of successful plantation as an end in itself. After stabilizing the donor
lung harvest. and reaching standard physiologic norms, initial evalu-
If volume alone is insufficient to attain these goals ation of the heart should be performed using echocar-
quickly, as may be the case in up to 80% of donors, vasoac- diography, evaluating for structural heart disease, left
tive agent support is required. The use of vasoactive agents ventricular ejection fraction, and gross wall motion abnor-
has been shown to improve organ viability by stabilizing malities. However, it should be noted that although single-
the donor, though there is still debate about the first-line use echocardiography is helpful in screening for anatomic
agent of choice.41 In deciding between dopamine and nor- abnormalities, it is not sufficient for the evaluation of physi-
epinephrine, it is important to consider the physiologic ologic suitability. Given evidence that young hearts with
problem being treated. In the setting of low SVR alone, left ventricular dysfunction can recover from the initial
norepinephrine may offer a more targeted treatment insult of autonomic storm, serial evaluation of cardiac func-
option, whereas if there is cardiac dysfunction separately tion after optimizing volume status and repleting hormone
or in addition, dopamine will provide single-agent treat- deficiencies (discussed later) provides the best data for deter-
ment for both. AVP (0.04 U/min) may also be an appro- mining suitability for transplant. Cardiac catheterization is
priate choice in the early periods of vasodilatory shock.47 reserved for the assessment of potential donor hearts from
Vasopressin has been shown to improve outcomes after patients 45 years of age or older, or at the particular request
kidney and lung transplants,47 a result that is ascribed to of the transplant center.
a decrease in microvascular thrombotic events after “pre-
treating” the endothelium. Coupled with effectiveness in RESPIRATORY
treating diabetes insipidus, it is a useful vasoactive agent
in the treatment of vasodilatory shock after brain death. Perhaps nowhere has the adoption of a standardized
If an inotrope is required, it is important to realize that the protocol made as much impact for organ recovery as in
vasodilatory effects of dobutamine, especially in the setting the management of potential lung donors. Historically,
of hypovolemia, may provoke additional hypotension and lung procurement has been as low as 7% from all avail-
tachycardia. However, at low doses (5 μg/kg/min), dobu- able organ donors, and hovers optimally around 15% to
tamine has been shown to have some protective effects in 25%,41 with multiple factors contributing to this rate.25
animal models of renal transplant.48 If the donor is tachy- These include acute lung injury after brain death, atel-
cardic at baseline, milrinone is a better first-line inotropic ectasis, aspiration, and pneumonia. Although previous
agent. work demonstrated that a standardized protocol could
Aside from standard physiologic monitoring, invasive improve recovery, implementation of the San Antonio
markers of volume status can be very useful, especially lung transplant donor management protocol, developed
when donors may have confounding factors affecting by Luis Angel and colleagues, has demonstrably doubled
their urine output (mannitol, diabetes insipidus). Central their ability to identify and transplant lungs from donors
venous pressure (CVP) has been the traditional parameter at their institution.51 Following in the footsteps of previous
of choice, with a target range of 4 to 12 mmHg. However, in work that suggested marginal lung donor candidates could
the setting of mechanical ventilation, this measure has sig- actually produce adequate lungs for transplant recipients,
nificant limitations due to alterations in intrathoracic pres- Angel was able to show not only an increase in the num-
sure. CVP monitoring does allow measurement of central ber of “poor” donors optimized to ideal or extended criteria
venous oxygen saturation (Scvo2), a marker of organ perfu- donors with implementation of their protocol, but also that
sion and delivery. Regardless of whether this parameter is no significant 30-day or 1-year mortality differences were
checked intermittently or continuously, the target range is observed between groups.51
still a Scvo2 > 70%. Despite an absence of data on “normal” Lung-protective ventilation, the strategy to prevent ven-
ranges for Scvo2 after brain death, this value still represents tilator-associated lung trauma from a combination of over-
a useful target for end organ oxygen delivery and consump- stretch of lung parenchyma and oxygen toxicity (as defined
tion. If there is a concern for myocardial depression, either as a delivered Fio2 greater than 0.60 for greater than 48
preexisting in the donor or as a result of brain death itself, hours), has been demonstrated to increase the available
additional monitoring can be used. Although pulmonary pool of potential lungs for transplantation.52 Arguably, the
artery catheters have been used to optimize cardiac output study that demonstrated that effect also left patients on con-
in the past, echocardiography is increasingly prevalent in tinuous positive pressure support during their apnea trials,
the intensive care unit (ICU), providing equivalent informa- so it is less clear how much the continuity of a closed circuit
tion. In addition, the use of dynamic indices of cardiac func- affected the ability to keep lungs recruited before transplan-
tion, such as pulse pressure variation and stroke volume tation. Regardless, baseline targets of oxygenation and ven-
variation, has been promoted in the intensive care litera- tilation (Po2 >80 mmHg and Pco2 30–35 mmHg) can be
ture as a more accurate measure of preload than static indi- met using this technique and should be even if patients are
ces such as CVP or pulmonary capillary wedge pressure, not candidates for lung donation, to maintain appropriate
6 • Brain Death and Cardiac Death: Donor Criteria and Care of Deceased Donor 99

oxygen delivery to the other organs. Lung recruitment If urine output targets exceed 4 mL/kg/h, diabetes insipi-
strategies need to be balanced with appropriate peak and dus should be suspected. The hypothalamic–posterior
plateau airway pressures to avoid negative effects on patient pituitary neuronal connections become hypoxic with the
hemodynamics. progression of brain death, and production of AVP ceases.
Strict monitoring of volume status is the next important As a result, despite relative hypotension in the setting of
component of lung management in the donor population. loss of sympathetic tone, the kidneys continue to secrete
Cardiac instability during the autonomic storm causes large volumes of dilute urine until the patient becomes pro-
significant shifts in blood pressure, and, without strict foundly hypovolemic and oliguric. This is also associated
parameters, can result in significant volume overload. With with hypernatremia, which is known to be detrimental
direct catecholamine effect on alveolar permeability, fluid for potential liver donors. Treatment is aimed at restoring
can redistribute into the pulmonary interstitium. In addi- intravascular volume, at the same time supplementing
tion, an increase in left atrial pressure due to increased SVR the loss of AVP. Supplementation can be done either with
can result in functional heart failure and cardiogenic pul- desmopressin (DDAVP 8 ng/kg loading dose followed by 4
monary edema. Several studies have demonstrated that ng/kg/h titrated to urine output), or with continuous infu-
pulmonary edema decreases the potential for lung trans- sion of AVP (1 U bolus followed by 0.01–0.04 U/min).56
plantation.53,54 Judicious fluid administration and adminis- Desmopressin is a more selective vasopressin receptor ago-
tering diuretics as tolerated are parts of the overall strategy nist, targeting V2 receptors, and therefore has significant
for decreasing pulmonary edema and optimizing oxygen- antidiuretic properties without the concomitant vasopres-
ation before donation, without significantly affecting renal sor activity of AVP.44,56 It can therefore be used without
graft function.55 increasing organ vasoconstriction and causing posttrans-
Standard ICU management for the prevention of venti- plant impairment. However, if the patient requires vaso-
lator-associated pneumonia and aspiration pneumonitis pressor support in addition to volume repletion, AVP is a
should be continued. This includes elevating the head of better choice.
the bed at least 30 degrees at all times, administration of Hypernatremia, a side effect of diabetes insipidus, has
deep vein thrombosis prophylaxis (mechanical and chemi- been associated with increased rates of liver graft loss, and
cal methods), and administration of a proton pump inhibi- possibly renal graft function.57,58,59,60 Serum sodium levels
tor for the prevention of gastritis. If, after lung recruitment should be kept between 140 and 155 mmol/L, in an effort
measures, infiltrates or contusions persist on chest x-ray, to decrease osmotic gradients between donor liver and
bronchoscopy with bilateral lavage can be performed to recipient vasculature upon reanastomosis.61 In addition, a
evaluate those areas. balanced nutritional approach using dextrose-based solu-
The most important concept in the management of tions is helpful for maintaining hepatic glycogen stores;
donors for lung transplantation is to remember that every however, this cannot be done at the expense of glycemic
donor should be considered a potential lung donor, because control.61
standardized attention to a few basic management prin-
ciples can result in optimization of initially poor-quality ENDOCRINE
organs.
Hormonal resuscitation is a concept in organ donor man-
agement aimed at addressing the three endocrine deficien-
RENAL
cies of brain death that can affect outcomes.
Most considerations relative to donor renal function have The first of these is thyroid hormone (thyroxine [T4]
been mentioned already in the section on cardiovascular and T3) depletion. The exact mechanism of thyroid hor-
management. Euvolemia with appropriate end-organ per- mone function is as yet not fully elucidated, but several
fusion and oxygen delivery is the goal of intensive donor studies have implicated low levels of thyroid hormone
management. It is important to mention here that, in the as a contributory factor to the hemodynamic instabil-
process of managing the brain injury before brain death, the ity after brain death. Novitzky and colleagues were able
donor is likely to have been exposed to a number of agents to demonstrate in a series of animal and human studies
with diuretic properties (mannitol, hypertonic saline). that the administration of T3 improved cardiac function,
These agents can complicate both the hemodynamic stabil- decreased hemodynamic instability, and transitioned
ity of an already hypovolemic patient, and affect the diagno- brain-dead donors from anaerobic metabolism to aero-
sis of renal-specific processes associated with brain injury. bic metabolism.14,62 Additional work by these and other
It is important for these agents to be carefully documented authors has confirmed these findings while using T4 and
and considered when evaluating urine output. suggested that the use of thyroid hormone replacement
The target for urine output, as previously mentioned, can increase the overall number of organs harvested
is the same as for any other patient in the ICU—approxi- from a single donor and decrease the incidence of delayed
mately 0.5 to 1 mL/kg/h. However, up to 2.5 to 3 mL/kg/h graft function.63–65 Rosendale et al. looked at recipient
can be considered normal in donors as they equilibrate from outcomes and were able to demonstrate that, for car-
fluid repletion during periods of hemodynamic instability. It diac recipients, 1-month survival was improved, with a
is important to avoid an excessively positive fluid balance— 50% decrease in early graft dysfunction, although this
there is a significant body of literature in critical care sug- improvement occurred in conjunction with the use of
gesting that this can lead to end-organ dysfunction, and in steroids.66 Although clinical research has suggested that
fact, fluid-restrictive strategies have demonstrated improve- oral repletion of T4 produces comparable bioavailability
ment in the numbers of lungs transplanted.55 at 91% to 93% of the available IV T4 dose in circulation,
100 Kidney Transplantation: Principles and Practice

overall, current recommendations call for thyroid hor-


mone supplementation, preferably with T3 due to its more BOX 6.5 Infection Diagnoses That Exclude
rapid onset and decreased susceptibility to influence by Transplantation
exogenous factors.67 T3 can be dosed at 4 μg bolus fol- □  ctive fungal, parasitic, or viral meningitis or encephalitis
A
lowed by 3 μg/h infusion for 10 hours.56 If T3 is unavail- □ Bacterial meningitis—conditional—okay if treated for at least
able, T4 can be used at 20 μg bolus followed by a 10 μg/h 48 hours
infusion for 10 hours.45
Vasopressin, as mentioned earlier, is a pharmacologic Bacterial
agent in both brain death-related peripheral vasodilata- □ Tuberculosis
tion and diabetes insipidus. Again, treatment is aimed at □ Gangrenous bowel or perforated bowel or intraabdominal
restoring intravascular volume, while at the same time sepsis
supplementing the loss of AVP. Corticosteroid administra- Viral
tion is the third component of hormone resuscitation, and □  epatitis B surface antigen positivity
H
is based on two possible mechanisms of function. The first □ Rabies
is the attenuation of the inflammatory process associated □ Retroviral infections, including HTLV-I/II
with brain death. Two studies looking at posttransplant □ Active herpes simplex, EBV, varicella, or CMV viremia, or pneu-
liver rejection markers, inflammatory markers, and post- monia
transplant cardiac function demonstrated improvement □ West Nile virus
in both parameters when steroid therapy was given to the □ Hep B—HBV core antibody okay
donor.66,68 In addition, lung procurement was demon- □ Hep C—Hep C positive donor to Hep C positive recipients
strated to be directly tied to the administration of steroids, okay
which was related to increased oxygenation.69 However, □ HIV—HIV positive donor to HIV positive recipients okay
□ HIV—HIV seronegative in high risk behaviors conditionally okay
more recent work has suggested that, although suppression
with adequate consent
of the inflammatory state does occur, the incidence of rejec-
tion in liver and kidney recipients was not improved with Fungal
steroid pretreatment.70 The second physiologic derange- □  ctive infection with Cryptococcus, Aspergillus, Histoplasma, Coc-
A
ment for which steroid administration has been suggested cidioides
to help is for adrenal insufficiency. During the CORTICOME □ Active candidemia or invasive yeast
trial from France, which randomized steroid supplementa-
Parasites
tion as part of brain death management, testing revealed
78% incidence of adrenal insufficiency in the population. □  ctive infection with Trypanosoma cruzi (Chagas), Leishmania,
A
However, significant decreases in vasopressor utilization Strongyloides, or Plasmodium sp. (malaria)
after steroid administration was not related to preexist- Prion
ing adrenal insufficiency, making it less likely this is the □ Creutzfeldt–Jakob disease
mechanism by which corticosteroid use improved donor
physiology.71 Current guidelines still endorse the use of
CMV, cytomegalovirus; EBV, Epstein–Barr virus; HIV, human immuno-
methylprednisolone (15 mg/kg bolus) as part of hormone
deficiency virus; HTLV, human T-lymphotropic virus.
resuscitation.70
Finally, although not part of hormone resuscitation, gly-
cemic control is an important ongoing component of inten- not contraindications to transplantation.75 Those that are
sive care management of the donor. Between stress-related serious are listed in Box 6.5.76 It is worth noting that sev-
insulin resistance after brain injury and the replacement of eral diseases that used to be absolute contraindications are
free water deficits during diabetes insipidus with free water now conditionally so; most notably hepatitis B and C, and
solutions that contain dextrose, donors can become quite human immunodeficiency virus (HIV), but also bacterial
hyperglycemic.42 Evidence suggests that renal function meningitis.41,77–79 Recipients of organs from these donors
over the course of donor management worsens significantly will need posttransplant monitoring, and in some situations
in the setting of persistent hyperglycemia and significant additional therapy, but donation from individuals with
glycemic shifts72; this could reflect both osmotic diuresis these diseases is no longer impossible. It is important to pur-
affecting donor resuscitation and tissue damage due to pro- sue accurate testing of these diseases in the donors; nucleic-
tein glycosylation. Management of hyperglycemia (glucose acid amplification testing (NAT) is now the recommended
>180 mg/dL) improves the number of organs transplanted method of testing where appropriate. Effective treatment of
per donor and results in higher rates of long term renal graft preexisting donor infections can result in successful trans-
survival.73 In addition, pancreas function in the recipient plantation, as can transplantation of a donor organ exposed
is affected by donor glucose levels, with normoglycemia to a chronic viral infection to a recipient with the same
improving outcomes.74 An insulin infusion therapy can be exposure history.80 It is important to recognize infections
titrated for this effect. in donors and treat them quickly and efficiently to keep the
donation process moving forward. Treatment starts with
good prevention—standard ICU policies for the prevention
INFECTIOUS DISEASE
of hospital-acquired infections (pneumonias, central line-
Although donor to recipient infection transmission is well associated bloodstream infections, and urinary tract infec-
documented and can result in serious, and potentially tions) should be maintained even after the focus of care has
fatal, outcomes, most donor infections independently are transitioned.
6 • Brain Death and Cardiac Death: Donor Criteria and Care of Deceased Donor 101

A high index of suspicion for infection is the next step. hypercoagulable states early in their management.83 Stan-
There are many reasons why a donor will not mount a dard ICU protocol for the prevention of deep vein thrombo-
standard response to infection: loss of thermoregulation sis should be continued.
preventing fevers, preexisting leukocytosis secondary to
the inciting injury, massive blood transfusions, or use of
steroids resulting in immunosuppression. Therefore it Other Topics in Donor
is important to pay attention to other clinical markers,
including the skin examination, sputum characteristics,
Management
chest x-ray findings, and urinalysis results. Choice and
ETHICS
interpretation of cultures, as well as guidelines for sur-
veillance cultures when no infection is obvious, are at the The ethical ramifications of organ transplantation and the
discretion of the Organ Procurement Agency; however, it responsibilities inherent in maintaining the spirit in which
is always useful to obtain a Gram stain of the initial speci- a donation is made are never far from the surface in the
men to allow early antibiotic tailoring. Antibiotic choice management of organ donors. With an increasing focus
is also a regional matter, as local sensitivities will change on the imbalance between support for organ donation and
regularly. However, it is important to tailor broad empiric actual registration as a donor, the concept of first-person
therapy as soon as culture results are available. In the authorization for donation, wherein the designation as
presence of renal or hepatic dysfunction, extended criteria “donor” of a given individual is held as a mandate for
donors (age), and obesity, it is important to adjust antibi- the care team to facilitate donation in appropriate cir-
otic dosing. cumstances, has been increasingly popular and has been
considered for policy implementation.84 However, recent
HEMATOLOGY publicity over manipulation of the organ transplant allo-
cation system has again brought to the forefront appropri-
Anemia should be avoided in organ donors to optimize oxy- ate allocation of a scarce and precious resource.85,86 It is
gen delivery to various organs. Interestingly, De la Cruz part of the responsibility of the care team to continue to
et al. found a positive association between number of units educate patients, family members, and the community
of blood transfused before transplantation and renal graft about both the process through which donation happens,
function; the more units transfused, the less likely the kid- and the importance of the act of donation for the donor and
ney was to develop delayed graft function.81 Recommen- the recipient. Only through education and transparency
dations are currently to target a hemoglobin level of 8 g/ will the transplantation community continue to engender
dL, in an effort to balance potential immunosuppressive patient support, and ensure an ongoing stream of willing
effects while maximizing hemodynamic stability and oxy- donors.
gen delivery.
Coagulopathy can occur for a variety of reasons in the
FUTURE TECHNOLOGIES
donor. Donors may have preexisting conditions that can
result in coagulopathy, including cardiac arrhythmias, With the current shortage of available organs for trans-
for which they are treated with anticoagulants. Iatrogenic plantation, much work is being done to extend the donor
causes need to be ruled out, including dilutional coagulopa- pool and to optimize the organs that are available for trans-
thy, acidosis, and hypothermia. The loss of hypothalamic plantation. Approaches range from the microcellular—
thermoregulation in the brain-dead donor, in addition to chemical and electrical modulation of the inflammatory
the inability to vasoconstrict or shiver, results in a poiki- cascade brought on by brain death—to the macrocellular,
lothermic donor. Close attention needs to be paid to envi- using direct peritoneal resuscitation practices to improve
ronmental control of temperature and the temperature of attainment of donor management goals and increase organ
infusing fluids to prevent the adverse effects of hypother- transplant rates.14,87,88 There has even been a resurgence
mia. Core temperature should be maintained above 34°C of interest in using ex-vivo organ management to optimize
with the use of warming blankets, fluid warmers, and ven- extended criteria organs for potential transplantation.89
tilator heating units for inhaled gases. This is an exciting time to be involved in the field of organ
Additionally, the release of cerebral proteins into the donation, as new technologies bring new possibilities for
circulation with brain death can trigger a consumptive donor management.
coagulopathy (disseminated intravascular coagulopathy)
or a hypercoagulable state.82 Any evidence of coagulopa-
thy should be identified early and treated based on bleeding Conclusion
risk. Preexisting coagulopathies (secondary to medication)
should be identified and reversed if at all possible. Donor The field of organ donor management continues to
acidosis and hypothermia should be corrected, and consid- evolve. The application of both critical care best prac-
eration should be given to a 1:1:1 ratio of packed red blood tices and organ transplantation research to the clinical
cells, fresh frozen plasma, and platelet resuscitation model management of organ donors has allowed for improve-
for patients who are bleeding (similar to trauma resusci- ments in both the number and quality of organs recov-
tation), as this has been shown to decrease both time to ered. Known as donor management guidelines, these best
hemodynamic stability and coagulopathy. If available, practices will allow practitioners to continue to provide
thromboelastography can augment traditional measures of optimal medical care to both the donor and eventually
coagulopathy, and potentially indicate patients at risk for the recipient.
102 Kidney Transplantation: Principles and Practice

References 28. Bellingham JM, Santhanakrishnan C, Neidlinger N, et al. Dona-


tion after cardiac death: a 29-year experience. Surgery 2011;150:
1. Coronado VG, Xu L, Basavaraju SV, et al. Surveillance for traumatic 692–702.
brain injury-related deaths – United States, 1997–2007. Morb Mortal 29. Monbaliu D, Pirenne J, Talbot D. Liver transplantation using donation
Wkly Rep Surveill Summ 2011;60:1–32. after cardiac death donors. J Hepatol 2012;56:474–85.
2. Novitzky D. Detrimental effects of brain death on the organ donor. 30. Oto T. Lung transplantation from donation after cardiac death (non-
Transplant Proc 1997;29:3770–2. heart-beating) donors. Gen Thorac Cardiovasc Surg 2008;56:533–8.
3. Dujardin KS, McCully RB, Wijdicks EF, et al. Myocardial dysfunction 31. Manara AR, Murphy PG, O’Callaghan G. Donation after circulatory
associated with brain death: clinical, echocardiographic, and patho- death. Br J Anaesth 2011;108:i108–21.
logic features. J Heart Lung Transplant 2001;20:350–7. 32. Suntharalingam C, Sharples L, Dudley C, et al. Time to cardiac death
4. Novitzky D, Wicomb WN, Rose AG, et al. Pathophysiology of pul- after withdrawal of life-sustaining treatment in potential organ
monary edema following experimental brain death in the chacma donors. Am J Transplant 2009;9:2157–65.
baboon. Ann Thorac Surg 1987;43:288–94. 33. de Groot YJ, Lingsma HF, Bakker J, et al. External validation of a prog-
5. van der Zee H, Malik AB, Lee BC, et al. Lung fluid and protein exchange nostic model predicting time of death after withdrawal of life support
during intracranial hypertension and role of sympathetic mecha- in neurocritical patients. Crit Care Med 2012;40:233–8.
nisms. J Appl Physiol 1980;48:273–80. 34. DeVita MA, Brooks MM, Zawistowski C, Rudich S, Daly B, Chaitin E.
6. Fisher AJ, Donnelly SC, Hirani N, et al. Enhanced pulmonary inflam- Donors after cardiac death: validation of identification criteria (DVIC)
mation in organ donors following fatal non-traumatic brain injury. study for predictors of rapid death. Am J Transplant 2008;8:432–41.
Lancet 1999;353:1412–3. 35. Brieva J, Coleman N, Lacey J, Harrigan P, Lewin TJ, Carter GL. Pre-
7. Kosleradzki M, Lisik W, Rowinski W, et al. Progress in abdominal diction of death in less than 60 minutes after withdrawal of cardio-
organ transplantation. Med Sci Monit 2011;17:RA282–91. respiratory support in potential organ donors after circulatory death.
8. Barklin A. Systemic inflammation in the brain-dead organ donor. Transplantation 2014;98:1112–8.
Acta Anaesthesiol Scand 2009;53:425–35. 36. Reich DJ, Mulligan DC, Abt PL, et al. ASTS recommended practice
9. Nagareda T, Kinoshita Y, Tanaka A, et al. Clinicopathology of kidneys guidelines for controlled donation after cardiac death organ procure-
from brain-dead patients treated with vasopressin and epinephrine. ment and transplantation. Am J Transplant 2009;9:2004–11.
Kidney Int 1993;43:1363–70. 37. Stiegler P, Sereinigg M, Puntschart A, et al. A 10 min “no-touch” time
10. Gramm H-J, Meinhold H, Bickel U, et al. Acute endocrine failure after – is it enough in DCD? A DCD animal study. Transpl Int 2012;25:481–
brain death? Transplantation 1992;54:851–7. 92.
11. Jassem W, Koo DD, Cerundolo L, et al. Leukocyte infiltration and 38. Malinoski DJ, Patel MS, Ahmed O, et al. The impact of meeting donor
inflammatory antigen expression in cadaveric and living-donor livers management goals on the development of delayed graft function in
before transplant. Transplantation 2003;75:2001–7. kidney transplant recipients. Am J Transplant 2013;13:933–1000.
12. Weiss S, Kotsch K, Francuski M, et al. Brain death activates donor 39. Abuanzeh R, Hashmi F, Dimarakis I, et al. Early donor management
organs and is associated with a worse I/R injury after liver transplan- increases the retrieval rate of hearts for transplantation in marginal
tation. Am J Transplant 2007;7:1584–93. donors. Europ J Cardio-Thorac Surg 2015;47:72–7.
13. Subramanian A, Brown D. Management of the brain-dead organ 40. Billeter AT, Sklare S, Franklin GA, et al. Sequential improvements
donor. Contemp Crit Care 2010;8:1–12. in organ procurement increase the organ donation rate. Injury
14. Novitzky D, Cooper DKC, Rosendale JD, et al. Hormonal therapy of 2012;43:1805–10.
the brain-dead organ donor: experimental and clinical studies. Trans- 41. Kotloff RM, Blosser S, Fulda GJ, et al. Management of the potential
plantation 2006;82:1396–401. organ donor in the ICU: Society of Critical Care Medicine/American
15. McKeating EG, Andrews PJ, Signorini DF, et al. Transcranial cytokine College of Chest Physicians/Association of Organ Procurement Orga-
gradients in patients requiring intensive care after acute brain injury. nizations consensus statement. Crit Care Med 2015;43:1291–325.
Br J Anaesth 1997;78:520–3. 42. Patel MS, De La Cruz S, Sally MB, Groat T, Malinoski DJ. Active donor
16. Stangl M, Zerkaulen T, Theodorakis J, et al. Influence of brain death management during the hospital phase of care is associated with more
on cytokine release in organ donors and renal transplants. Transplant organs transplanted per donor. J Am Coll Surg 2017;225:525–31.
Proc 2001;33:1284–5. 43. Audibert G, Charpentier C, Seguin-Devaux C, et al. Improvement of
17. Poppelaars F, Seelan MA. Complement-mediated inflammation and donor myocardial function after treatment of autonomic storm during
injury in brain dead organ donors. Mol Immunol 2017;84:77–83. brain death. Transplantation 2006;82:1031–6.
18. Ad Hoc Committee of the Harvard Medical School to Examine the 44. Wood KE, Becker BN, McCartney JG, et al. Care of the potential organ
Definition of Brain Death. A definition of irreversible coma. JAMA donor. N Engl J Med 2004;351:2730–9.
1968;205:337–49. 45. Dosemeci L, Yilmaz M, Cengiz M, et al. Brain death and donor man-
19. Guidelines for the determination of death: report of the medical con- agement in the intensive care unit: experiences over the last 3 years.
sultants on the diagnosis of death to the President’s Commission for Transplant Proc 2004;36:20–1.
the study of ethical problems in medicine and biochemical and behav- 46. West S, Soar J, Callaway CW. The viability of transplanting organs
ioral research. JAMA 1981;246:3. from donors who underwent cardiopulmonary resuscitation: a sys-
20. Quality Standards Subcommittee of the American Academy of Neu- tematic review. Resuscitation 2016;108:27–33.
rology. Practice parameters for determining brain death in adults. 47. Plurad DS, Bricker S, Neville A, Bongard F, Putnam B. 2012 Arginine
Neurology 1995;45:1012–4. vasopressin significantly increases the rate of successful organ pro-
21. Wijdicks EF, Varelas PN, Gronseth GS, et al. Evidence-based guideline curement in potential donors. Am J Surg 2012;204:856–61.
update: determining brain death in adults. Report of the quality stan- 48. Gottmann U, Brinkkoetter PT, Bechtler M, et al. Effect of pre-treatment
dards subcommittee of the american academy of neurology. Neurol- with catecholamines on cold preservation and ischemia/reperfusion-
ogy 2010;74:1911–8. injury in rats. Kidney Int 2006;70:321–8.
22. Wijdicks EF, Pfeifer EA. Neuropathology of brain death in the modern 49. Al-Khafaji A, Elder M, Lebovitz DJ, et al. Protocolized fluid therapy in
transplant era. Neurology 2008;70:1234–7. brain-dead donors: the multicenter randomized MOnIToR trial. Inten-
23. Conci F, Procaccio F, Arosio M, et al. Viscero-somatic and viscero- sive Care Med 2015;41:418–26.
visceral reflexes in brain death. J Neurol Neurosurg Psychiatry 50. D’Aragon F, Dhanani S, Lamontagne F, et al. Canada-donate study
1986;49:695–8. protocol: a prospective national observational study of the medical
24. Saposnik G, Basile VS, Young GB. Movements in brain death: a sys- management of deceased organ donors. BMJ Open 2017;7.
tematic review. Can J Neurol Sci 2009;36:154–60. 51. Angel LF. Impact of a lung transplantation donor-management pro-
25. Wijdicks EF, Rabinstein AA, Manno EM, et al. Pronouncing brain tocol on lung donation and recipient outcomes. Am J Respir Crit Care
death: contemporary practice and safety of the apnea test. Neurology Med 2006;174:710–6.
2008;71:1240–4. 52. Mascia L, Pasero D, Slutsky AS, et al. Effect of a lung protective strategy
26. Weill D. Donor criteria in lung transplantation: an issue revisited. for organ donors on eligibility and availability of lungs for transplan-
Chest 2002;121:2029–31. tation a randomized controlled trial. JAMA 2010;304(23):2620–7.
27. Barlow AD, Metcalfe MS, Johari Y, et al. Case-matched comparison of 53. Reilly PM, Grossman M, Rosengard BR, et al. Lung procurement from
long-term results of non-heart beating and heart-beating donor renal solid organ donors: role of fluid resuscitation in procurement failures.
transplants. Br J Surg 2009;96:685–91. Chest 1996;110. 220S-7S.
6 • Brain Death and Cardiac Death: Donor Criteria and Care of Deceased Donor 103

54. Venkateswaran RV, Patchell VB, Wilson IC, et al. Early donor man- 72. Blasi-Ibanez A, Hirose R, Feiner J, et al. Predictors associated with ter-
agement increases the retrieval rate of lungs for transplantation. Ann minal renal function in deceased organ donors in the intensive care
Thorac Surg 2008;85:278–86. unit. Anesthesiology 2009;110:333–41.
55. Minambres E, Perez-Villares JM, Terceros-Almanza L, et al. An inten- 73. Sally MB, Ewing T, Crutchfield M, et al. Determining optimal thresh-
sive lung donor treatment protocol does not have negative influ- old for glucose control in organ donors after neurologic determination
ence on other grafts: a multicentre study. Euro J Cardio-Thorac Surg of death: A United Network for Organ Sharing Region 5 Donor Man-
2016;49(6):1719-1724. agement Goals Workgroup prospective analysis. J Trauma Acute Care
56. Linos K, Fraser J, Freeman WD, et al. Care of the brain-dead organ Surg 2014;76:62–9.
donor. Curr Anaesth Crit Care 2007;18:284–94. 74. Gores PF, Gillingham KJ, Dunn DL, et al. Donor hyperglycemia as a
57. Figueras J, Busquets J, Grande L, et al. The deleterious effect of donor minor risk factor and immunologic variables as major risk factors for
high plasma sodium and extended preservation in liver transplanta- pancreas allograft loss in a multivariate analysis of a single institu-
tion: a multivariate analysis. Transplantation 1996;61:410–3. tion’s experience. Ann Surg 1992;215:217–30.
58. Kazemeyni SM, Esfahani F. Influence of hypernatremia and polyuria 75. Mehta SR, Logan C, Kotton CN, Kumar D, Aslam S. Use of organs
of brain-dead donors before organ procurement on kidney allograft from donors with bloodstream infection, pneumonia, and influenza:
function. Urol J 2008;5:173–7. results of a survey of infectious diseases practitioners. Transpl Infect
59. Totsuka E, Fung U, Hakamada K, et al. Analysis of clinical vari- Dis 2017;19.
ables of donors and recipients with respect to short-term graft out- 76. Domínguez-Gil B, Delmonico FL, Shaheen FAM, et al. The critical
come in human liver transplantation. Transplant Proc 2004;36: pathway for deceased donation: reportable uniformity in the approach
2215–8. to deceased donation. Transpl Int 2011;24:373–8.
60. Totsuka E, Fung JJ, Ishii T, et al. Influence of donor condition on post- 77. Trotter PB, Robb M, Hulme W, et al. Transplantation of organs from
operative graft survival and function in human liver transplantation. deceased donors with meningitis and encephalitis: a UK registry anal-
Transplant Proc 2000;32:322–6. ysis. Transplant Infect Dis 2016;18:862.
61. Powner D. Factors during donor care that may impact liver transplant 78. Coilly A, Samuel D. Pros and cons: usage of organs from donors
outcomes. Prog Transplant 2004;14:241–9. infected with hepatitis C virus – revision in the direct-acting antiviral
62. Novitzky D. Novel actions of thyroid hormone: the role of triiodothyro- era. J Hepatology 2016;64:226.
nine in cardiac transplantation. Thyroid 1996;6:531–6. 79. Levitsky J, Formica RN, Bloom RD, et al. The American society of
63. Salim A, Martin M, Brown C, et al. Using thyroid hormone in brain- transplantation consensus conference on the use of hepatitis c viremic
dead donors to maximize the number of organs available for trans- donors in solid organ transplantation. Am J Transplantat 2017;17(11):
plantation. Clin Transplant 2007;21:405–9. 2790–2802.
64. Salim A, Vassiliu P, Velmahos GC, et al. The role of thyroid hormone 80. Lumbreras C, Sanz F, Gonzalez A, et al. Clinical significance of donor-
administration in potential organ donors. Arch Surg 2001;136: unrecognized bacteremia in the outcome of solid-organ transplant
1377–80. recipients. Clin Infect Dis 2001;33:722–6.
65. Novitsky D, Mi Z, Sun Q, Collins JF, Cooper DKC. thyroid hormone 81. De la Cruz JS, Sally MB, Zatarain JR, et al. The impact of blood transfu-
therapy in the management of 63,593 brain-dead organ donors: a sions in deceased organ donors on the outcomes of 1,884 renal grafts
retrospective analysis. Transplantation 2014;98:1119. from United Network for Organ Sharing Region 5. J Trauma Acute
66. Rosendale JD, Kauffman HM, McBride MA, et al. Hormonal resuscita- Care Surg 2015;79.
tion yields more transplanted hearts, with improved early function. 82. Laroche M, Kutcher ME, Huang MC, et al. Coagulopathy after trau-
Transplantation 2003;75:1336–41. matic brain injury. Neurosurgery 2012;70:1334–45.
67. Sharpe MD, van Rassel B, Haddara W. Oral and intravenous thyrox- 83. Powner D. Thromboelastography during adult donor care. Prog
ine (T4) achieve comparable serum levels for hormonal resuscitation Transplant 2010;20:163–8.
protocol in organ donors: a randomized double-blinded study. Can J 84. Human Transplantation (Wales) Act. 2013 (anaw 5). Available
Anesth/J Can Anesth. 2013;60:998–1002. online at: http://www.legislation.gov.uk/anaw/2013/5/pdfs/anaw_
68. Kotsch K, Ulrich F, Reutzel-Selke A, et al. Methylprednisolone ther- 20130005_en.pdf. Accessed April 22, 2019.
apy in deceased donors reduces inflammation in the donor liver and 85. Shaw D. Lessons from the German organ donation scandal. JICS
improves outcome after liver transplantation: a prospective random- 2013;14(3):200.
ized controlled trial. Ann Surg 2008;248:1042–50. 86. Caplan A. Bioethics of organ transplantation. Cold Spring Harb Per-
69. Follette DM, Rudich SM, Babcock WD. Improved oxygenation and spect Med 2014;4.
increased lung donor recovery with high-dose steroid administration 87. Hoeger S, Bergstraesser C, Selhorst J, et al. Modulation of brain dead
after brain death. J Heart Lung Transplant 1998;17:423–9. induced inflammation by vagus nerve stimulation. Am J Transplant
70. D’Aragon F, Belley-Cote E, Agarwal A, et al. Effect of corticosteroid 2010;10:477–89.
administration on neurologically deceased organ donors and trans- 88. Smith JW, Matheson PJ, Morgan G, et al. Addition of direct peritoneal
plant recipients: a systematic review and meta-analysis. BMJ Open lavage to human cadaver organ donor resuscitation improves organ
2017;7. procurement. J Am Coll Surg 2015;220(4):539–47.
71. Pinsard M, Ragot S, Mertes PM, et al. Interest of low-dose hydrocorti- 89. Cypel M, Keshavjee S. Strategies for safe donor expansion: donor man-
sone therapy during brain-dead organ donor resuscitation: the COR- agement, donations after cardiac death, ex-vivo lung perfusion. Curr
TICOME study. Crit Care 2014;18:R158. Opin Organ Transplant 2013;18:513–7.
7 Medical Evaluation of
the Living Donor
MATTHEW J. ELLIS, BRADLEY HENRY COLLINS, BRIAN EZEKIAN, and
STUART J. KNECHTLE

CHAPTER OUTLINE History of Living Donation and Donor- Kidney Paired Donation
Related Ethics ABO Incompatibility
The Rationale for Living Donation Medical Evaluation
Matching Donor and Recipient Surgical Evaluation
Global Variations in Living Donor Kidney Long-Term Outcomes After Living
Transplantation Donation
Organ Supply Problem Financial Considerations
Quality of the Living Donor Kidney Living Donor Quality Initiatives
Types of Living Donors

to living donor renal transplants. The arrival of cyclospo-


History of Living Donation and rine and muromonab-CD3 (OKT3) in the 1980s further
Donor-Related Ethics enhanced outcomes with both deceased donor and living
donor transplantation. Laparoscopic live donor nephrec-
The first long-term successful organ transplant was a liv- tomy, initially described by Ratner in 1995, reduced
ing donor kidney transplant between monozygotic twin the pain and suffering associated with live donation and
brothers performed in 1954 at the Peter Bent Brigham resulted in a substantial growth of living kidney donation
Hospital in Boston.1 This procedure marked the first time in the US.3 Improvements in outcomes and reduction in
in history that a healthy person underwent a major surgi- morbidities of surgery led to a growth in living kidney dona-
cal procedure that they did not need, solely for the benefit tion to a maximal number in 2004 of 6647. Unfortunately,
of another person, and this reality surrounded the ethical the number of living donor transplants declined to 5626 in
discussion of the case. The success of the procedure from 2015.4 Reasons for the decline in total numbers of living
an immunologic perspective was predicated on the genetic donor nephrectomy, despite the continual growth of the
identity of the donor and recipient, by obstetric records of recipient waitlist, are difficult to attribute accurately, but
the donor and recipient’s birth, and by skin grafting before may include increasing reliance on deceased donor kidney
the kidney transplant to confirm compatibility. Because this transplantation. Utilization of living donors varies by geo-
transplant predated the development of dialysis, it was truly graphic region, race and culture, and transplant program.
a life-saving procedure, given the end-stage renal disease of Additionally, the increasing body mass index (BMI) of the
the recipient. The donor lived another 56 years with a sin- US population discourages the use of living donors.
gle kidney without any apparent sequelae. Additional ethi- As living donation comes under increasing regulatory
cal considerations and unknowns at the time were whether scrutiny, long-term medical follow-up has been mandated
the twin donor would develop renal failure either because for such donors. Paired kidney donation has developed as
of the procedure (considered very unlikely) or because of a means of maximizing benefits and possibilities of living
development of the same kidney disease as his genetically donation. In an attempt to increase the number of people
identical brother (unlikely but nevertheless possible). This eligible to be a living donor, donors who are older, have a
transplant was followed by a small but growing number higher BMI, are on one antihypertensive medication, and
of similar living donor transplants between closely related have other more complex medical conditions have been
family members, even while immunosuppressive drug ther- considered. Attitudes toward covering the medical costs
apy for transplantation was in its infancy. of living donation vary around the world (see section on
Although living donor renal transplants initially were the Financial Considerations). The implementation of living
only option, deceased donor renal transplantation began kidney donation reflects both medical and societal attitudes
shortly thereafter and grew considerably with the advent toward risk and cost sharing. However, one constant reality
of brainstem death criteria.2 The development of immuno- in the field is that the best long-term outcomes have been
suppressive drug therapy, consisting initially of Imuran, achieved using living donors, likely because of the superior
steroids, and antilymphocyte globulin, allowed the growth quality of the kidneys and short preservation times that
of deceased donor renal transplantation as an alternative minimize graft injury associated with transplantation.
104
7 • Medical Evaluation of the Living Donor 105

The Rationale for Living Donation because even a poorly matched living donor kidney may
perform better than a deceased donor kidney. However,
Although the survival benefit of kidney transplantation is each additional HLA mismatch comes with a significant
superior to any other form of renal replacement therapy, linear adverse effect on allograft survival.11 Furthermore,
the allograft’s source confers additional benefits. For exam- kidneys from deceased donors are associated with the low-
ple, kidney transplantation before the initiation of dialysis, est effect of HLA mismatch, kidneys from living related
also known as preemptive transplantation, is associated donors are associated with an intermediate effect from HLA
with higher graft and patient survival compared with those mismatch, and kidneys from living unrelated donors are
recipients who receive their kidneys after initiating dialy- associated with the highest effect from HLA mismatch. ABO
sis.5 This survival benefit is most pronounced in the liv- incompatibility (ABOi) may serve as another immunologic
ing donor kidney population, because 31% of living donor barrier to living donor kidney transplantation. However,
transplants performed in the US in 2015 were preemptive, 1-, 3-, and 5-year ABOi graft survival rates with peritrans-
whereas only 9% of deceased donor kidney transplants plant immunomodulatory therapies are now comparable to
were performed in patients before chronic dialysis.4 Stud- United Network for Organ Sharing data for compatible live
ies have shown that renal replacement therapy by kidney donor kidney transplants (see section on ABO Incompatibil-
transplantation is less costly than dialysis, so it is likely ity).12–15 Age disparity between donors and recipients also
that the increased use of preemptive transplantation in plays a role in matching. Theoretically, age matching is of
living donation decreases the financial burden associated considerable importance, because younger kidney trans-
with renal failure.6 Recipients of living donor grafts do not plant recipients receiving older allografts may outlive their
wait as long for their organs as the recipients of grafts from allografts and require retransplantation, whereas older
deceased donors, who may wait for years, depending on recipients may die of other causes before their allograft fails,
the region in which they are listed and their level of pre- which both lead to suboptimal organ usage. Additionally,
formed antibodies to the major histocompatibility complex. recent data have demonstrated that increasing living donor
Another advantage of living donor kidney transplantation age is associated with reduced allograft survival, particu-
is virtual absence of delayed graft function (DGF) as a result larly in longer-term follow-up.16,17
of relatively short cold ischemia times and the lack of the
physiologic perturbations associated with brain death that GLOBAL VARIATIONS IN LIVING DONOR KIDNEY
may lead to allograft dysfunction. The avoidance of DGF is TRANSPLANTATION
advantageous as it has been associated with an increased
incidence of acute rejection and linked to the development Comprehensive global assessments of living kidney dona-
of chronic allograft nephropathy, which both lead to infe- tion have seldom been performed because of the varying
rior graft survival.7,8 Kidneys obtained from living donors transparency of practices between countries. Recently
outperform those from deceased donors at all time points global trends of 69 countries with available national data
as evidenced, for example, by a 5-year graft survival rate of have been examined.18 In 2006 about 27,000 legal liv-
>85% for living donors versus 75% for deceased donors.5 ing donor kidney transplants were performed worldwide,
Finally, when surgical complications, primary nonfunc- accounting for nearly 40% of all kidney transplants. In
tion, and other causes of early graft loss are excluded, the addition to legal practices, the World Health Organization
1-year conditional half-life of living donor transplants was estimates a significant fraction of kidney transplants world-
15.3 years for transplants performed from 2009 to 2010, wide involve unacceptable or illegal practices.19 In general,
whereas the half-life of deceased donor grafts was 12.5 the number of living kidney donor transplants has grown,
years during that same period.9 with 62% of countries reporting at least a 50% increase in
Living donor kidney transplantation has a number of volume from the preceding decade. The highest volume of
other benefits that are harder to quantify. The procedures living donor kidney transplants was performed in the US
are scheduled electively, usually at the discretion of the (6435), Brazil (1768), Iran (1615), Mexico (1459), and
donor, so they are performed during the day when the Japan (939).
operative team is rested and all ancillary services are fully The current proportion of all kidney transplants from
staffed. Living donation allows for the optimization of any living and deceased donors shows substantial geographic
medical comorbid conditions that might affect recipient variation.20 Most European countries, such as Poland,
outcome adversely. Theoretically, every living donor trans- Spain, Italy, France, and Germany, derive a majority of
plant performed leaves a deceased donor kidney for another their kidney allografts from deceased donors; thus the
recipient on the transplant list. However, in the US, despite proportion of living donor kidney transplantation is small
the growth of the waiting list, the number of living donor (<20%). In countries such as the US, Canada, United King-
kidney transplants continues to decline, and there were dom, Sweden, and Switzerland, the proportion of living
1000 fewer live donors in 2016 than in 2004.10 donor kidney transplants is increasing (now 20%–50%)
alongside robust deceased donor networks. In countries
such as Brazil, Korea, Turkey, Japan, and Iran, the majority
MATCHING DONOR AND RECIPIENT
of kidney transplants are from living donors (>50%). Nota-
Optimal human leukocyte antigen (HLA) matching of bly in Japan, laws that were in place until 2010 required
a living kidney donor with a recipient influences long- family consent for organ recovery despite the documented
term allograft survival. A few key donor/recipient factors will of a donor, a practice that significantly hampered rates
have emerged as predictors of long-term outcomes. Ideal of deceased donation on the basis of cultural norms. More-
HLA matching is not required for living kidney donation, over, Iran is the only country in the world with a paid living
106 Kidney Transplantation: Principles and Practice

unrelated kidney donation program, which accounts for deceased donor organ allocation and provide the best avail-
about 75% of all its kidney transplants.21 Finally, in coun- able basis for patient and provider decisions regarding
tries such as Egypt, Jordan, Pakistan, Oman, and Iceland, accepting or declining a given kidney offer. Recently the
all kidney transplants are from living donors, because there first Living Kidney Donor Profile Index (LKDPI) was devel-
is no deceased donor program in place. oped.27 The national data used in developing the LKDPI
demonstrated that donor age >50 years, African Ameri-
can race, ABO incompatibility, and HLA-B and HLA-DR
ORGAN SUPPLY PROBLEM
mismatches were associated with worse graft survival,
The need for kidney transplants continues to far exceed the whereas higher donor estimated glomerular filtration rate
demand, although the absolute number of registrants on (eGFR), donors unrelated to the recipient, and male-to-male
the list has plateaued over the past few years to just under donation were associated with better graft survival. This
100,000 in the US.10 This observation has been attributed system allows direct comparison between two living donor
to a combination of factors, including implementation of kidneys, or comparison between a living donor kidney and
the kidney allocation system (KAS).4 Now that waiting one from a deceased donor. The LKDPI is particularly use-
time is calculated from the date that dialysis was initiated, ful in decision making for recipients fortunate enough to
there is no benefit to maintaining a patient with end-stage have multiple potential living donors, or those who receive
renal disease (ESRD) on the waitlist in an inactive status deceased donor kidney offers while potential living donors
while outstanding medical, financial, or psychosocial issues are under evaluation. Additionally, this index is useful in
are addressed. Over the past decade, the number of kidney kidney paired donation (KPD). Incompatible pairs entering
transplants performed each year has generally increased; a KPD system can use this index to decide which alterna-
however, gains in the number of kidneys recovered from tive living donors are acceptable, and, in the case of paired
deceased donors have been offset by a decline in number donation, to evaluate the possibility of receiving a deceased
of living donors. For example, in 2004, 16,007 patients donor kidney in exchange for their living donor donating to
underwent kidney transplantation (9359 from deceased another person on the waiting list.28
donors and 6648 from living donors).10 The number of
allografts increased to 19,060 in 2016. However, that TYPES OF LIVING DONORS
growth was driven entirely by an increase in the number
of kidneys recovered from deceased donors (13,431) as the When educating patients and their families about kid-
number of living donor grafts declined to 5629.10 Commu- ney transplantation, it is imperative that the advantages
nity outreach education initiatives, and programs through of the living donor are emphasized: shorter waiting time,
state divisions of motor vehicles where millions of drivers better quality kidney/function, and longer allograft sur-
have provided first-person donor consent, have likely led vival. Many patients are still under the impression that
to an increase in deceased donors, but a corresponding living donors must be blood relatives; however, transplant
increase in living donors has yet to be realized.22 The cur- professionals should encourage recipients to expand their
rent opioid epidemic in the US has also yielded an increase searches. In this era of social media, a wider net can be cast,
in deceased organ donors despite fears of disease transmis- although the ethical considerations are still under debate.29
sion blunting the potential. In this era of sophisticated dis- Patients should also be taught that any interested, reason-
ease detection methodologies (e.g., nucleic acid testing), the able donor should present for screening, because blood type
risk of dying without being transplanted is higher than the matching is not always necessary given desensitization
risk of unintentional disease transmission.23 protocols and the expanding role of paired donation. Once
Another sequela of the organ donor shortage is patient judged with skepticism, altruistic donors are now coveted
mortality while on the waiting list. Of the approximately because the paired donation experience has demonstrated
99,000 patients waiting for kidney transplantation in their importance in initiating transplant chains.30 Some
2015, almost 5000 died.4 The role for living donor kidney centers take the patient out of the role of soliciting for a
transplantation to improve waitlist mortality may be lim- kidney and transfer that responsibility to an advocate or
ited given the decreasing number of individuals willing to “champion.”31 The effect that these efforts will have on the
donate and the large percentage of living donor kidneys living kidney donor pool is not known at this time.
that are transplanted preemptively. Of the 31,672 patients
removed from the kidney transplant waitlist in 2015 KIDNEY PAIRED DONATION
(because of transplantation, medical deterioration, psycho-
social reasons, or death), 18% received kidneys from liv- KPD is now an option for recipients with willing and medi-
ing donors whereas 39% underwent transplantation from cally fit donors who are deemed poorly compatible on the
deceased donors.4,10 basis of immunologic (e.g., ABO or HLA incompatibility) or
other factors (e.g., age, gender differences, BMI mismatch),
which affects up to 30% of cases.32 In these circumstances,
QUALITY OF THE LIVING DONOR KIDNEY
incompatible donor/recipient pairs can be entered into a pool
The need for risk stratification of deceased donor organs of one or more additional incompatible pairs who are in a
has long been recognized. Accordingly, an evolution has similar situation. Subsequently, by exchanging donors, all
occurred from the simplistic expanded criteria donor (ECD) recipients have the potential to receive a compatible organ
classification to the more sophisticated Kidney Donor match. Early paradigms consisted solely of same-day, two-
Risk Index (KDRI) and related Kidney Donor Profile Index way swaps performed at the same location. However, the
(KDPI).24–26 In aggregate, these tools have helped shape demonstrated safety of shipping living donor kidneys from
7 • Medical Evaluation of the Living Donor 107

one center to another33 and the addition of nondirected protocols variably include recipient treatment with one or
donors to the donor pool has allowed feasibility of KPD across more of the following: (1) plasmapheresis to remove anti-
much larger geographic areas.34 The US is unique in that A/B antibodies, (2) intravenous immunoglobulin, and (3)
single-center, multicenter, and national programs that oper- B cell–depleting therapies such as rituximab or splenec-
ate independently of one another coexist.35 Programs have tomy, which are combined with powerful maintenance
uniformly reported at least equivalent graft survival rates immunosuppression posttransplantation.42 The ultimate
compared with traditional non-KPD living donor trans- goal of these protocols is to decrease the level of anti-A/B
plants.36–38 A single, national KPD system that includes antibodies below a safe threshold in the immediate post-
the need for uniform tissue-typing platforms, computerized transplant period. After an approximately 2-week period
matching algorithms, and a standardized organ acquisition of engraftment, rebound anti-A/B antibody production
charge has been proposed,39 but faces several logistical chal- inevitably occurs, but this rebound does not appear to cause
lenges that have impeded implementation. Such a system significant injury to the kidney allograft (a process termed
exists in other geographic areas, including the United King- accommodation).43 Increasing experience with ABOi kidney
dom, where a national sharing scheme has been established transplantation has led to outcomes that are now equiva-
for living donor kidneys. Matching runs are undertaken lent to ABO-compatible transplantation in both pediatric
every 3 months and, from January 2018, all nondirected and adult patient populations.14,15
altruistic donors in the UK are entered into the scheme unless
the donor takes exception to this. This has led to a steady
increase in the number of kidneys transplanted through the Medical Evaluation
scheme, with results that are comparable to related living
donor transplantation40 (see Chapter 23 for more details). Donors are evaluated in a multidisciplinary manner that
includes an independent living donor advocate, nephrolo-
gist, surgeon, social worker, medical psychologist, and
ABO INCOMPATIBILITY
financial counselor. The medical evaluation has multiple
ABOi transplants had long been considered a contraindica- components, some of which can start before the patient is
tion to kidney transplantation with Hume et al. in the 1950s seen at the transplant center, and culminates in the review
remarking, “[W]e do not feel that renal transplantation in of all data by a multidisciplinary team and a final decision
the presence of blood incompatibility is wise.”41 However, regarding candidacy. These components are listed in Box
the increasing organ shortage has stimulated the devel- 7.1, but the broad categories include: (1) a thorough medi-
opment of strategies to allow transplantation across this cal history and physical examination including family his-
immunologic barrier. Current ABOi immunomodulation tory focused on renal disease; (2) general health laboratory

BOX 7.1 Components of the Living Donor Evaluation


History and Physical Examination Urinalysis and culture
Interview with focus on renal disease and family history of renal disease Electrocardiograph
Detailed health questionnaire Chest x-ray
Donor education and consent Crossmatch with recipient
History and physical examinations by transplant nephrologist and Human leukocyte antigen (HLA) typing of the donor
surgeon Identify Transmissible Infectious Disease
Multiple complete vital signs
Evaluation by mental health expert Human immunodeficiency virus, hepatitis B and C
Interview with independent living donor advocate Rapid plasma reagin (syphilis screen)
Testing for tuberculosis (TB)—TB skin testing or QuantiFERON-TB Gold
Laboratory Testing to Evaluate Renal Function and Determine Testing for Strongyloides, Trypanosoma cruzi, and West Nile virus for
Immunologic Compatibility donors from endemic areas
Blood pressure—two to three separate measurements Evaluation of Renal Anatomy With Cross-Sectional Imaging
Additional 24-hour blood pressure monitoring as indicated
Urine protein assessment (via 24 h urine or spot protein to creati- Abdominal imaging using computed tomography angiography or
nine ratio) magnetic resonance angiography
Glomerular filtration rate assessment (via 24 h creatinine clearance, Age-Appropriate Health Screening, Including Cancer Screening
iothalamate clearance, or radioisotope clearance)
Oral glucose tolerance test and/or HbA1c Prostate-specific antigen (recommendations based on donor age
Metabolic workup if previous history of renal stones and family history)
Donor ABO typing Gynecologic examination with Papanicolaou smear
Complete blood count with platelet count and differential Colonoscopy
Comprehensive metabolic panel to include fasting serum glucose Mammogram
and measurement of transaminases Pregnancy test if indicated
Fasting lipid profile Echocardiography and cardiac stress testing as indicated
Coagulation studies to include the prothrombin time, international Pulmonary function studies and computed tomography scanning
normalized ratio, and partial thromboplastin time of the chest as indicated

Modified from kidney transplantation, 7th ed., Chapter 7, MacConmara and Newell.
108 Kidney Transplantation: Principles and Practice

evaluation with focus on renal function, HLA typing, HLA The primary surgical approach to living kidney donation
antibody screening, ABO typing, and screening for trans- is laparoscopic, and 97% of living donor kidneys in 2015
missible infection; (3) age-appropriate health and can- were retrieved by this method.4 Many surgeons prefer the
cer screening; and (4) cross-sectional imaging of kidney hand-assisted technique. However, almost 40% of cases
anatomy. are performed completely laparoscopically. The kidney is
The medical history focuses on possible comorbidities extracted from the abdomen via either a periumbilical or
such as hypertension, diabetes, cardiovascular or cerebro- Pfannenstiel incision. The choice of the kidney is usually
vascular disease that must be quantified. Multiple blood driven by anatomic considerations, with preference for the
pressure readings are obtained to provide a reliable base- left because of the longer, more substantial renal vein (for
line. Laboratory profile includes the tests shown in Box more details on donor nephrectomy see Chapter 8). The
7.1 and specifically includes urine studies to estimate renal presence of multiple renal arteries may present challenges
function and to evaluate for proteinuria. In the US, measur- at implantation, so both the recovering and implanting
ing renal function is a requirement and can be completed by surgeons should review preoperative imaging. Small upper
collecting 24-hour urine samples to calculate the 24-hour pole arteries can be ligated should they pose undue risk dur-
creatinine clearance, or glomerular filtration rate (GFR) ing implantation (prolonged warm ischemia); however,
can be measured by nuclear imaging studies. every effort should be made to preserve lower pole arteries,
Immunologic testing includes ABO and HLA typing of because they usually provide blood supply to the ureter.
the donor to allow interpretation of compatibility with the Techniques include anastomosis to the main renal artery
recipient, choosing between donors, if there are more than on the back table or separate arterial anastomoses in the
one, or pairing the donor with the most appropriate recipi- recipient. To minimize warm ischemia time, some surgeons
ent in the case of paired exchange. Measurement of possible reperfuse the kidney once the main arterial anastomosis
communicable infectious diseases such as hepatitis B and has been completed, then perform the second anastomosis
C and HIV is necessary. Screening for tuberculosis (TB) is at a more distal site on the native iliac artery.
highly recommended using TB skin testing or QuantiF- Immediate complications of living kidney donation that
ERON-TB Gold. Donors who test positive should be treated may require reoperation include injury to intraabdominal
for 6 to 9 months before proceeding with donation. Screen- viscera, bleeding, and wound dehiscence (Table 7.1).46
ing for syphilis is also highly recommended, and, if there is Potential donors should also be made aware of other compli-
history of travel to Central or South America, Chagas’ dis- cations including lymphatic leak (chylous ascites), wound
ease as well. West Nile virus screening is also advised. infection, hernia, and deep venous thrombosis. Death
Abdominal imaging is usually delayed until donors resulting from donor nephrectomy is rare; however, donors
pass the medical and laboratory screening stages to avoid should be counseled that it is possible. Between 2011 and
unnecessary risk and cost. Depending on local expertise, 2015 in the US, 7 out of 28,291 living kidney donors died
either computed tomography (CT) angiography or mag- (approximate incidence of 1 in 4000) because of what was
netic resonance angiography is performed to identify vas- termed “donation-related” causes.4
cular anatomy, size of kidneys, and the anatomy of the Most living donors are discharged within 2 days of sur-
urinary collecting system. gery. In an attempt to decrease hospital stay prolonged by
All donors older than 50 and those older than 40 with
cardiovascular risk factors should undergo appropriate car-
diovascular diagnostic studies and imaging, such as stress TABLE 7.1 Donor Complication Risks
testing. All donors are screened with chest x-ray, and those Risk (Percentage of
with a history of smoking, lung disease, or abnormal chest Donations Unless Stated)
x-ray are screened with chest CT scan as well. Guidelines
Mortality 0.02–0.03
for cancer screening are published by the American Cancer
Society and others according to age and gender, and guide MAJOR COMPLICATION
the performance of these screening tests for donors.44 Bleeding 2.2
Bowel obstruction 1.0
Vascular injury 0.27
Open conversion 0.7–1.1
Surgical Evaluation Reoperation 0.52
Blood transfusion 0.4
The surgeon’s unique role is to assess the operative risk of Wound infection 2.1
Urinary tract infection 4.5
the donor. Factors that can contribute to increased morbid- Readmission (including nausea, 2–5
ity include prior abdominal operations and elevated BMI. vomiting, gastroenteritis, abdomi-
Transplant centers have various thresholds for maximum nal pain, ileus, bowel obstruction)
BMI in donors, with some extending their upper limit to 35 Hernia repair 0.8
kg/m2.45 In this population, consideration should be given End-stage renal failure Hazard ratio 11.38
Additional risk in obese, elderly,
to the increased risk of hypertension, diabetes, heart dis- and African American donors
ease, and renal disease before acceptance as a donor. The Hypertension 15–25
Scientific Registry of Transplant Recipients (SRTR) obesity
rate for living donors in 2015 was 20% with a BMI range Data from OPTN/SRTR 2015 Annual Data Report. In: Scientific registry of
transplant recipients 2017; Mjøen G, Hallan S, Hartmann A, et al. Long-term
of 30 to 35 kg/m2 and 3% over 35 kg/m2.4 Elevated BMI risks for kidney donors. Kidney Int 2014;86:162–7; Lam NN, Lentine KL,
is also a risk factor for prolonged surgical time and wound Levey AS, et al. Long-term medical risks to the living kidney donor. Nat Rev
complications. Nephrol 2015;11:411–9.
7 • Medical Evaluation of the Living Donor 109

gastrointestinal complications (constipation, etc.), some Rehospitalizations: Patients who donate a kidney are
□ 

centers have applied an enhanced recovery after surgery at increased risk of subsequent hospitalizations in the 3
(ERAS) protocol to living donors. This includes preoperative years after donation; some of these are related to dona-
bowel preparation, measured use of crystalloids in the oper- tion, whereas other admissions are not.
ating room, and narcotic-free pain control. Convalescence Mortality: Fortunately, there have been no clear signals
□ 

for donors follows that of most major abdominal operations for changes in mortality compared with the general pop-
and includes avoiding driving and weight-lifting restric- ulation, with the caveat that most studies’ duration is not
tions. However, walking is encouraged. Return to work long enough to credibly state that there is no change.
  
depends on clinical assessment of recovery and the type of
employment (desk job vs. construction). The rehospitaliza- In 2013 the United Network of Organ Sharing (UNOS)
tion rate for living donors is 5% at 1 year.4 mandated that centers report on patients’ laboratory and
Given the thorough medical evaluations that living clinical outcomes and follow-up at 6, 12, and 24 months
donors are subjected to, the incidence of renal failure is low. postdonation. Before the 2013 UNOS mandate, fewer than
However, as living donor criteria are extended, the inci- 50% of patients had clinical data reported, which has more
dence may increase. There are calculators that clinicians recently improved to approximately 65%; similarly, less than
should employ to determine each candidate’s lifetime risk 40% of patients had laboratory data reported, which has
for renal failure.47 also improved to 56%.53,54 Patient factors associated with
higher rates of follow-up include female gender, younger
age, college or higher education level, and residence in the
Long-Term Outcomes After Living same state as the performing transplant center. Center char-
Donation acteristics associated with limited follow-up include lack of
reimbursement and a lack of transplant center resources to
Follow-up after living donation and characterization of accomplish follow-up.54 Donor surveys suggest that follow-
the long-term outcomes have historically been limited by up would be better if postdonation communication was
the short duration of follow-up, the high number of donors more regular, predonation education about the need and
lost to follow-up, and the small number of minority donors; effect of follow-up was improved, and more resources were
these limitations interfere with living donor programs accu- available to assist in covering donation-related follow-
rately describing the inherent risks of kidney donation.48 up expenses.55 Although follow-up is commonly lacking,
One of the most worrisome outcomes after living dona- increased follow-up success can be achieved. Single-center
tion is the development of ESRD. Current estimates in reports describe living donor program improvement projects
the US and Norway document a 7- to 11-fold increase that resulted in dramatic (14-fold) increases in completed
in risk of ESRD after donation.49,50 White donors have 2-year follow-up and reporting,56 with modest financial
up to a 6.1% increased risk for proteinuria develop- impact to the center (less than $200 per patient), despite
ment (a known risk factor for chronic kidney disease), increased services offered to the patient.
whereas as many as 30% of donors will develop GFRs An important consideration related to postdonation
less than 60 mL/min/1.73 m2.51 The risks for protein- medical complications includes the timing of these events.
uria and “low” GFR are augmented by elevated systolic Whereas 2-year follow-up is required after donation in
and/or diastolic predonation blood pressure, increased the US, very few of the important complications, namely
age, decreased predonation kidney function, and higher increased blood pressure, increased urinary protein excre-
BMI (for each unit increase in BMI over 30, there is a 3% tion, or a fall in GFR, occur during that time. Furthermore,
to 10% increase rate of new proteinuria and decreased long-term follow-up is challenging, even in countries with
GFR, respectively).51 universal, single-payer health insurance.57 Importantly,
In addition to renal-specific outcomes, donors may follow-up care is even more important in the current dona-
experience a number of other long-term risks. Matas and tion climate, in which transplant centers seek more living
colleagues recently summarized what is known about post- donors and allow more comorbid condition(s), as attention
donation, non-ESRD risks.52 Although many of these out- is paid to narrowing the gap between the increasing num-
comes are known to be only associations, several risks have ber of waitlisted patients and the relatively flat number of
concrete, causal connections. transplants performed each year. Currently, donors more
  
often tend to be unrelated to the recipient, older, insured,
Cardiovascular risk: In the first few years after donation,
□  employed, college educated, have a history of smoking and/
a donor’s GFR most often increases, which is indepen- or hypertension, lower GFR pretransplant, and living out-
dently associated with increases in left ventricular mass, side of the US.
decreased aortic distensibility, and increased troponin An important component to medical and/or psychological
measurements. These changes are all associated with in- follow-up is having reliable access to the healthcare system,
creased rates of future cardiovascular events, including principally a primary care provider (PCP). However, close to
cardiac ischemia and heart failure. 25% of patients who donate have either never had a PCP or
Pregnancy: Postdonation pregnancy does not carry an
□  visit with a PCP irregularly before donation.58 The rate of fol-
increased risk of maternal or fetal complications. Howev- low-up after donation continues to be low, with only about
er, donors tend to experience higher than expected rates 20% of donors seeking regular care with a PCP; this is par-
of gestational hypertension and preeclampsia, similar to ticularly true among men who have completed less than a
what is expected in women with prepregnancy hyperten- college education and who did not have a regular PCP before
sion, diabetes, and higher BMI. donation.58 The importance of having a PCP is highlighted
110 Kidney Transplantation: Principles and Practice

by the fact that a donor is 14 times more likely to not have perspectives. Most would argue that a donor who has altru-
any follow-up in the first year after transplant if they had no istically undergone nephrectomy for no direct medical ben-
regular PCP before transplant.53 Finally, the demographic efit should incur as few negative consequences (including
risk factors for not having a PCP mirror those risk factors for financial) as possible. Furthermore, when patients undergo
not completing required follow-up and for increased risk of financial hardship, including time away from activities
lower GFR, higher blood pressure and proteinuria, and other of daily living and/or work, there is a significant societal
deleterious outcomes after donation.53,58 This suggests that strain. Finally, financial constraints and concerns may
screening for these factors may be as important as other, contribute to limiting enthusiasm for donation, a concept
more traditional laboratory, physical examination, or medi- cited frequently for the recent stagnation in living donor
cal history elements of a donor’s workup. rates. Pretransplant, more than 9 of 10 donors state that
Because the donor-derived benefits of living donation are they incurred a cost related to their donation workup.
psychological and not medical, specific attention to psy- These costs include travel, lodging, or other health-related
chological outcomes is important. Additionally, donors are costs.60 Whereas the demographic characteristics of donors
required to undergo extensive pretransplant psychological vary across the country, data suggest that a high percent-
screening to understand their motivations for donation, to age of donors (85%) work outside of the home, with approx-
rule out any type of coercion, and to assure that each patient imately 30% of these donors in professional lines of work.
is free from any significant mental health issues that might After donation, about 75% of donors indicate that they
influence the donation decision or complicate postdonation were out of work for 4 weeks, with approximately 20% stat-
recovery. In general, there is a low rate of mental health ing that they were unable to perform their activities of daily
issues among donors compared with the general popula- living for this same time period.61 Impaired function trans-
tion. However, the risk for postdonation depression and lates into costs not commonly considered: 30% of donors
anxiety is real, along with its potential to negatively affect a stated that they spent money on child care or help cover-
donor’s quality of life. Up to 20% of donors report decreased ing required house maintenance, and so on.60 Attempts to
postdonation quality of life; this is particularly true for non- place a financial value on direct and indirect costs is chal-
related, nonspouse donors who experience a medical com- lenging and convoluted. However, after taking a number of
plication or who witness their graft fail in the recipient.52,59 factors into consideration, just less than 40% of donors will
Furthermore, if the donor is ambivalent before transplant spend up to $500 related to their donation in the 12 months
or has a longer recovery (related to or independent of com- after the event, with 20% spending more than $5000 dur-
plications), they have a higher risk of postdonation psycho- ing that same time period.60 A similar effect is noted in other
logical complications, specifically anxiety.59 countries around the world, including Canada.62
Another consideration for donors pretransplant is the
effect of donation on the attainment of postdonation insur-
Financial Considerations ance (health, life, or other). As many as 57% of transplant
centers indicate that a lack of predonation insurance is
Long-term donor outcomes can also be measured finan- an absolute (15%) or relative (42%) contraindication to
cially, from a number of different perspectives, related to donation (Table 7.2; Figs. 7.1 to 7.4).60 Although the rate
both direct and indirect costs. Understanding the financial of insurance among donors in the US is proportional to
impact of donation is critical from both patient and societal the rate of uninsured people in the general population,

TABLE 7.2 Absolute and Relative Contraindications to Living Kidney Donation


Absolute Relative
Age Less than 18 years Over 65 excluded from many programs
Informed consent Impaired ability to make an autonomous decision because of
mental or psychiatric condition
Substance abuse Active substance abuse
Hypertension Multiple agents or high doses of single agents for control Borderline or control with single agents
Evidence of end-organ damage
End-organ injury
Additional strong risk factors for cardiovascular disease
Diabetes Diabetes mellitus Impaired glucose tolerance
Obesity Morbid obesity (BMI >35) with comorbid conditions Obesity (BMI >30) with comorbid conditions
Renal disease Evidence of renal disease with reduced creatinine clearance Borderline creatinine clearance, microscopic hema-
(GFR <80 mL/min), proteinuria (>250 mg), or hematuria turia low levels of proteinuria (<30 mg/24 h)
Renal stones Multiple or recurrent renal calculi of a metabolic condition that Single renal stone
predisposes to the recurrence of renal calculi
Inherited renal disease ADPKD, SLE, Alport’s syndrome, IgA nephropathy TBMD, mutations in APOL1
Infection HIV, hepatitis B, hepatitis C, West Nile virus, Chagas’ disease Hepatitis B core antibody
Cancer Cancer current or treated but at significant risk for recurrence
Cardiovascular disease Coronary or peripheral vascular disease, or heart valve disease
Renal anatomic abnormalities Significant discrepancy in the kidney sizes Vascular anomalies (see Figs. 7.1 to 7.4)

ADPKD, adult polycystic kidney disease; APOL1, apolipoprotein L1; BMI, body mass index; GFR, glomerular filtration rate; HIV, human immunodeficiency virus;
IgA, immunoglobulin A; SLE, systemic lupus erythematosus; TBMD, thin basement membrane disease.
Modified from kidney transplantation, 7th ed., Chapter 7, MacConmara and Newell.
7 • Medical Evaluation of the Living Donor 111

A B
Fig. 7.1 Duplication of the renal collecting system. (A) Image from a computed tomography angiogram demonstrating a left kidney with duplicated
ureters. (B) Magnetic resonance imaging demonstrating duplication of the collecting systems of both the right and left kidneys. Arrows in each panel
identify the duplicated ureters.

A B
Fig. 7.2 Renal venous anomalies—retroaortic left renal vein. (A) Image from a computed tomography angiogram demonstrating a retroaortic left
renal vein (arrow identifies the retroaortic left renal vein). Note the characteristic inferior course of the retroaortic left renal vein relative to the left renal
artery. (B) Reconstructed image clearly demonstrating the left renal vein coursing posterior to the aorta.

there are disparities among black and Hispanic donors. for the donor. However, coverage for postdonation compli-
This is concerning, because these groups of donors are cations is not always uniformly and durably covered. Addi-
more likely to experience hypertension, proteinuria, or tionally, postdonation coverage becomes even more unclear
other outcomes that would require subsequent medical when donors have preexisting health issues, an increasingly
diagnostic and therapeutic attention after donation.60 common problem, because as the acceptable donor age rises,
This is further complicated by the fact that nearly 10% so do potential health issues, such as a history of hyperten-
of donors indicate that they had trouble obtaining health sion, obesity, and kidney stones. Although a great deal of
insurance after donation, either because insurance com- additional data is needed to understand how to maintain kid-
panies would not insure them or because the premium to ney donation as cost neutral, having a clear framework for
do so was too high, even after the Affordable Care Act was understanding what costs a donor may encounter is helpful,
passed in the US.60 including being specific about what outcomes are affected by
Most recipient insurance plans cover the costs of the dona- donation; this allows for more straightforward attribution of
tion evaluation, nephrectomy, and associated hospitalization risk to the act of donating.63
112 Kidney Transplantation: Principles and Practice

A B
Fig. 7.3 Renal venous anomalies—duplication of the inferior vena cava and circumaortic left renal vein. (A) Duplication of the inferior vena cava
(arrow identifies the duplicated, left-sided inferior vena cava and double arrow demonstrates the left-sided inferior vena cava crossing the aorta to join
the main, right-sided inferior vena cava). (B) Circumaortic left renal vein (arrows indicate the component of the left renal vein that cross anterior and
posterior to the aorta and the relationship to the left renal artery).

A B
Fig. 7.4 Anatomic abnormalities of the renal arteries. (A) Left kidney with two renal arteries with a small-caliber lower pole artery arising from the
aorta immediately inferior to the main left renal artery (arrow indicates the position of the two renal arteries). (B) Reconstructed image from a computed
tomography angiogram demonstrating a left renal artery with an early bifurcation (arrow indicates the larger, superior and smaller, inferior branches
of the left renal artery).

A number of factors influence living donor rates in the


Living Donor Quality Initiatives US, including donor and recipient demographics, cultural
beliefs about transplantation, the number of unrelated
Because potential health consequences follow kidney dona- versus related donors available (particularly as the donor/
tion, and because the long-term effect of donation is not recipient relationship affects HLA and blood type compat-
completely known, a comprehensive quality program at ibility), and the availability of the laparoscopic approach to
each transplant center is increasingly important. Public nephrectomy. Many centers track the success of their living
reporting of transplant outcomes includes reporting of liv- donor program based on the percentage of the total num-
ing donor outcomes. Living donor quality programs are ber of transplants performed from living donors, a ratio that
a particular emphasis as donors become more medically hovers around 30% in the US. However, the rate of living
complex. Furthermore, with the growth of paired exchange and deceased donation may not be related; a more reflec-
programs, the logistics of living donation have become tive measure may be the number of living donor transplants
increasingly complicated. Finally, because living donor rates performed (or donors who register for evaluation) as a per-
have plateaued over the last several years, providing quality centage of the number of new waitlist registrants.64 Mea-
data may help in understanding impediments to growth. suring the living donor transplant rate in this way allows
7 • Medical Evaluation of the Living Donor 113

programs to more precisely understand barriers (race, 12. Wang JMG, Montgomery RA, Kucirka LM, et al. Incompatible live-
socioeconomic status, age, HLA, or ABO incompatibility) in donor kidney transplantation in the United States: results of a national
survey. Clin J Am Soc Nephrol 2011;6:2041–6.
the regional donor pool. It also allows a different, and per- 13. Montgomery RA, Locke JE, King KE, et al. ABO incompatible renal
haps more accurate way to compare centers across a region transplantation: a paradigm ready for broad implementation. Trans-
or a country. plantation 2009;87:1246–55.
Whereas more than 90% of donors say they would 14. Tyden G, Kumlien G, Berg UB. ABO-incompatible kidney transplanta-
tion in children. Pediatr Transplant 2011;15:502–4.
donate again, more than 25% say that their quality of life 15. Montgomery JR, Berger JC, Warren DS, et al. Outcomes of ABO-
was affected negatively and 44% indicate that the donation incompatible kidney transplantation in the United States. Transplan-
experience could be improved, citing the time in evaluation tation 2012;93:603–9.
was too long; too many required visits to complete in-person 16. Lee SH, Oh CK, Shin GT, et al. Age matching improves graft sur-
assessments and testing; and the number of postdonation vival after living donor kidney transplantation. Transplant Proc
2014;46:449–53.
encounters was not enough, leading to donors’ perceptions 17. Noppakun K, Cosio FG, Dean PG, et al. Living donor age and kidney
of feeling “lost to follow-up.”55 Toward this end, transplant transplant outcomes. Am J Transplant 2011;11:1279–86.
centers’ quality programs should consider a focus on the 18. Horvat LD, Shariff SZ, Garg AX. Global trends in the rates of living
patient experience, including the duration and number of kidney donation. Kidney Int 2009;75:1088–98.
19. Steering Committee of the Istanbul Summit. Organ trafficking and
visits required to complete a donor workup, and how many transplant tourism and commercialism: the declaration of Istanbul.
postdonation encounters are completed (either associated Lancet 2008;372:5–6.
with or independent of mandated UNOS requirements). 20. Ghods AJ. Living kidney donation: the outcomes for donors. Int J
Quality of life after donation is critical to any definition of Organ Transplant Med 2010;1:63–71.
donor success and there is no doubt that donation can have 21. Ghods AJ, Savaj S. Iranian model of paid and regulated living-unre-
lated kidney donation. Clin J Am Soc Nephrol 2006;1:1136–45.
a dramatic direct and indirect financial impact on a donor.61 22. Salim A, Malinoski D, Schulman D, et al. The combination of an
Transplant centers should track time back to normal activi- online organ and tissue registry with a public education campaign can
ties of daily living, time back to work, and costs incurred by increase the number of organs available for transplantation. J Trauma
donors. Certain groups of donors are particularly important 2010;69:451–4.
23. Goldberg DS, Blumberg E, McCauley M, et al. Improving organ uti-
to understand their course: donors who are female, have lization to help overcome the tragedies of the opioid epidemic. Am J
attained lower education, are related to the recipient, and Transplant 2016;16:2836–41.
have suffered complications at the time of donation are at 24. Port FK, Bragg-Gresham JL, Metzger RA, et al. Donor characteristics
the highest risk for prolonged return to daily activities and associated with reduced graft survival: an approach to expanding the
work.61 Additionally, as centers encourage more medically pool of kidney donors. Transplantation 2002;74:1281–6.
25. Rao PS, Schaubel DE, Guidinger MK, et al. A comprehensive risk
complex and racially diverse donors, attention must be paid quantification score for deceased donor kidneys: the kidney donor risk
to the financial impact of donation. Helping track insurance index. Transplantation 2009;88:231–6.
rates postdonation would help future donors better under- 26. Massie AB, Luo X, Chow EK, et al. Survival benefit of primary deceased
stand posttransplant risks and liabilities. donor transplantation with high-KDPI kidneys. Am J Transplant
2014;14:2310–6.
Finally, in light of the fact that quality programs and ini- 27. Massie AB, Leanza J, Fahmy LM, et al. A risk index for living donor
tiatives, including improved donor follow-up, cost money,56 kidney transplantation. Am J Transplant 2016;16:2077–84.
transplant centers should prospectively track the amount 28. Gentry SE, Segev DL, Montgomery RA. A comparison of popula-
of effort directed toward these initiatives to quantify their tions served by kidney paired donation and list paired donation. Am
effort and expenses to better understand the “value” of the J Transplant 2005;5:1914–21.
29. Henderson ML, Clayville KA, Fisher JS, et al. Social media and organ
donor experience. donation: ethically navigating the next frontier. Am J Transplant
2017;17:2803–9.
References 30. Ferrari P, Weimar W, Johnson RJ, et al. Kidney paired dona-
1. Murray JE. Ronald Lee Herrick memorial: June 15, 1931–December tion: principles, protocols and programs. Nephrol Dial Transplant
27, 2010. Am J Transplant 2011;11:419. 2015;30:1276–85.
2. A definition of irreversible coma. Report of the ad hoc committee of 31. Garonzik-Wang JM, Berger JC, Ros RL, et al. Live donor champion:
the Harvard Medical School to examine the definition of brain death. finding live kidney donors by separating the advocate from the
JAMA 1968;205:337–40. patient. Transplantation 2012;93:1147–50.
3. Ratner LE, Ciseck LJ, Moore RG, et al. Laparoscopic live donor nephrec- 32. Goody AJ. Donor exchange for renal transplantation. N Engl J Med
tomy. Transplantation 1995;60:1047–9. 2004;351:935–7; author reply 935-7.
4. 2015 Annual Data Report. Scientific Registry of Transplant Recipi- 33. Butt FK, Gritsch HA, Schulam P, et al. Asynchronous, out-of-
ents. Available online at: http://srtr.transplant.hrsa.gov/annual_ sequence, transcontinental chain kidney transplantation: a novel
reports/Default.aspx (accessed 02.02.19). concept. Am J Transplant 2009;9:2180–5.
5. Kasiske BL, Snyder JJ, Matas AJ, et al. Preemptive kidney trans- 34. Wongsaroj P, Kahwaji J, Vo A, Jordan SC. Modern approaches to incom-
plantation: the advantage and the advantaged. J Am Soc Nephrol patible kidney transplantation. World J Nephrol 2015;4:354–62.
2002;13:1358–64. 35. Pham TA, Lee JI, Melcher ML. Kidney paired exchange and desensiti-
6. Voelker R. Cost of transplant vs dialysis. JAMA 1999;281:2277. zation: strategies to transplant the difficult to match kidney patients
7. Siedlecki A, Irish W, Brennan DC. Delayed graft function in the kidney with living donors. Transplant Rev (Orlando) 2017;31:29–34.
transplant. Am J Transplant 2011;11:2279–96. 36. Bingaman AW, Wright J, Kapturczak M, et al. Single-center kidney
8. Yarlagadda SG, Coca SG, Formica RN, et al. Association between paired donation: the Methodist San Antonio experience. Am J Trans-
delayed graft function and allograft and patient survival: a systematic plant 2012;12:2125–32.
review and meta-analysis. Nephrol Dial Transplant 2009;24:1039–47. 37. Leesear DB, Aull MJ, Afaneh C, et al. Living donor kidney paired dona-
9. 2012 Annual Data Report. Scientific Registry of Transplant Recipi- tion transplantation: experience as a founding member center of the
ents. Available online at: http://srtr.trasnplant.hrsa.gov/annual_ National Kidney Registry. Clin Transplant 2012;26:E213–22.
reports/Default.aspx (accessed 27.02.19). 38. Melcher ML, Leeser DB, Gritsch HA, et al. Chain transplantation:
10. United Network for Organ Sharing. In: initial experience of a large multicenter program. Am J Transplant
11. Williams RC, Opelz G, Weil EJ, et al. The risk of transplant failure with 2012;12:2429–36.
HLA mismatch in first adult kidney allografts 2: living donors, sum- 39. Irwin FD, Bonagura AF, Crawford SW, Foote M. Kidney paired dona-
mary, guide. Transplant Direct 2017;3:e152. tion: a payer perspective. Am J Transplant 2012;12:1388–91.
114 Kidney Transplantation: Principles and Practice

40. Johnson RJ, Allen JE, Fuggle SV, et al. Early experience of paired 54. Henderson ML, Thomas AG, Shaffer A, et al. The national landscape
living kidney donation in the United Kingdom. Transplantation of living kidney donor follow-up in the United States. Am J Transplant
2008;86(12):1672–7. 2017;17:3131–40.
41. Hume DM, Merrill JP, Miller BF, Thorn GW. Experiences with renal 55. Li T, Dokus MK, Kelly KN, et al. Survey of living organ donors’ expe-
homotransplantation in the human: report of nine cases. J Clin Invest rience and directions for process improvement. Prog Transplant
1955;34:327–82. 2017;27:232–9.
42. Morath C, Zeier M, Döhler B, et al. ABO-incompatible kidney trans- 56. Keshvani N, Feurer ID, Rumbaugh E, et al. Evaluating the impact of
plantation. Front Immunol 2017;8:234. performance improvement initiatives on transplant center reporting
43. Muramatsu M, Gonzalez HD, Cacciola R, et al. ABO incompatible compliance and patient follow-up after living kidney donation. Am J
renal transplants: good or bad? World J Transplant 2014;4:18–29. Transplant 2015;15:2126–35.
44. Smith RA, Manassaram-Baptiste D, Brooks D, et al. Cancer screening 57. Kulkarni S, Thiessen C, Formica RN, et al. The long-term follow-up
in the United States, 2015: a review of current American Cancer Soci- and support for living organ donors: a center-based initiative founded
ety guidelines and current issues in cancer screening. CA Cancer J Clin on developing a community of living donors. Am J Transplant
2015;65:30–54. 2016;16:3385–91.
45. Naik AS, Cibrik DM, Sakhuja A, et al. Temporal trends, center-level 58. Alejo JL, Luo X, Massie AB, et al. Patterns of primary care utilization
variation, and the impact of prevalent state obesity rates on accep- before and after living kidney donation. Clin Transplant 2017;31.
tance of obese living kidney donors. Am J Transplant 2018;18:642–9. 59. Jacobs CL, Gross CR, Messersmith EE, et al. Emotional and financial
46. Rege A, Leraas H, Vikraman D, et al. Could the use of an enhanced experiences of kidney donors over the past 50 years: the RELIVE
recovery protocol in laparoscopic donor nephrectomy be an incentive Study. Clin J Am Soc Nephrol 2015;10:2221–31.
for live kidney donation? Cureus 2016;8:e889. 60. Rodrigue JR, Schold JD, Morrissey P, et al. Direct and indirect costs
47. Grams ME, Sang Y, Levey AS, et al. Kidney-failure risk projection for following living kidney donation: findings from the KDOC Study. Am J
the living kidney-donor candidate. N Engl J Med 2016;374:411–21. Transplant 2016;16:869–76.
48. Lentine KL, Segev DL. Understanding and communicating medi- 61. Larson DB, Jacobs C, Berglund D, et al. Return to normal activities and
cal risks for living kidney donors: a matter of perspective. J Am Soc work after living donor laparoscopic nephrectomy. Clin Transplant
Nephrol 2017;28:12–24. 2017;31:e12862.
49. Mjøen G, Hallan S, Hartmann A, et al. Long-term risks for kidney 62. Klarenbach S, Gill JS, Knoll G, et al. Economic consequences incurred
donors. Kidney Int 2014;86:162–7. by living kidney donors: a Canadian multi-center prospective study.
50. Muzaale AD, Massie AB, Wang MC, et al. Risk of end-stage renal dis- Am J Transplant 2014;14:916–22.
ease following live kidney donation. JAMA 2014;311:579–86. 63. Gill JS, Delmonico F, Klarenbach S, Capron AM. Providing cover-
51. Ibrahim HN, Foley RN, Reule SA, et al. Renal function profile in white age for the unique lifelong health care needs of living kidney donors
kidney donors: the first 4 decades. J Am Soc Nephrol 2016;27:2885–93. within the framework of financial neutrality. Am J Transplant
52. Matas AJ, Hays RE, Ibrahim HN. Long-term non-end-stage renal 2017;17:1176–81.
disease risks after living kidney donation. Am J Transplant 64. Matar AJ, Files J, Burkholder R, et al. Evaluating living donor kidney
2017;17:893–900. transplant rates: are you reaching your potential? Clin Transplant
53. Schold JD, Buccini LD, Rodrigue JR, et al. Critical factors associated 2017;31:e12914.
with missing follow-up data for living kidney donors in the United
States. Am J Transplant 2015;15:2394–403.
8 Donor Nephrectomy
JOHN C. LAMATTINA and ROLF N. BARTH

CHAPTER OUTLINE Deceased Donor Nephrectomy Total Laparoscopic Approach


Donation After Brain Death Right Donor Nephrectomy
Donation After Circulatory Death Single-Port Donor Nephrectomy
Living Donor Nephrectomy Robotic Donor Nephrectomy
Anesthetic Management Complications
Open Donor Nephrectomy Summary
Laparoscopic Donor Nephrectomy
Hand-Assisted Technique

Deceased Donor Nephrectomy the maximal benefit in terms of life years. In essence, donor
kidneys are evaluated for quality using the Kidney Donor
Deceased donor renal donation predominates as the source Risk Index (KDRI; see Fig.8.2).3 Donor-specific factors from
of transplantable kidneys. In the US, deceased donors pro- the KDRI (including age, ethnicity, height, weight, his-
vide approximately 13,000 kidneys per year or 70% of the tory of hypertension or diabetes, hepatitis C status, and
available pool of transplantable kidneys (Fig. 8.1).1 donation after cardiac death status) are used to create a
Recent years have seen an increase in the number of Kidney Donor Profile Index that assigns a score between 0
deceased donors secondary to the opioid crisis in North and 100, setting the median donor as the 50th percentile.
American (and Europe) and the resultant number of refer- A kidney with a lower perceived risk (and likely of higher
rals for organ donation. Despite assumptions that these quality) scores lower. Recipients are likewise risk stratified
donors would be predominantly younger and without for expected posttransplant survival (EPTS) using age, his-
comorbidities, data support growth in all age categories and tory of diabetes, duration of dialysis, and history of prior
this growth in donor population has been associated with transplant. The lowest 20th percentile kidneys (which are
extensive medical and substance abuse histories.2 Anoxic expected to have the longest graft survival) are allocated
brain injury is the primary mechanism of terminal injury preferentially to recipients in the highest 20th percentile
in these donors and this can be accompanied by significant of expected survival (who are expected to have the longest
hypotensive periods before resuscitation. Such an insult patient survival). The new system also offers priority allo-
can frequently manifest itself as acute kidney injury at the cation to highly sensitized recipients, with patients having
time of donation. a calculated panel reactive antibody (cPRA) ≥99 receiving
Deceased donors are divided into subgroups of dona- the greatest benefit, but also increasing access for patients
tion after brain death (DBD) and donation after circula- with a cPRA ≥98%. Zero-antigen mismatch donors will still
tory death (DCD). DBD donors are pronounced dead by be offered priority under the new system. Recipients are still
both clinical and radiologic evaluations that are clearly offered points based on years on the waiting list (or main-
defined. The operations from these donors are conducted in tenance dialysis), prior living donation, degree of sensitiza-
controlled settings with careful physiologic monitoring to tion, and one degree of HLA mismatch).4
ensure optimal organ perfusion and oxygenation until per-
fusion and cooling of donor organs. DBD donors account
for the overwhelming majority of deceased donors repre- Donation After Brain Death
senting 82.3% of donors in 2015. DCD donation rates have
increased over the past decade, and the proportion of DCD Kidneys from DBD donors are usually recovered in conjunc-
donors contributing to the donor pool more than doubled tion with recovery of other abdominal and thoracic organs
from 7.3% to 17.7% between 2005 and 2015 (Fig. 8.2).1 and require coordination of the surgical teams performing
Other countries have higher rates of DCD donation, for different roles. Often, thoracic teams will have complicated
example, in the UK in 2017 to 2018, 40% of all deceased recipients with redo-transplantation, mechanical support
donors were DCD (https://nhsbtdbe.blob.core.windows- devices, or other complicated histories, and this can result
.net/umbraco-assets-corp/11628/transplant-activity- in significant delays until cross-clamp for abdominal recov-
report-2017-2018.pdf). ery teams.
The US introduced a new deceased donor renal trans- Initial midline laparotomy, and isolation and control of
plant allocation system in 2013. This system was designed the infrarenal abdominal aorta is accomplished. In cases
to match donor kidneys with recipients who would receive where the liver is recovered, isolation of either the inferior

115
116 Kidney Transplantation: Principles and Practice

Deceased
15000
donor
Living donor

Transplants
All

5000

0
2004 2006 2008 2010 2012 2014 2016
Year
Fig. 8.1 Transplanted kidneys in US from 2004 to 2015 by donor type. Recent years have observed increased numbers of deceased donors. (OPTN/
SRTR 2015 Annual Data Report. HHS/HRSA.)

50
Percentage of donors

40
Donor age >50
Black race
30 Diabetes
Hypertension
20 Weight >80 kg
Terminal SCr >1.5 mg/dL
DCD
10 CVA death

0
2004 2008 2012 2016
Year
Fig. 8.2 Components of the Kidney Donor Risk Index 2004 to 2015. Significant increases in the percent of DCD donors has been observed over the
past decade. These components have resulted in no net changes in overall KDRI since 2004. (OPTN/SRTR 2015 Annual Data Report. HHS/HRSA.)

mesenteric vein or portal vein can also be achieved. The superior cuff that can be used to construct a venous exten-
aorta is cannulated after administering heparin, and in sion when necessary.
appropriate cases venous cannulation is performed. In Individual recovery starts with either side by isolating
coordination with thoracic recovery, perfusion is initiated, the ureter and gonadal vein and transecting distally. Care
the aorta is clamped in a supraceliac location, and either should be taken to leave tissue around the ureter with
the inferior vena cava or the right atrium are transected sharp dissection to minimize the risk of devascularizing the
and suction or drainage devices are placed to facilitate ureter by stripping it of adjacent tissues. The anterior wall
perfusion. The abdominal organs are packed with ice for of the aorta can be longitudinally sharply transected, fol-
cooling while flushing and recovery of other organs are lowed by division of the posterior wall, with care taken to
performed. Mobilization of the ascending and descending identify single or multiple renal arteries and leave sufficient
colon can be performed to allow for more direct exposure tissue for Carrel patches around each vessel. From an infe-
of the kidneys to ice. Recovery of thoracic organs, liver, rior approach, working from the midline and posterior to
and pancreas generally precedes recovery of the kidneys the aorta, all tissues can be sharply divided with attention
(Fig. 8.3). to the location of the ureter to avoid inadvertent injury.
Recovery of the kidneys can be performed either individ- Working both superiorly and laterally, the vasculature and
ually or en bloc. Individual recovery of the kidneys is per- kidneys can be separated from the lateral abdominal and
formed by transecting the left renal vein at the vena cava. retroperitoneal attachments. Posterior dissection proceed-
The aorta and vena cava can both be transected superior ing directly against the psoas muscle will minimize the risk
to the level of cannulation (usually just superior to bifur- of injuring renal arteries. Regardless of extent, Gerota’s
cation) and at the origin of the superior mesenteric artery fascia should be removed with the kidneys to be separated
(SMA) and superior to the right renal vein. Division of the at later time. The right kidney should be removed with all
aorta should be performed by incising the base of the SMA remaining vena cava to preserve the conduit for venous
and, with an oblique angle, entering the aorta superiorly extension grafts when necessary.
to identify renal arteries because they often enter at this En bloc recovery of the kidneys is performed without lon-
level or higher. Superior transaction of the aorta should gitudinal transection of the aorta or division of the renal
be performed ideally to preserve a Carrel patch on both the vein. Inferior to superior dissection is performed posterior
right and left renal arteries. The right renal vein should be to the aorta and vena cava, and with initial isolation of the
identified before transaction of the vena cava to preserve a ureters to avoid injury. Separation of the kidneys is then
A

Incised Portal vein


gallbladder Aorta
Left gastric artery
Transected Celiac axis
common bile duct Splenic artery
Right gastric artery Hepatic artery
Catheter in
Inferior vena cava splenic vein

Gastroduodenal
artery
Superior mesenteric
artery and vein
B

R. gastric artery
Hepatic artery Bowel,
duodenum,
and pancreas
Splenic artery and vein retracted

Short gastric arteries


L. gastric artery
Loose ligament
Portal vein around retracted
Aorta superior
mesenteric artery

Superior mesenteric
artery and vein
C D

Celiac artery
Superior mesenteric
artery
L. kidney
Kidney perfusion

Aorta
Ureter Vena cava
R. kidney Inferior mesenteric
Perfusion artery Lumbar vessels
catheters Aorta
IVC

Ureter

E F
Fig. 8.3 Deceased donor organ retrieval. (A) The chest and abdomen are opened through a midline incision. The abdominal cavity is inspected for
any evidence of disease or injury. Initial control of the distal aorta is obtained in the case that urgent flushing becomes necessary. (B) The splenic vein is
catheterized through the inferior mesenteric vein for portal perfusion. Limited portal dissection should include distal ligation of the common bile duct
and incision and flushing of the gallbladder. (C) Pancreas dissection can be performed by division of the short gastric vessels and medial visceral rota-
tion of the spleen and pancreas in a plane posterior to the splenic vein and artery. (D) The duodenum should be widely mobilized allowing for access
to the superior mesenteric artery and infrahepatic vena cava. (E) The distal aorta is cannulated with a perfusion catheter after heparinization, and drain-
age devices may be placed in the distal vena cava. (F) Aortic cross-clamp is performed in the supraceliac location, and perfusion and cooling with ice is
performed. After removal of thoracic organs, liver, and pancreas, kidney dissection is commenced. Additional attention should be directed to preserving
ureteral length and aortic cuff around the origin of single or multiple renal arteries. Kidneys can be recovered individually or en bloc.
118 Kidney Transplantation: Principles and Practice

performed after removal with the similar goals of leaving an explant of the organs from the abdominal cavity, and facil-
aortic cuff for all renal arteries and the vena cava with the itate organ cooling may improve renal outcomes as has
right kidney. been reported with other organs.6
Local procurement centers have preferences for marking While infusion of the preservation solution is underway,
the ureters with ligature or other labeling to identify right surgical recovery of the kidneys can be performed with
versus left kidneys. a similar technique to recovery in DBD donors. Whereas
If concerns exist for the quality of perfusion based on an expedient technique is important, precision remains
the appearance of the kidneys, direct cannulation and paramount in these circumstances to prevent the higher
perfusion of the right and left renal arteries can be per- rates of surgical damage and organ discard that have been
formed on the back table. Although this step is not usu- reported.7 After removal of the kidneys from the abdomi-
ally necessary, concern regarding poor perfusion or the nal cavity, additional perfusion can be performed once the
mottled appearance of the kidneys should direct addi- kidneys are in cold solution depending on either preference
tional flushing. or concerns regarding the quality of intraabdominal per-
fusion. Modified technical approaches including balloon
catheter placement for in situ preservation or use of extra-
Donation After Circulatory Death corporeal support after death have not achieved substan-
tial effect in improving results.5,6 Although supportive data
Whereas in total numbers DBD donors account for the exist regarding the ability of hypothermic pulsatile perfu-
vast majority of recovered and transplanted kidneys, DCD sion to improve outcomes for DCD kidneys, conflicting data
has become an increasingly common source of deceased still exist and thus this conclusion is not uniform.8,9
donor kidneys in recent years. In the US in 2015, DCD Additional interest in normothermic perfusion devices
donors provided more than 2000 donor kidneys (nearly has been generated in recent years. Although no defini-
20% of the total deceased donation). DCD donors yielded tive conclusions can be reached, potential for improved
more kidneys per donor (1.56) than brain dead donors function and decreased rates of DGF are under investiga-
(1.44).1 tion.10 Although a variety of strategies have been proposed,
Recovery of kidneys from DCD donors is performed in a including initiating normothermic perfusion in situ, ex vivo
similar manner to brain dead donors with a few modifica- at the donor hospital, or on return to the recipient center, a
tions. Individual hospitals and organ procurement orga- single system that has reliably improved outcomes has yet
nizations (OPOs) set specific guidelines for time limits for to be reported.
recovery to be performed after withdrawal of life support
that vary between 60 and 120 minutes. These waiting peri-
ods occur either in a perioperative setting or in the operat- Living Donor Nephrectomy
ing room; outside the US, this time may extend to 180 to
240 minutes. According to local practice, patients are Living renal donation provides an invaluable resource in
declared deceased after cessation of pulse, cardiac rhythm, regard to both organ quantity and quality. There are 5600
or electrical activity. The cessation of all pulseless electri- living kidney donations annually in the US, which accounts
cal activity (PEA) has not been deemed necessary as a cri- for approximately 30% of annual US renal transplant vol-
terion for declaration in the absence of pulse pressure.5 An ume. The absolute number of living donors nearly equals
additional waiting period between 2 and 5 minutes occurs the number of deceased donors and thus remains critical as
before initiation of organ recovery. a source of transplantable organs. Living donor kidneys are
The period of warm ischemic time between withdrawal of superior quality in every objective measurement includ-
of life support and the initiation of cooling and perfu- ing immediate graft function rates, graft half-life, and life
sion with organ preservation solutions contributes to the years gained for recipients. The available pool of donors has
increased rates of delayed graft function and organ dys- remained relatively stable over the recent decade, although
function seen with DCD organs. Thus the surgical proce- demographics have demonstrated increases in donors over
dure needs to be performed in an expedited fashion to cool 50 years old (29.5%) and women (63.5%; Fig. 8.4). The pri-
and perfuse organs as soon as possible. A midline incision mary responsibility of the donor surgeon is patient safety,
from sternal notch to pubis is rapidly performed and, using and this overarching concern must guide every pre-, intra-,
combinations of sharp and blunt dissection, the abdomi- and postoperative decision. The reliably safe outcomes with
nal cavity is entered and the distal aorta is cannulated donor nephrectomy and good long-term renal function
with a perfusion catheter with or without isolated con- of donors are paramount to preserve the justification for
trol. Immediate perfusion should commence at this point. removing a kidney from a healthy donor.
Transection of the distal inferior vena cava and placement As the transplant community has continued to gain
of drainage device or suction catheter can be performed to experience caring for living donors, conditions that had
facilitate perfusion. Depending on recovery of the liver or previously served as absolute or relative contraindications
thoracic organs, the aorta can be clamped at the level of to living donation are being reconsidered. Select centers
the descending thoracic aorta after opening the chest. DCD now accept donors with prior surgical histories that affect
recoveries for kidneys alone can avoid opening the chest, the technical complexity of the operation. These include
and clamping of the aorta should occur at the supraceliac prior histories of bariatric procedures, gynecologic opera-
level. The abdominal cavity should then be packed with tions, hernia repair, appendectomy, and cholecystectomy.
ice during perfusion. A rapid surgical technique designed Although not contraindications for surgery, these proce-
to minimize the time taken to achieve cross-clamp and dures may predict a more complicated technical operation.
8 • Donor Nephrectomy 119

65

60

Percentage
55

50 Male
Female
45

40

35
2004 2006 2008 2010 2012 2014 2016
Year
Fig. 8.4 Living kidney donors by sex. Increasing rates of female donors has resulted in approximately 2:1 ratio of female:male donors. (OPTN/SRTR
2015 Annual Data Report. HHS/HRSA.)

Anatomic variants that once precluded donation, such as Regardless of the technical approach, control of postop-
multiple renal arteries and circum- or retroaortic renal erative pain should be initiated during the operative case.
veins, no longer eliminate potential donors at many cen- Intraoperative administration of narcotics provides tran-
ters. These more complicated patients may be eligible for sient pain control. The use of local anesthetics and systemic
renal donation, and consideration should be given for refer- nonsteroidal agents can minimize postoperative pain and
ral to centers with experience in these conditions. narcotic requirements. We routinely inject 0.5% liposomal
bupivacaine into port and extraction sites and consider the
use of intravenous ketorolac for most patients. The use of
Anesthetic Management liposomal bupivacaine preparations has added to the dura-
tion of local anesthesia for up to 72 hours and we have
Communication with anesthesia personnel is important to found this useful to facilitate early postoperative pain con-
ensure that good urine output is achieved throughout the trol and discharge. Additionally, initiating regularly sched-
case. Pneumoperitoneum has been demonstrated to impair uled oral narcotics soon after surgery can prevent intense
venous return influencing renal perfusion, and volume pain spikes as local agents diminish.
expansion has been demonstrated as the primary inter-
vention to counteract this effect.11 Patients often require
greater than 5 L of crystalloid to achieve a robust urine out- Open Donor Nephrectomy
put. Mannitol can be administered in divided doses of 12.5
g to augment urine output. Urine output should be moni- Open donor nephrectomy has been nearly completely
tored and low output addressed aggressively by admin- replaced by minimally invasive surgical techniques. In fact,
istering additional intravenous fluids and decreasing or surgeons trained in recent eras may have little or no expe-
eliminating pneumoperitoneum until adequate urine out- rience with standard open or miniopen techniques. None-
put is achieved. theless, these techniques may be employed by select centers
Whereas inadequate volume resuscitation is the most and/or surgeons based on indication or preference. Rela-
likely factor, identifying other confounding factors for low tive indications may include the presence of complicated
urine output, including relative hypotension, inability to vascular anatomy, prior operations that complicate lapa-
tolerate pneumoperitoneum, or other physiologic events roscopic approaches, or right nephrectomy. Whereas these
are important to determine whether the case should pro- techniques deserve an appropriate place in the arsenal of
ceed. Although extremely unusual, our practice is not to living donor nephrectomy, laparoscopic techniques can be
proceed with donation if adequate urine output cannot be successfully used in almost all cases. Despite reduced inva-
achieved. siveness of miniopen incisions, laparoscopic techniques
Adequate relaxation is necessary to achieve sufficient still result in comparatively decreased pain, faster return to
pneumoperitoneum to provide abdominal domain to per- work, and higher patient satisfaction.12,13
form surgery. Diminishing abdominal domain will result Standard open techniques depend on the division of
from patients that are inadequately paralyzed and will result muscle and possible rib resection compared with min-
in difficulty making surgical progress. This may be real- iopen approaches that are muscle-sparing and avoid rib
ized at midpoints of the case as initial paralytic agents may resection. The miniopen techniques have been reported to
require redosing. Participation by experienced anesthesi- improve donor outcomes compared with standard open
ologists is important throughout the procedure, but espe- techniques.14 After the induction of general anesthesia,
cially immediately before division of vascular structures. patients are positioned in a flexed lateral decubitus orien-
We do not routinely administer heparin before division of tation on the operative table. The patient is prepped and
the renal vessels and have not observed complications from draped from the inferior rib margin to the superior iliac
this practice. Some surgeons administer low-dose intrave- crest. A lateral oblique incision is performed inferior to
nous heparin (3000 units) before vascular division with the 12th rib with division or separation of the oblique and
reversal by administering protamine after removal of the transverse musculature. Segmental resection of the infe-
kidney. rior rib may be necessary to improve exposure of the upper
120 Kidney Transplantation: Principles and Practice

pole of the kidney. Combinations of manual and electro- Laparoscopic Donor Nephrectomy
cautery dissection are performed around Gerota’s fascia to
permit retractor placement. The peritoneal cavity is swept Initial reports were made of a laparoscopic approach to
anteromedially as planes are created to the level of the nephrectomy for tumor with morcellation and extraction
renal vein and artery. Retractors can be placed either on in 1991.15 In 1995 this approach had been successfully
fixated platforms or by handheld techniques. Retroperito- applied to living donor nephrectomy as Ratner made the
neal dissection continues around the kidney and inferiorly first report of laparoscopic nephrectomy for transplantation
to identify and isolate the ureter and gonadal vessels. The with immediate graft function.16 Initial comparisons of open
ureter should be dissected close to the level of the iliac ves- and laparoscopic approaches reported substantial improve-
sels to ensure adequate length. Complete mobilization of ments in donor recovery.17 These donor benefits were con-
the kidney is performed, and the artery and vein are iso- firmed in subsequent studies.18,19 A recent randomized
lated at their origin and insertion in the aorta and vena controlled trial demonstrated improved donor satisfaction,
cava, respectively. The adrenal gland can be separated less morbidity, and equivalent graft outcomes.20 Early large
from the parenchyma of the kidney lateral to medial. The series reported concerns regarding complications, espe-
adrenal vein on the left side may be divided between liga- cially with regard to the ureter, associated with the laparo-
tures or clips to maximize renal vein length. Lumbar veins scopic technique. These decreased as progressive technical
posterior to the renal vein should be divided to also maxi- experience was achieved.21 The improved patient recovery
mize renal vein length. This can be performed between and minimally invasive approach has permitted discharge
vascular clamps, surgical clips, or stapling devices. After for select patients on the first postoperative day.22 Addition-
complete isolation of the vascular pedicle, division of the ally, the advent of laparoscopic donor nephrectomy was
ureter and renal vessels proceeds. The ureter and gonadal associated with increased living donation rates and overall
vein are divided distally with ligatures, clips, or stapling volumes, providing important recipient benefits.23
devices. The renal artery or arteries are then divided. This In the US in 2015, 97% of living donor nephrectomies
can be performed by ligation with or without suture or sta- were performed by a laparoscopic approach, with a major-
pling device. Finally, the renal vein can be divided with a ity performed with a hand-assisted approach (Fig. 8.5). The
similar technique. Vascular clamps can be used to maxi- number of cases performed via an open approach has con-
mize vessel length with subsequent ligation and sutur- tinued to decrease over the past 5 years with 3% of donor
ing of vessels after removal of the kidney. The presence nephrectomies performed via an open retroperitoneal or
of multiple vessels requires planning for the order of divi- transabdominal approach.1
sion. Placement of multiple vascular clamps may prove
difficult with limited space, thus stapling devices may be
HAND-ASSISTED TECHNIQUE
preferred. Transfixing techniques with either sutures or
staples should be used for the renal artery and vein stumps Hand-assisted techniques are the preferred approach for
to minimize bleeding risk. many groups and allow for combinations of manual dis-
Inspection for good hemostasis with or without place- section and retraction with laparoscopic visualization and
ment of hemostatic adjuncts is then performed. Abdomi- energy devices. Most series report improved speed with
nal wall closure is performed in multiple layers and with hand-assisted techniques compared with other laparoscopic
preference for absorbable suture. Local anesthetic can be approaches.24 The intraabdominal presence of a hand may
injected to provide local pain control and minimize sys- also be perceived as an improved safety benefit for manual
temic requirements. Similarly, intravenous nonsteroidal control of bleeding or other injury that may occur. Whereas
antiinflammatory drugs may be used to provide pain relief large series do not specifically support these concepts, most
and minimize narcotic requirements. These should be dis- surgeons are comfortable with open techniques providing
continued within 48 hours. Postoperatively, patients can direct manual control. The hand port site is also gener-
receive intravenous and oral narcotics and can return to ally used as the extraction site and does not add morbid-
normal diet and activity. ity, although its location is generally in an upper midline

80

60
Percentage

Transabdominal
40 Flank (retroperitoneal)
Laparoscopic
not assisted
20 Laparoscopic
hand assisted
0 Unknown

2004 2008 2012 2016


Year
Fig. 8.5 Living kidney donation by procedure type. Laparoscopic approaches dominate technical procedure type with the majority performed hand
assisted. (OPTN/SRTR 2015 Annual Data Report. HHS/HRSA.)
8 • Donor Nephrectomy 121

location and is thus more visible than alternate extraction to a level immediately superior to the iliac vessels to provide
sites. The technical steps for recovery are similar to the total adequate recipient length. Proximal to the kidney, elevation
laparoscopic approach (discussed next) with the obvious of the ureter and gonadal vein is performed as lymphatics
addition of a hand to assist. and vessels are identified and divided. We generally do not
divide the gonadal vein; however, in certain cases this may
provide additional exposure to the renal artery and vein.
TOTAL LAPAROSCOPIC APPROACH
Almost all donors have lumbar veins that can vary
Total laparoscopic approach for donor nephrectomy is per- from small and singular to multiple and large. Preopera-
formed with the donor in either a total or modified lateral tive imaging will identify larger lumbar vessels but may
position with flexion of the operative table to open the space be less effective in identifying multiple branches. Addition-
between the iliac crest and the ribs (Fig. 8.6). After sterile ally, aberrant venous anatomy including multiple renal
preparation and draping of the abdomen, a Veress needle
can be placed in the left lower quadrant and used to insuf-
flate the abdomen to 15 mmHg pressure. Our preference
is to use camera visualization through the first port that is
placed with subsequent direct visualization of each addi-
tional port. A combination of 5-mm and 12-mm ports are
placed in periumbilical, superior and inferior midabdomi-
nal, and lateral locations. Placement of a 12-mm port in the
periumbilical location allows for interchange of dissecting
and stapling devices.
The left colon is mobilized along the line of Toldt by divid-
ing this line with the harmonic scalpel or other energy
device (Fig. 8.7). The colon and mesentery should be swept
medially with care not to cause a mesenteric defect or
injure mesenteric vessels. If a mesenteric defect is identified
this should be repaired with suture or clips to avoid poten-
tial for internal hernia formation. After the mesentery has
been swept medially off the psoas and kidney, the ureter and
gonadal vein are identified. The ureter can be identified just
superior to the iliac vessels or near the inferior pole of the
kidney. Care should be taken not to directly grasp the ureter
or use energy devices in direct proximity given the potential
for unrecognized injury. A plane should be created under
the ureter and gonadal vein that can then be elevated while
remaining tissues are dissected (Fig. 8.8). The ureter and
gonadal vein are elevated as a bundle, and a lateral window
is created with an energy device. Our preference is to use an
energy device because small vessels can be present in some Fig. 8.7 Mobilization of colon. The line of Toldt and splenocolic liga-
of these tissue planes. Dissection should be carried distally ment are divided with an energy device (i.e., harmonic scalpel) and the
colon and mesentery are swept medially. Care must be taken to avoid
or identify a mesenteric defect during medial mobilization of the colon
and mesentery.

Fig. 8.6 Positioning and incision placement for laparoscopic left


donor nephrectomy. The donor is positioned in a right lateral decubi- Fig. 8.8 Elevation and dissection of the ureter and gonadal vein. A
tus position with flexion opening the space between costal margin and plane is created under the ureter and gonadal vein. Anterior elevation
iliac crest. A 12-mm port is placed periumbilically and options for 5- to of these structures allows for dissection from a distal level near the iliac
12-mm ports in superior and inferior midclavicular and lateral midaxil- vessels to a proximal location of the renal hilum. Attention to the pres-
lary positions. Extraction can be performed through a transverse Pfan- ence of additional renal arteries and lumbar vessels is necessary as dis-
nenstiel incision. section proceeds superior to the hilum.
122 Kidney Transplantation: Principles and Practice

developed, the adrenal gland requires separation from the


upper pole of the kidney. The adrenal gland is often identifi-
able and can be gently positioned with a grasper as dissec-
tion is performed immediately lateral to the gland. In obese
donors, it may be difficult to identify the adrenal gland,
and dissection along separate tissue planes that belong
either medially with the adrenal gland or laterally with
Gerota’s fascia is performed in potentially ambiguous ter-
ritory. Upper pole vessels often occupy the space between
the adrenal gland and kidney; therefore closer proximity
to the adrenal gland and away from the renal hilum is pre-
ferred. Dissection of the upper pole of the kidney should be
carried posteriorly to the level of the psoas muscle and supe-
riorly to the diaphragm. We perform separation of Gerota’s
Fig. 8.9 Division of the adrenal and lumbar veins. As the kidney fascia from the diaphragm and psoas with energy devices
is elevated with instruments dissection of the lumbar vein from sur- to minimize even small amounts of venous bleeding. Care
rounding lymphatics can be performed. Smaller adrenal and lumbar
veins can be divided with energy devices; larger vessels should either
needs to be taken with dissection around the diaphragm to
be clipped and divided or divided with a stapling device. Consideration not inadvertently injure or perforate it with resultant pneu-
of likely division sites of the renal artery and vein should be performed mothorax. Unrecognized injuries manifest as progressive
to avoid interference of clips with stapling devices. billowing of the diaphragm into the operative field. These
injuries can be repaired laparoscopically by suturing the
identified defect with reduced pneumoperitoneum and Val-
veins, circumaortic, and or retroaortic renal veins may also salva maneuvers.
be associated with variants in lumbar venous anatomy. The renal and adrenal veins are apparent at various
Smaller lumbar veins can be divided with energy devices. stages during the procedure. The adrenal vein is usually
Larger lumbar vessels (>6 mm) should be divided with divided from the renal vein to provide additional venous
either clip appliers or stapling devices (Fig. 8.9). Judgment length on the left kidney. The adrenal vein should be com-
as to the possible interference of metallic clips with subse- pletely isolated from the renal vein and with clear poste-
quent stapling devices needed to divide renal artery and rior planes as the renal artery and aorta are in immediate
vein is important, because clips can interfere with proper proximity. The adrenal vein can be divided with either
closing and stapling. Vascular staples, in contrast, do not energy devices or clip appliers. Care with clip placement
generally interfere with the ability to place stapling devices is important to not interfere with stapling devices neces-
over or in close proximity. sary for division of the renal vein. After division of the
Elevation of the kidney from the lower pole can then adrenal vein, the renal artery is easier to identify and can
reveal the renal artery and vein. Lymphatics are pres- be dissected from the periaortic lymphatics. Tissues can be
ent in varying amounts and density around the vessels, swept medially off the anterior surface of the renal vein to
and require dissection, isolation, and division. Division of provide maximal length. Elevation of the renal vein with
the periaortic lymphatics demands precision with energy a blunt instrument and clearance of a posterior plane can
devices because the risk of injury and bleeding exists if also be completed.
these devices come into contact with blood vessels. The We perform separation of the kidney from its poste-
artery should be completely isolated from the renal vein rior retroperitoneal attachments at varying times during
by careful dissection and should be exposed as proximal to individual cases, depending on our ability to visualize key
the aorta as is safely possible. This is of increased impor- structures. As a general rule, if the kidney is mobilized too
tance in early branching arteries that may require more early in the case, the kidney can inadvertently rotate medi-
extensive dissection down to the level of the aorta. Mul- ally and complicate vascular dissection. We typically leave
tiple arteries are present in approximately one-quarter of Gerota’s fascia intact with the kidney as we perform this ret-
donors. Preoperative knowledge of the location of each roperitoneal dissection. Separation of the kidney from the
additional vessel is important to anticipate as dissection is Gerota’s fascia, even when extensive, can be difficult with
performed. Arteries that originate significantly inferior to densely adherent fat that may risk capsular injury to the
the main renal artery require additional care in elevation kidney. As the kidney is completely mobilized medially, the
of the ureter, gonadal vein, and kidney, because traction psoas muscle and origin of the renal artery become appar-
injuries become increasingly possible. Likewise, avoiding ent. Dissection of the posterior and superior aspects of the
overdissection of smaller, more inferior arteries can pre- renal artery can sometimes be facilitated with the kidney in
vent inadvertent injury. a medial location. Likewise, lumbar vessels may sometimes
The upper pole of the kidney is separated from the reno- be easier to identify and or divide with the kidney medially
colic and splenorenal ligaments using a combination of rotated.
blunt dissection and energy devices. The spleen can be Extraction can be performed via either a Pfannenstiel or
retracted medially in combination with lateral retraction lower midline incision. Through either approach, the rec-
of the kidney to open this space. Retraction of the spleen tus is exposed and a 15-mm port is inserted to accommo-
and surrounding tissues can also be a potentially hazard- date a large endocatch bag for retrieval. Alternatively, the
ous maneuver and should be done with blunt instruments endocatch bag can be directly placed through a small defect
or graspers to avoid injury to the spleen. As this space is in the peritoneum. Care needs to be taken not to have an
8 • Donor Nephrectomy 123

Fig. 8.10 Division of the renal artery. The kidney can be retracted
laterally and anteriorly with instruments as a vascular stapling device is
placed just beyond the origin of the renal artery. Cutting or noncutting
staplers can be used, but attention to the distal location of the stapler Fig. 8.11 Placement of the kidney into endocatch bag. After divi-
is important to avoid clips or other improper positioning. sion of the ureter and vascular structures, the kidney and entire ure-
ter should be placed into an endocatch bag under direct visualization.
Once all structures are inside the bag, it can be closed and extracted
through the selected incision site.
uncontrolled violation of the peritoneal cavity, otherwise
pneumoperitoneum will not be maintained during vascular
stapling. may offer advantages when preserving early bifurcations
Division of the ureter and gonadal vein is performed first because of a decreased width of the device. Additional, non-
with a vascular staple load. This is performed distally with cutting devices allow for confirmation of the correct place-
direct observation of the distal stapler to confirm that the ment of staples before subsequently dividing the vessel with
iliac vessels are not in proximity. The renal artery is then endoscopic scissors. Plastic or metallic clips should not be
divided with the kidney elevated to maximal height. If mul- used to ligate main renal vessels and the US Food and Drug
tiple arteries exist and are separated by more than 5 to 10 Administration (FDA) issued a specific alert in 2011 that
mm, we divide the inferior vessel first followed by new staple updated 2006 warnings regarding the use of Weck Hem-
loads for superior arteries. If arteries are in close proximity o-Lok Ligating Clip for renal artery ligation in living kidney
they can be taken with a single staple load. After arteries are donors secondary to risk of postoperative dislodgement and
divided, the kidney is retracted to a maximally lateral extent hemorrhage.26 Furthermore, it should be noted that trans-
and a stapler is placed on the renal vein as close to the vena fixion of tissue is the only acceptable method for renal artery
cava as is safely possible. division in living donor nephrectomy.
Stapling requires care and attention to the stapling After all vessels have been transected, the kidney should
device and staple loads. An experienced scrub nurse or be confirmed to be free of all retroperitoneal attachments.
technician should be familiar with expedient reloading of Residual attachments can be divided with energy devices
the stapler. Any concern regarding the function or proper or additional staple loads if necessary. The endocatch bag
reloading of the device justifies replacement with a new is directly deployed under the kidney and the kidney and
stapling device that should be immediately available in the ureter are placed completely in the bag under direct visu-
operating room. Before firing the stapling device, direct alization (Fig. 8.11). The bag should also be closed under
visualization of proper alignment of the stapler, including direct visualization because injury to the kidney or ureter
the distal extent of the device and proper position of the can occur if they are not contained within the bag. Rarely,
staple cartridge, should be confirmed by the surgical team a kidney may be unable to be placed in the bag because
(Fig. 8.10). This includes care not to close the stapler across of size or other technical issues. Manual retrieval should
metal clips, which can cause misfire. There have been lim- then be performed quickly and if possible while retaining
ited reports of stapler misfires; however, they can be cata- pneumoperitoneum to aid in identification and control of
strophic and require expedient management.25 The most the kidneys. The rectus can be opened in either a vertical
significant danger is for improper stapling before division or transverse direction as the kidney is retrieved from the
of the vessel with a cutting device. A transected bleeding abdominal cavity.
artery or vein should be controlled directly with a laparo- The kidney is immediately placed on ice and brought to
scopic instrument or hand if possible. If the vessel can be the back table for preparation. Staple lines should be tran-
controlled, determination can be made whether the vessel sected and direct cannulation and flushing of all renal arter-
can be restapled, clipped, or oversewn with laparoscopic ies is performed until clear effluent is achieved.
techniques. If this is not possible, direct pressure should The extraction site can be closed with either running or
be attempted while a midline laparotomy incision is made interrupted absorbable (PDS or Maxon) #1 sutures. After
for direct exposure and repair. Noncutting stapling devices closure of the extraction incision, pneumoperitoneum is
124 Kidney Transplantation: Principles and Practice

reestablished and confirmation of good hemostasis at all


dissection and vascular division sites is performed. Occa-
sionally, gonadal vessels will require additional clip place-
ment at the level of the ureteral division. The renal artery
and vein should be directly observed. Blood is aspirated
from the retroperitoneal space and near the spleen to con-
firm no unidentified bleeding. Hemostatic agents are not
generally required, but can be placed as an adjunct to
hemostatic maneuvers when necessary. Mesenteric defects
can be repaired at this point if identified (as discussed ear-
lier). Rarely, challenging bleeding can occur from adrenal,
splenic, and lumbar venous sources. If adrenal or splenic
bleeding cannot be confirmed to be controlled, consider-
ation for removal should be given. Lumbar venous bleeding
can be difficult to manage, and if direct control and sealing
with energy device or clip placement cannot be achieved,
an attempt to directly oversew the vessel should be made.
The procedure should never be concluded in the absence of
perfect hemostasis, and drains are almost never indicated.
Ports can be removed under direct visualization. Larger
port (12- and 15-mm) sites can be closed as practiced per
routine of the surgical team. The extraction and port sites
are anesthetized with injectable agents (lidocaine or mar-
caine) and closed with absorbable subcuticular sutures. Fig. 8.12 Cosmetic result of single-port donor nephrectomy 2 years
Ketorolac can be used per physician discretion to minimize postdonation. Single-port donor nephrectomy performed through
the umbilicus with transumbilical extraction allows for minimization
postoperative discomfort and narcotic use. of the apparent incision length with minimal residual scar. (Ann Surg.
2013;257:527–33.)

Right Donor Nephrectomy


donor procedure in the world, select centers perform lapa-
Laparoscopic right donor nephrectomy is rarely performed, roscopic donor nephrectomy through a single small inci-
with rates between 1% to 4% at large US centers. Initial sion. The technique was first described in 2008 as a feasible
technical challenges with laparoscopic right nephrectomy approach with good outcomes.28 Some supportive evidence
resulted in increased vascular complications compared exists that this approach offers improved recovery in com-
with left kidneys. Modifications in donor techniques to pre- parison to standard laparoscopic techniques.29 Our cen-
serve donor vein length, and in the recipient to mobilize the ter transitioned to a single-port laparoendoscopic (LESS)
iliac venous system, can improve these outcomes.27 None- technique in 2009. This is now our standard approach in
theless, multiple renal arteries and anomalous renal venous all cases. The rationale for this transition lay in the ability
anatomy are not contraindications for left donor nephrec- to perform the entire dissection and extraction through a
tomy. The presence of stones, cysts, or lesions within the small incision concealed within the umbilicus of the donor.
right kidney are strong indications for right, rather than This has led to a very small residual scar once healed (Fig.
left, nephrectomy. 8.12), and we have shown equivalent safety with improved
The operative approach is modified by the requirement of patient satisfaction compared with multiport laparoscopy.30
liver retraction. This is achieved through limited right lobe The single-port devices that are commercially available
mobilization with placement of a liver retractor to elevate typically allow for entry of 3 to 4 instruments (Fig. 8.13).
the liver over the superior pole of the right kidney. The Although the technical steps in LESS donor nephrectomy
operation is further modified by the requirement for divi- are not significantly different from those in standard mul-
sion of the right gonadal vein that inserts directly into the tiport approaches, the surgeon’s hands are often closely
vena cava. The right adrenal vein generally does not need opposed to each other, and instruments must frequently
to be divided because it is separate from the right renal vein. cross within the confines of the abdominal cavity. Although
The shorter right renal vein also requires extra attention to these conditions are anathema to the basic tenets of lapa-
stapling with either cutting or noncutting vascular stapling roscopy as taught to earlier generations, basic maneuvers
devices. Maximum retraction of the kidney and exposure of can lessen their effect. The procedure can be performed
the vena cava are performed to allow placement of the sta- with standard laparoscopic instrumentation and cam-
pling device for maximum vein length. era. The substitution of the single-port device for the
multiple ports required in either hand-assisted or total lapa-
roscopic approaches does not demonstrate substantial cost
Single-Port Donor Nephrectomy differences.
It is difficult to describe the technique and instrumenta-
Although multiport laparoscopic and hand-assisted tion with absolutism, and optimum exposure and safety is
donor nephrectomy techniques have supplanted the open typically achieved after a brief period of trial and error using
donor nephrectomy and are clearly the most widely offered alternative bed positioning (both Trendelenburg/reverse
8 • Donor Nephrectomy 125

Fig. 8.13 Single port donor nephrectomy. Patient position and operating room setup are similar to standard laparoscopic nephrectomy. Surgeon
and assistant are in close proximity as multiple instruments and camera are inserted through the umbilical port. The assistant operating the camera and/
or additional instrument must pay attention toward avoiding interference with the primary surgeon’s instruments.

Trendelenburg and leftward/rightward tilting), operating should be determined for extraction of the kidney after divi-
instruments of differential length (bariatric length in one sion of the vasculature. Depending on the device used, the
hand, standard length in the other), and alternative port skin and fascial incisions require extension to safely deliver
positioning within the single-port device. We have found the kidney without significant trauma. Removal through
that the deflectable tip 5-mm camera provides ideal visu- too small a fascial or skin incision can injure the kidney and
alization without undue steric hindrance to the operating should not be aggressively attempted.
surgeon. In cases of right donor nephrectomy, we use a Ultimately, additional port placement may be required.
single-port device that can accommodate a fourth port for Once substantial experience has been gained with the tech-
the liver retractor. nique, this tends to be required in less than 10% of cases.
We found four techniques that were important to mastery Commitment to early placement of additional ports in chal-
of the single-port approach and normalization of operative lenging cases has allowed for equivalent safety in our expe-
times compared with total laparoscopic approaches. First, rience. The most critical maneuvers in any laparoscopic
ventilation of smoke and vapor through the single-port donor nephrectomy are the vascular dissection around the
device is critical, and recent generations of port devices have vein and artery, and subsequent stapling. These maneuvers
incorporated this into their design. Second, elevation of the involve fine movements that we have found not to be hin-
lower pole of the kidney anteriorly and medially allows for dered by a single-port approach.
opening of the space between the renal artery and vein for We have reported on the normalization of our operative
dissection. Third, retraction of the upper pole inferiorly and times and the ability to perform right and left nephrecto-
laterally provides separation of the kidney from splenic and mies with single or multiple arteries and veins.30 Although
adrenal attachments, and facilitates dissection of the renal complication rates are similar to other techniques, concern
artery from a superior/cephalad approach. Finally, a plan for umbilical hernia necessitates careful attention to the
126 Kidney Transplantation: Principles and Practice

proper technique for closure of the fascia. The umbilical 100


hernia rate is approximately 3% at our center, and the sur- 6 weeks
gical technical complication rate is comparable to the rates 80
6 months
seen in total laparoscopic and hand-assisted approaches

Percentage
60 12 months
at other centers.31 This technique continues to be our pre-
ferred approach for all living kidney donors and fits into an 40
algorithm of preferred approaches consisting of (in order)
single-port laparoscopy, multiport laparoscopy, hand- 20
assisted laparoscopy, and open techniques. The improved
cosmesis and potential other benefits of this technique may 0
also translate to increased interest in living kidney dona- No Yes Unknown
tion with a further slight reduction in the invasiveness of Response
the surgery. Fig. 8.14 Complications of living kidney donation from 2010 to 2014.
Overall complication rate of 8.8% observed at 12 months after kidney
donation. (OPTN/SRTR 2015 Annual Data Report. HHS/HRSA.)
Robotic Donor Nephrectomy
Robotic technology has been incorporated into laparo- hernia formation, and bowel obstruction (Fig. 8.14).1 In
scopic donor nephrectomy. Thus far, reports have been the 5-year period between 2010 and 2015, a total of 17
limited to single-center studies, and the techniques have deaths occurred in prior living renal donors within 1 year of
not become widely used by the community as a whole.32–34 donation. Seven of these deaths were medically related and
Although the purported advantages of robotic technology five were the result of accident or homicide. Prior analysis
include (1) improved visualization with three-dimensional of more than 80,000 renal donors in the US who donated
(3D) camera systems, (2) articulating laparoscopic instru- between 1994 and 2009 revealed a surgical mortality rate
mentation allowing meticulous dissection of complicated of 0.03%.40
vascular anatomy, and (3) ease of intracorporal sutur- Single center reports from large centers tend to provide
ing, their applicability to donor nephrectomy has yet to be more granular information regarding the types and fre-
broadly accepted or defined. Present robotic approaches quencies of complications after donation.
are performed with multiple laparoscopic ports and require An early single-center study reporting 1200 laparoscopic
bedside manual assistant ports for the use of energy devices, renal donors demonstrated an intraoperative complication
staplers, and for renal extraction. Whereas the feasibility rate of 1.6% with a conversion rate of 0.92% (most com-
of robotic-assisted laparoscopic nephrectomies have been monly as a result of renovascular injury). An additional
clearly demonstrated, early reports have not demonstrated 4.0% of patients presented with a postoperative complica-
significant advantages over total laparoscopic and hand- tion with only three patients requiring surgery for internal
assisted techniques, while demonstrating increased cost.35 hernia or ileus.41 Our center had previously reported our
Single-port platforms have recently been introduced, and experience with more than 700 donors, where we dem-
these platforms have thus far been used predominantly for onstrated an open conversion rate of 1.6% (again most
LESS cholecystectomy.36 Following studies demonstrating commonly as a result of vascular injury); 1.2% of patients
the use of the single-port platform in renal surgery,37,38 required blood transfusion.42 Five patients also had a bowel
our group embarked on a brief clinical trial evaluating obstruction requiring subsequent exploration and one
the robotic single-port platform in donor nephrectomy.39 patient required splenic laceration repair. In another report
Although robotic technology (which offers an improved of 1000 hand-assisted donor nephrectomies, a hernia rate
3D field of view and articulating instrumentation) has the of 4% was noted, with 0.3% of patients requiring transfu-
potential to compensate for the visual and technical limi- sion and 1.5% requiring reoperation.43 We have recently
tations associated with LESS nephrectomy, current instru- reported our results with nearly 400 consecutive single-
mentation for the single-port platform does not allow for port donor nephrectomies.31 We noted an umbilical hernia
adequate instrument articulation or the use of energy rate of 3%. Although only one patient required conversion
devices. Continued technologic advancement may improve to an open procedure, an additional seven required a return
the ability of robotic approaches to add significant advan- to the operating room for internal hernia (2), evisceration
tages to donor surgery. (1), bleeding (1), enterotomy (1), and wound infection (2).

Complications Summary
The 2015 report from the Organ Procurement and Trans- Living donor nephrectomy can be successfully performed
plantation Network (OPTN) and Scientific Registry of through a variety of techniques, both open and laparo-
Transplant Recipients (SRTR) provides the most recent scopic. Minimally invasive techniques have been associ-
compilation of donor complications rates in the US. Com- ated with decreased morbidity and improved recovery and
plications occurred in 5.3% of donors within 6 weeks of should be viewed as the preferred approach for the majority
donation. Overall 8.8% of donors reported a complication of patients. Nonetheless, the key principle of donor safety
potentially related to the organ donation within the first should be used to make final decisions regarding donation
year of donation. Reported complications included bleeding, techniques. Different centers and surgeons may have a
8 • Donor Nephrectomy 127

variety of approaches that result in good and safe outcomes. 21. Jacobs SC, Cho E, Dunkin BJ, et al. Laparoscopic live donor nephrec-
Thus surgeon experience becomes an important consider- tomy: the University of Maryland 3-year experience. J Urol
2000;164(5):1494–9.
ation in determining the specific approach that is best for 22. Kuo PC, Johnson LB, Sitzmann JV. Laparoscopic donor nephrectomy
each patient. with a 23-hour stay: a new standard for transplantation surgery. Ann
Acknowledgement is made to Phil Brazio, MD, for illus- Surg 2000;231(5):772–9.
trating the technique of laparoscopic donor nephrectomy 23. Schweitzer EJ, Wilson J, Jacobs S, et al. Increased rates of donation
with laparoscopic donor nephrectomy. Ann Surg 2000;232(3):392–
accompanying this chapter. 400.
24. Slakey DP, Wood JC, Hender D, Thomas R, Cheng S. Laparoscopic
References living donor nephrectomy: advantages of the hand-assisted method.
1. OPTN/SRTR 2015 Annual Data Report. Department of Health and Transplantation 1999;68(4):581–3.
Human Services, Health Resources and Services Administration; 25. Hsi RS, Ojogho ON, Baldwin DD. Analysis of techniques to secure the
2016. Available online at: http://srtr.transplant.hrsa.gov/annual_ renal hilum during laparoscopic donor nephrectomy: review of the
reports/Default.aspx. (accessed 28.02.19). FDA database. Urology 2009;74(1):142–7.
2. Goldberg DS, Blumberg E, McCauley M, Abt P, Levine M. Improving 26. Friedman AL, Peters TG, Ratner LE. Regulatory failure contributing to
organ utilization to help overcome the tragedies of the opioid epi- deaths of live kidney donors. Am J Transplant 2012;12(4):829–34.
demic. Am J Transplant 2016;16(10):2836–41. 27. Mandal AK, Cohen C, Montgomery RA, Kavoussi LR, Ratner LE.
3. Rao PS, Schaubel DE, Guidinger MK, et al. A comprehensive risk Should the indications for laparascopic live donor nephrectomy of the
quantification score for deceased donor kidneys: the kidney donor risk right kidney be the same as for the open procedure? Anomalous left
index. Transplantation 2009;88(2):231–6. renal vasculature is not a contraindiction to laparoscopic left donor
4. Israni AK, Salkowski N, Gustafson S, et al. New national allocation nephrectomy. Transplantation 2001;71(5):660–4.
policy for deceased donor kidneys in the United States and possible 28. Gill IS, Canes D, Aron M, et al. Single port transumbilical (E-NOTES)
effect on patient outcomes. J Am Soc Nephrol 2014;25(8):1842–8. donor nephrectomy. J Urol 2008;180(2):637–41; discussion 641.
5. Wind J, Snoeijs MG, van der Vliet JA, et al. Preservation of kidneys from 29. Canes D, Berger A, Aron M, et al. Laparo-endoscopic single site (LESS)
controlled donors after cardiac death. Br J Surg 2011;98(9):1260–6. versus standard laparoscopic left donor nephrectomy: matched-pair
6. Magliocca JF, Magee JC, Rowe SA, et al. Extracorporeal support for comparison. Eur Urol 2010;57(1):95–101.
organ donation after cardiac death effectively expands the donor pool. 30. Barth RN, Phelan MW, Goldschen L, et al. Single-port donor nephrec-
J Trauma 2005;58(6):1095–101; discussion 1092–101. tomy provides improved patient satisfaction and equivalent outcomes.
7. Ausania F, White SA, Pocock P, Manas DM. Kidney damage during Ann Surg 2013;257(3):527–33.
organ recovery in donation after circulatory death donors: data from UK 31. LaMattina JC, Powell JM, Costa NA, et al. Surgical complications of
National Transplant Database. Am J Transplant 2012;12(4):932–6. laparoendoscopic single-site donor nephrectomy: a retrospective
8. Jochmans I, Moers C, Smits JM, et al. Machine perfusion versus study. Transpl Int 2017;30(11):1132–9.
cold storage for the preservation of kidneys donated after car- 32. Hubert J, Renoult E, Mourey E, Frimat L, Cormier L, Kessler M. Com-
diac death: a multicenter, randomized, controlled trial. Ann Surg plete robotic-assistance during laparoscopic living donor nephrec-
2010;252(5):756–64. tomies: an evaluation of 38 procedures at a single site. Int J Urol
9. Watson CJ, Wells AC, Roberts RJ, et al. Cold machine perfusion 2007;14(11):986–9.
versus static cold storage of kidneys donated after cardiac death: 33. Gorodner V, Horgan S, Galvani C, et al. Routine left robotic-assisted
a UK multicenter randomized controlled trial. Am J Transplant laparoscopic donor nephrectomy is safe and effective regardless of the
2010;10(9):1991–9. presence of vascular anomalies. Transpl Int 2006;19(8):636–40.
10. Minambres E, Suberviola B, Dominguez-Gil B, et al. Improving the 34. Horgan S, Vanuno D, Sileri P, Cicalese L, Benedetti E. Robotic-assisted
outcomes of organs obtained from controlled donation after circula- laparoscopic donor nephrectomy for kidney transplantation. Trans-
tory death donors using abdominal normothermic regional perfusion. plantation 2002;73(9):1474–9.
Am J Transplant 2017;17(8):2165–72. 35. Boger M, Lucas SM, Popp SC, Gardner TA, Sundaram CP. Comparison
11. London ET, Ho HS, Neuhaus AM, Wolfe BM, Rudich SM, Perez RV. of robot-assisted nephrectomy with laparoscopic and hand-assisted
Effect of intravascular volume expansion on renal function during pro- laparoscopic nephrectomy. JSLS 2010;14(3):374–80.
longed CO2 pneumoperitoneum. Ann Surg 2000;231(2):195–201. 36. Konstantinidis KM, Hirides P, Hirides S, Chrysocheris P, Geor-
12. Kok NF, Lind MY, Hansson BM, et al. Comparison of laparoscopic and giou M. Cholecystectomy using a novel Single-Site® robotic plat-
mini incision open donor nephrectomy: single blind, randomised con- form: early experience from 45 consecutive cases. Surg Endosc
trolled clinical trial. BMJ 2006;333(7561):221. 2012;26(9):2687–94.
13. Perry KT, Freedland SJ, Hu JC, et al. Quality of life, pain and return to 37. Khanna R, Stein RJ, White MA, et al. Single institution experience
normal activities following laparoscopic donor nephrectomy versus with robot-assisted laparoendoscopic single-site renal procedures.
open mini-incision donor nephrectomy. J Urol 2003;169(6):2018–21. J Endourol 2012;26(3):230–4.
14. Yang SL, Harkaway R, Badosa F, Ginsberg P, Greenstein MA. Minimal 38. Kaouk JH, Autorino R, Laydner H, et al. Robotic single-site kidney
incision living donor nephrectomy: improvement in patient outcome. surgery: evaluation of second-generation instruments in a cadaver
Urology 2002;59(5):673–7. model. Urology 2012;79(5):975–9.
15. Clayman RV, Kavoussi LR, Soper NJ, et al. Laparoscopic nephrectomy: 39. LaMattina JC, Alvarez-Casas J, Lu I, et al. Robotic-assisted single-port
initial case report. J Urol 1991;146(2):278–82. donor nephrectomy using the da Vinci single-site platform. J Surg Res
16. Ratner LE, Ciseck LJ, Moore RG, Cigarroa FG, Kaufman HS, Kavoussi LR. 2018;222:34–8..
Laparoscopic live donor nephrectomy. Transplantation 1995;60(9): 40. Segev DL, Muzaale AD, Caffo BS, et al. Perioperative mortal-
1047–9. ity and long-term survival following live kidney donation. JAMA
17. Flowers JL, Jacobs S, Cho E, et al. Comparison of open and laparoscopic 2010;303(10):959–66.
live donor nephrectomy. Ann Surg 1997;226(4):483–9; discussion 41. Leventhal JR, Paunescu S, Baker TB, et al. A decade of minimally
489–90. invasive donation: experience with more than 1200 laparo-
18. Wolf Jr JS, Merion RM, Leichtman AB, et al. Randomized controlled scopic donor nephrectomies at a single institution. Clin Transplant
trial of hand-assisted laparoscopic versus open surgical live donor 2010;24(2):169–74.
nephrectomy. Transplantation 2001;72(2):284–90. 42. Jacobs SC, Cho E, Foster C, Liao P, Bartlett ST. Laparoscopic donor
19. Nogueira JM, Cangro CB, Fink JC, et al. A comparison of recipient nephrectomy: the University of Maryland 6-year experience. J Urol
renal outcomes with laparoscopic versus open live donor nephrec- 2004;171(1):47–51.
tomy. Transplantation 1999;67(5):722–8. 43. Serrano OK, Kirchner V, Bangdiwala A, et al. Evolution of living donor
20. Simforoosh N, Basiri A, Tabibi A, Shakhssalim N, Hosseini Moghad- nephrectomy at a single center: long-term outcomes with 4 different
dam SM. Comparison of laparoscopic and open donor nephrectomy: a techniques in greater than 4000 donors over 50 years. Transplanta-
randomized controlled trial. BJU Int 2005;95(6):851–5. tion 2016;100(6):1299–305.
9 Kidney Preservation
JOHN O’CALLAGHAN, GABRIEL ONISCU, HENRI LEUVENINK,
PETER J. FRIEND and RUTGER J. PLOEG

CHAPTER OUTLINE Introduction Normothermic Regional Perfusion


Organ Damage After Death Technical Variations
Cold Storage Clinical Outcomes
Cell Swelling Normothermic Machine Perfusion
Energy and Acidosis Experimental Evidence
Calcium Clinical Evidence
Intracellular Enzymes and Organelles Future Directions
Reactive Oxygen Species Tailoring Preservation Strategies
Clinical Use of Preservation Solutions
Pharmacologic Additives
Hypothermic Machine Perfusion

Introduction transplant programs are therefore turning to older and


higher risk donors, previously defined as expanded criteria
At the start of the first transplant programs, the donor and donation (ECD) and DCD.
recipient would be operated in the same surgical center. The The change in the risk profile for kidneys we now use to
only preservation method therefore undertaken would be meet the demand for organs means that it is now neces-
to flush out the kidney with either blood or Ringer’s lactate sary to revisit our methods of preservation. The UK Kidney
and to use surface cooling with ice water, because hypo- Donor Risk Index (UKKDRI)5 uses donor factors shown in
thermia was supposed to be protective by reducing metab- Table 9.1 to predict overall transplant survival. The pro-
olism. Very early transplant work in twins and animal portion of DBD kidneys transplanted in the UK from high-
studies suggested that an ischemic time of less than 1 hour risk donors (HR ≥1.35) using this index has increased from
should be aimed for, with irreversible damage suspected 29% to 39% in the past 10 years.6 The Kidney Donor Profile
beyond 3 hours.1 This target would later be extended to 12 Index (KDPI) is used to predict the risk of failure for a kid-
hours with improved flushing and cooling.1 The necessity ney transplant in the US; the current proportion of deceased
of transporting kidneys to compatible recipients in other donor kidneys with the worst KDPI (86%–100%) is 18%,
local centers meant adequate preservation for longer peri- having risen from 14% in 2012.7
ods was needed. The first method of addressing this was the The increasing use of DCD kidneys has also prompted a
machine preservation system developed by Belzer (Fig. 9.1). renewed interest in preservation methods; despite the ade-
It was not until the late 1960s that preservation fluids were quate long-term function of kidneys from DCD, they still
developed that allowed static cold storage without continu- have a higher risk of delayed graft function (DGF) compared
ous perfusion.2 with kidneys from DBD. In the UK the use of DCD kidneys has
Modern transplant programs often involve the shar- risen dramatically, from 3% of deceased donor transplants
ing of organs over much larger geographic distances. The in 2000 to 42% in 2017.6 In the Eurotransplant region the
­sharing of kidneys in this way permits better matching use of DCD kidneys has permitted a large increase in avail-
between donor and recipient, but may have the additional able kidneys for transplantation.8 The use of DCD in the
effect of longer cold ischemic times, which can affect graft US has increased, although not at such a rapid rate; DCD
survival.3 accounted for approximately 17% of deceased donor kidney
The first transplant programs used either live donors or transplants in the US in 2016, having increased from 10%
donation after circulatory death (DCD) when the opportu- in 2007.7
nity to quickly perform organ retrieval was available. In
1968 a committee at Harvard Medical School met to exam-
ine and redefine the definition of brain death.4 This was Organ Damage After Death
followed by an expansion in the use of organs from dona-
tion after brain death (DBD). These were typically younger Organ damage around the time of death can be divided into
people with irreversible brain damage during motor five broad phases that have different implications for the
vehicle accidents, referred to as standard criteria donors transplant and may potentially be addressed by targeting
(SCD). There is a declining number of this donor type, and preservation techniques at each phase (Fig. 9.2).
128
9 • Kidney Preservation 129

Cerebral injury and brain death are associated with the DCD, however, undergo a period of warm ischemia before
release of cytokines and the initiation of inflammatory pro- retrieval. The extent of this damage is related to the situ-
cesses that can directly injure organs and lead to further ation of the donor, and this may be categorized using the
immune damage at reperfusion in the recipient.9–12 These Maastricht system, which is shown in a modified form in
changes in immunogenicity are characterized by increased Table 9.2.16,17 It should be noted that not all of these cate-
expression of damage-associated molecular patterns (DAMPs) gories are acceptable in some countries. In the case of Maas-
that may include cytokines, adhesion molecules, and major tricht category III donors, the warm ischemic period follows
histocompatibility complex class II antigens in the retrieved the withdrawal of supportive treatment once a retrieval
organ.11,12 Hormonal changes after brain death include team is ready, intending to minimize warm ischemic time.
reduced levels of antidiuretic hormone and thyroid hor- In some cases the withdrawal of support may be prolonged,
mones.13 Brain death also causes myocardial suppression and and a systolic blood pressure of less than 50 mm Hg results
decreased vascular tone, which results in hypotension and in functional warm ischemia and may affect the transplant
inadequate perfusion of organs.14 Kidneys in this environment outcome. In the case of the so-called “uncontrolled” donors
are exposed to perfusion dynamics and inflammatory cyto- of Maastricht categories I and II, there may be a prolonged
kines that can lead to necrosis of renal tubules and fibrous warm ischemic period after circulatory arrest and before
proliferation of the arterial intima.15 cold perfusion of the organs can be achieved.16
Organs from DCD are exposed to inflammatory cyto- The period between the initial flush of cold preserva-
kines, but in a less profound way to DBD. Organs from tion fluid through the organ’s arteries in the donor and
the reperfusion of an organ with blood in the recipient is
the cold ischemic time (CIT). The length of this potentially
modifiable period is an important factor in determining the
outcome of solid-organ transplantation. Analysis of the Col-
laborative Transplant Study (CTS), a voluntary, multina-
tional database of transplant outcomes, found that kidneys
preserved for longer than 19 hours had worse graft sur-
vival.18 A longer CIT is also associated with an increasing
risk of DGF of kidneys.19,20
The simplest method of dealing with the harmful cellular
processes after donor death has been to cool the kidneys. This
slows down the metabolism but does not completely stop it,
leading to ongoing damage in cold-preserved organs, and
hence the critical importance of limiting cold ischemic times.

Cold Storage
When a kidney is removed from the donor’s body, perfu-
Fig. 9.1 The first Belzer machine perfusion unit and its transport sion completely ceases, leading to an absence of oxygen and
­system. nutrient delivery to the renal cells, which will rapidly lead
to serious metabolic problems. Suppression of metabolism
TABLE 9.1 Comparing the UK Kidney Donor Risk Index is therefore essential to maintain organ viability during the
(UKKDRI) with the US Kidney Donor Risk Index (KDRI) and preservation period. Reduction of the core temperature of the
the Kidney Donor Prediction Index (KDPI) kidney below 4°C will result in a reduction of metabolism to
5% to 8% and will diminish enzyme activity.21 The concept
Factors Used to Calculate the Factors Used to Calculate the
UKKDRI, Watson et al.5 KDRI and KDPI, OPTN7
of simple cooling with ice proved to be successful; however,
it was unfit for longer preservation periods.1 The concept
Donor age Donor age of simple cold static storage was investigated by several
History of hypertension History of hypertension groups, resulting in new solutions and providing an insight
Donor weight Donor weight
Use of adrenaline Donor height
into the side effects of ischemia in the preserved organ.2,22,23
Days in hospital Ethnicity/race Transplant pioneers started perfusion of kidneys with cryo-
History of diabetes preserved plasma-based solutions, leading to improved pres-
Cause of death ervation, although accompanied by logistical problems.24 In
Serum creatinine current clinical practice several preservation fluids are used.
Hepatitis C virus status
DCD/DBD Both in situ in the deceased donor, and on the theater work-
bench, kidneys are flushed through the renal artery with a
DBD, donation after brain death; DCD, donation after circulatory death. preservation fluid until donor blood is completely cleared.

Organ Preservation Reperfusion


Donor
Patient retrieval awaiting in the
management
process transplant recipient

Fig. 9.2 Potential stages of the retrieval process during which kidney allografts may accrue damage that affects the future transplant function. The
mechanisms of damage during each stage differ.
130 Kidney Transplantation: Principles and Practice

The kidney is then packaged in a sterile bag of this fluid and creates a hyperosmolar intracellular environment and sub-
kept on ice in a cool box. It is important that handling of the sequently an influx of water.25 To reestablish the disturbed
kidney and workbench preparation are undertaken with the Donnan equilibrium and to prevent cell swelling, imperme-
organ submerged in preservation fluid. The fluid should also ants such as colloids (hydroxyethyl starch in the University
be kept on ice until required, to prevent warming to ambient of Wisconsin (UW) solution or polyethylene glycol in IGL-1)
temperature. This method is relatively cheap, easily trans- are added to preservation solutions.26,27 It was thought
portable, and does not require input from the retrieval or that intracellular composition was required to diminish the
implant team during the preservation period. exchange of electrolytes. The high potassium, however, also
The classic unwanted side effects of hypothermia are swell- leads to initial vasoconstriction, hampering flush-out in the
ing, acidosis, altered enzyme activity, and production of radi- donor.28 The extracellular-type solutions are as effective as
cal oxygen species upon reperfusion. Effective preservation the intracellular solutions indicating that creating an osmotic
solutions are therefore composed to counteract these processes pressure is more important than the composition.29
using different types of buffers, electrolyte compositions, and
additives. These processes are summarized in Fig. 9.3. ENERGY AND ACIDOSIS
The absence of oxygen during storage results in a rapid
CELL SWELLING
fall in intracellular ATP levels.30 Even at 0°C to 4°C cellu-
The main cause of cell swelling is the impaired activity of lar ATP content is rapidly depleted, and cells will switch to
Na+/K+ ATPase.23 As a result, sodium passively enters cells, anaerobic metabolism of glucose.31 This will lead not only to
attracted by the negative charge of cytoplasmic proteins. This a much less efficient production of ATP but also to produc-
tion of lactic acid. Severe acidosis activates phospholipases
and proteases causing lysosomal damage and eventually
TABLE 9.2 Paris Modified Maastricht Classification for cell death.32 Adequate control of pH by a buffering agent is
Donation After Circulatory Death (DCD) therefore an important function of preservation solutions.
UNCONTROLLED DCD
I. Found dead Sudden unexpected cardiac arrest CALCIUM
without any resuscitation by a
medical team; may be in hospital or Under normal physiologic conditions the free calcium con-
out of hospital centration difference between the intracellular and extracel-
II. Witnessed cardiac arrest Sudden unexpected irreversible lular fluid is maintained by ATP-dependent transporters.33
cardiac arrest with unsuccessful During cold preservation, cellular ATP concentrations are
resuscitation by a medical team;
may be in or out of hospital
low, leading to increased intracellular calcium. Preservation
solutions therefore contain a low concentration of calcium.
CONTROLLED DCD Experimentally calcium blockers in preservation solutions
III. Withdrawal of life- Planned withdrawal of life-sustaining are shown to be effective in preventing activation of calcium-
sustaining treatment therapy, expected cardiac arrest
IV. Cardiac arrest while brain Sudden cardiac arrest after brain death
dependent processes such as calpain activation, an enzyme
dead diagnosis during donor life- involved in the breakdown of the cytoskeleton.34,35
management but before organ
recovery
INTRACELLULAR ENZYMES AND ORGANELLES
Adapted from Thuong M, Ruiz A, Evrard P, et al. New classification of dona-
tion after circulatory death donors definitions and terminology. Transpl Int Intracellular proteases are involved in the breakdown of
2016;29(7):749–59. proteins during preservation, most likely due to the absence

Na+

– Na+ H2O
Protein– ROS
K+
– Cell Swelling
Ca2+
Continued, slow Organelle and membrane damage
metabolism
Ca2+
Hypoxia Lysosome rupture
Lactic acidosis

Acidosis

Fig. 9.3 Negative effects of cold ischemia on cells and organelles. Continued but slowed metabolism results in hypoxia, acidosis, reduced ATP synthesis,
and activity of sodium and calcium pumps. Resultant sodium influx with water leads to cell swelling. Reactive oxygen species (ROS), along with acidosis
and lysosomal enzymes, damage cellular organelles and membranes.
9 • Kidney Preservation 131

of oxygen. Also, matrix metalloproteinases (MMPs) may be TABLE 9.3 Key Preservation Fluids (Generic Names),
activated during cold preservation leading to detachment of Common Acronyms/Abbreviations, and When Each Was
endothelial cells from the underlying matrix.36 Hydroxyethyl Developed
starch present in UW solution may reduce this detrimental
Euro-Collins’ Solution (developed from Col- EC 1960s–1970s
effect.37 Another relevant family of enzymes activated dur- lins’ Solution)
ing cold preservation are apoptosis-related caspases.38 The Hyper-Osmolar Citrate (Marshall’s Solution) HOC 1970s
release of mitochondrial proteins leads to endoplasmic retic- Histidine-Tryptophan-Ketoglutarate HTK 1970s
ulum stress and dysfunctional autophagy fluxes.39,40 University of Wisconsin Solution UW 1980s
Belzer Machine Perfusion Solution MPS 1980s
Celsior Solution Celsior 1990s
REACTIVE OXYGEN SPECIES Institut Georges Lopez-1 Solution IGL-1 1990s

Reactive oxygen species (ROS) are generated by several pro-


cesses in ischemic and postischemic reperfused organs.41 A
well-studied generator of ROS is xanthine oxidase, which of preservation.51 Several pharmacologic additives have
simultaneously produces hydrogen peroxide (H2O2) and the been investigated in preclinical models. The most promis-
superoxide anion (O2−).42 The subsequent reduction of H2O2, ing additives are trimetazidine (an antiischemic agent),52,53
catalyzed by iron, leads to hydroxyl radical formation.43 innate immunity inhibitors such as complement inhibi-
The infiltration of leukocytes in the graft after reperfusion tors,54,55 and regulators of endothelial function.56,57
also results in the production of superoxides (the respiratory
burst). Lastly, mitochondrial dysfunction resulting from par-
tial reduction of the respiratory chain is an important con- Hypothermic Machine Perfusion
tributor to ROS formation after reperfusion.43 These ROS
react rapidly with other intracellular molecules, causing The decreasing number of high-quality SCD kidneys trig-
severe damage to lipids, nucleic acids, and proteins during gered the reintroduction of hypothermic machine perfusion
reperfusion. Some reports suggest that oxygen radicals are (HMP). The retrieved kidney is placed within a chamber
formed during reperfusion and during cold preservation.44 filled with chilled preservation solution surrounded by an
Modern preservation solutions contain scavengers or precur- ice box. The renal artery is cannulated by one end of a sys-
sors for ATP production, which are shown to be effective in tem of tubing, and a pump is used to generate a pulsatile or
experimental research but less evident in clinical trials.45 continuous flow of preservation solution through the renal
vessels. The fluid pours from the renal vein into the reser-
voir where the pump collects it again to recirculate it. The
CLINICAL USE OF PRESERVATION SOLUTIONS
concept of machine perfusion was first described by Charles
Worldwide, the UW solution and histidine-tryptophan- Lindberg in 1935 in a fully mechanical device able to main-
ketoglutarate (HTK) solution have been most commonly tain an oxygenated pulsatile perfusion.58 In the context of
used since the early 1990s when two randomized con- organ transplantation, Professor Belzer experimented in
trolled trials (RCTs) showed lower DGF rates compared the 1960s using techniques adapted from cardiac bypass
with Euro-Collins’ solution.46,47 Registry data from the CTS machines. Through a series of preclinical experiments
suggests that at very long preservation times (beyond 24 using canine kidneys, he was able perform the first success-
hours) UW-stored kidneys have a reduced risk of graft loss ful human HMP kidney transplant in 1968.59 The intro-
compared with other preservation fluids.18 Registry data duction of cold storage solutions overcoming the problems
from the United Network for Organ Sharing (UNOS) show with logistics and high costs meant that machine perfusion
that kidney preservation in UW has improved graft survival was almost abandoned from clinical practice, although in
independent of preservation time.48 The lack of adequately some centers, mainly in the US, it never totally disappeared.
powered studies comparing Institut Georges Lopez-1 (IGL-1) Over more recent years the development of more trans-
solution, Celsior, or hyperosmolar citrate (HOC) against portable stand-alone machines resolved many logistic
HTK or UW so far prevents definite conclusions regarding hurdles. Commercially available machines for HMP of
the superiority of any newer preservation fluids. From small kidneys include the RM3 and Waves (IGL Group), the Life-
published studies one could conclude that results are similar port (Organ Recovery Systems), the Kidney Assist (Organ
to UW or HTK.49 Most studies were conducted with rela- Assist), and the Airdrive (Portable Organ Perfusion)
tively good quality donor kidneys. Globally UW and HTK are (Figs. 9.4–9.6).
most predominant, with some countries using other pres- Several retrospective and small prospective studies indi-
ervation fluids that were developed locally being popular cated superior preservation compared with static cold stor-
alternatives, see Tables 9.3 and 9.4. There is retrospective age, but high-quality studies have only been undertaken in
evidence from the UK, where HOC is still used for the preser- more recent years. In 2009 the machine perfusion (MP) trial
vation of large numbers of kidneys, that HOC is an accept- consortium reported that continuous HMP results in lower
able alternative to UW with a mean CIT of 17 hours.50 DGF rates for SCD, ECD, and DCD kidneys and improved graft
survival for SCD and ECD kidneys.60 Overall, a reduction in
DGF was observed with HMP compared with cold storage
PHARMACOLOGIC ADDITIVES
(20.8% vs. 26.5%) with no significant effect on primary non-
A better understanding of the pathophysiology and role of function (PNF) (2.1% vs. 4.8%). Graft survival in the first
ischemia reperfusion injury in organ transplantation has year was also improved by HMP (94% vs. 90%), and in later
given us the opportunity to target certain negative aspects reports the 3-year graft survival was also better (91% vs.
132 Kidney Transplantation: Principles and Practice

TABLE 9.4 Composition of Preservation Solutions for Static Cold Storage and Machine Perfusion
Component Type Component Celsior EC HOC HTK IGL-1 UW MPS
Colloids (mM) HES — — — — — 0.25 0.25
PEG — — — — 0.03 — —
Impermeants (mM) Citrate — — 80 — — — —
Gluconate — — — — — — 85
Glucose — 195 — — — — 10
Histidine 30 — — 198 — — —
Lactobionate 80 — — — 100 100 —
Mannitol 60 — 185 38 — — 30
Raffinose — — — — 30 30 —
Ribose — — — — — — 5
Buffers (mM) HEPES — — — — — — 10
K2HPO4 — 15 — — — — —
KH2PO4 — 43 — — 25 25 25
NaHCO3 — 10 10 — — — —
Electrolytes (mM) Calcium 0.25 — — 0.0015 — — 0.5
Chloride 42 15 — 32 20 20 1
Magnesium 13 — 40 4 5 5 5
Potassium 15 115 84 9 25 120 25
Sodium 100 10 84 15 120 30 100
ROS scavengers (mM) Allopurinol — — — — 1 1 —
Glutathione 3 — — — 3 3 —
Tryptophan — — — 2 — — —
Substrates (mM) Adenine — — — — — — 5
Adenosine — — — — 5 5 —
Glutamate 20 — — — — — —
Ketoglutarate — — — 1 — — —
Osmolality (mOsm) — 255 406 400 310 320 320 300

Citrate, histidine, and lactobionate also act as buffers. Histidine, lactobionate, and mannitol also act as free radical scavengers.
EC, Euro-Collins’; HEPES, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid; HES, hydroxyethylene starch; HOC, hyperosmolar citrate; HTK, histidine-tryptophan-
ketoglutarate; IGL-1, Institut Georges Lopez-1; MPS, machine perfusion solution; PEG, polyethylene glycol; UW, University of Wisconsin solution.

Fig. 9.5 Lifeport hypothermic machine perfusion pump from Organ


Recovery Systems.

Fig. 9.4 RM3 hypothermic machine perfusion pump from IGL Group. The use of HMP in SCD and ECD (both DBD donor types)
seems beneficial in terms of reduced DGF and graft survival.
Although initially used for DCD donors, the effect of HMP on
87%).61 Maybe even more interesting was that the severity long-term survival of DCD kidneys is less evident. One of only
of DGF was less in HMP-preserved kidneys, illustrated by 3 two adequately powered RCTs in DCD kidneys showed that
days shorter DGF on average (10 days vs. 13 days).60 continuous HMP resulted in lower risk of DGF (54% vs. 70%)
The beneficial effect of HMP seems to be more pronounced but no improvement in 1-year or 3-year graft survival.63 The
in ECD kidneys as shown in the extension trial of the same other RCT was not able to show a difference in DGF between
MP consortium.62 The rate of PNF was also lower for ECD HMP (58%) and cold storage (56%).64 The main difference
kidneys preserved with MP compared with cold storage between the two studies is that in the UK study kidneys were
(3% vs. 12%). Both 1-year and 3-year graft survival was first subjected to cold storage before connection to the machine,
higher after HMP (92.3% vs. 80.2%, and 86% vs. 76%). In whereas in the Eurotransplant study kidneys were continually
this study, DGF significantly affected 1-year graft survival pumped. Meta-analyses comparing HMP with cold storage
of cold stored kidneys (41% vs. 97%) but not as much if the have established a reduction in DGF with HMP,65 but improve-
kidney had been stored by HMP (41% vs. 97%). ment in graft survival has largely been among ECD only.66
9 • Kidney Preservation 133

Reservoir with cold


UW solution

Fig. 9.6 Kidney Assist hypothermic machine perfusion pump from


Organ Assist.
Leukocyte
depletion filter

P1 O in
One could speculate that the effect of MP on DCD kid- P2

neys is different from the effect on DBD kidneys because Reservoir with
prime Solution
Centrifugal
pump Oxygenator Markers show the sites of descending
Aorta and supra-hepatic IVC clamps
DCD kidneys experience substantial ischemic injury due Heat
exchanger Pressure Transducers

to the donation procedure. The addition of oxygen during P1: Premembrane


P2: Postmembrane

preservation is therefore an interesting approach and in


preclinical models shows potential, but clinical proof is not Hb, HCT, SvO , venous temperature sensor

yet available.67 The Consortium for Organ Preservation in Fig. 9.7 Illustration of a normothermic regional perfusion circuit cur-
Europe (COPE) is trying to confirm whether the addition of rently used in the UK.
oxygen during HMP is helpful or not and whether end-HMP
is beneficial, which may significantly simplify logistics.68
cannulation is undertaken via the femoral vessels using a
percutaneous or a cut-down technique. A similar approach
Normothermic Regional Perfusion has been adopted for controlled DCD in France, Spain, and
Italy. In contrast, in the UK, where premortem administra-
After pioneering work in Maastricht category II donors in tion of heparin is not allowed, cannulation is undertaken
Spain, abdominal normothermic regional perfusion has via a rapid laparotomy with distal aortic and vena cava
emerged as potentially beneficial strategy among alterna- cannulation, similar to the ultrarapid recovery technique.
tive preservation techniques, with improved organ recov- The optimal duration of NRP is yet to be determined but
ery rates and better outcomes in Maastricht category I and around 120 minutes appears to be sufficient to restore the
II donors. cellular ATP72 and increase the levels of endogenous anti-
Conceptually, normothermic regional perfusion (NRP) oxidants.73 A prolonged NRP time of 4 hours has been
establishes an extracorporeal circuit via arterial and maintained in the setting of Maastricht category II donors
venous femoral or distal aortic and vena cava cannulation with no evidence of additional benefit.
to recirculate the donor blood and isolates the abdominal One of the key benefits of NRP is the real-time, in situ,
compartment by occluding the descending aorta. The extra- dynamic assessment of organ function, which is compos-
corporeal circuit includes an oxygenator, a heat exchanger, ite measurement of blood gases and biochemistry every 30
a centrifugal pump, and an open circuit with a reservoir minutes while on the pump.70
(Fig. 9.7).69–72

TECHNICAL VARIATIONS CLINICAL OUTCOMES


After the mandatory “no-touch” period, NRP is started The use of NRP has been associated with an increased
with a flow of 2 to 3 L/min and oxygen or air/oxygen mix is organ recovery rate compared with standard DCD retrieval
administered to maintain a PaO2 more than 12 kPa. Bicar- in controlled donation. In the UK, this was particularly
bonate is added to maintain a pH of 7.35 to 7.45, while 2 to noticeable for livers, where utilization increased from 27%
4 units of red cells may be required to maintain the hema- to over 50% with a significant reduction in the ischemic bil-
tocrit at more than 20%.69–72 There are several key differ- iary complications and reduced incidence of early allograft
ences between NRP in controlled and uncontrolled DCD dysfunction.70
donors, related to administration of heparin timing and site The effect of NRP on the outcomes of kidney transplanta-
of vascular cannulation. In the uncontrolled DCD setting, tion from DCD donors is summarized in Table 9.5.
vascular cannulation and heparinization occur during Despite a high incidence of DGF in category II DCD
a period of mechanical cardiac compression and ventila- donors Valero et al. reported an excellent 1-year survival
tion to preserve organ perfusion post mortem. Vascular of 87.5%.74 The authors also noted that NRP reduced the
134 Kidney Transplantation: Principles and Practice

TABLE 9.5 Clinical Outcomes for Kidney Transplantation Using Normothermic Regional Perfusion for Donation After
Circulatory Death Organs
Study Number of Patients Maastricht Category 1-Year Graft Survival (%) 1-Year Patient Survival (%) DGF (%) PNF (%)
Valero et al.74 16 II 87.5 90 61 4.4
Demiselle et al.75 19 II NR NR 53 5
Reznik et al.76 44 II 95.5 100 52 0
Magliocca et al.77 24 III NR NR 8.3 0
Oniscu et al.70 32 III 87.5 96.8 40 6
Rojas-Pena et al.78 48 III NR NR 31 3
Minambres et al.71 37 III 91.8 NR 27 5

DGF, delayed graft function; NR, not reported; PNF, primary nonfunction.

incidence of DGF compared with hypothermic regional


perfusion. In France, Demiselle et al. reported similar Normothermic Machine Perfusion
findings. The use of NRP led to a 27% reduction in the
incidence of DGF compared with in situ cold perfusion.75 Normothermic machine perfusion (NMP) is based on the
Furthermore, in multivariate analysis, the use of NRP was assumption that the most effective means of preserving an
associated with a significantly lower risk of DGF (odds ratio organ is to replicate its physiologic milieu. The potential
[OR] = 0.17, 95% confidence interval [CIF], 0.03–0.87). of maintaining tissue viability by isolated perfusion was
At 12 months posttransplant, in an analysis adjusted for recognized as long ago as 1812 by the physiologist Le Gal-
donor serum creatinine and age as well as recipient age lois, and its experimental feasibility was first demonstrated
and the incidence of posttransplant acute rejection, the in much cited experiments by Carrel and Lindbergh in the
use of NRP was the only predictive factor of a better renal 1930s.80,81
function (eGFR > 40 mL/min/173 m2). This study also The postulated benefits of normothermic perfusion as
suggested that the use of NRP can improve graft function a means of preservation are: (1) restoration of cellular
in uncontrolled DCD transplantation, given that these energy, repair of donation/retrieval injury, minimization
patients achieved comparable DGF and early graft func- of ischemia-reperfusion; (2) measurement of viability; (3)
tion compared with recipients of kidneys from brain-dead delivery of organ-specific therapies; and (4) prolonged stor-
donors.75 age. All four are relevant to the most pressing current clini-
The use of NRP may also allow an extension of cur- cal challenge in kidney transplantation—the need to use
rent acceptance criteria in uncontrolled DCD as sug- suboptimal (marginal) donor organs.
gested by Reznik et al.76 Four patients were transplanted The wide variation in the acceptance rates of ECD organs
with kidneys from uncontrolled DCD with 60 minutes of (e.g., ranging from 29% to 62%) within the UK illustrates
asystole and a 95% 1-year graft survival and renal func- the need for a better way to preserve and assess donor
tion comparable with standard DBD at 3 and 12 months organs.6 Current methods for assessing the transplantabil-
posttransplant. ity of a marginal donor organ, based on donor factors and
The expansion of NRP in controlled donation has been embedded in donor risk algorithms, do not provide suffi-
driven primarily by the improvement in liver transplant cient information to determine whether to discard an indi-
outcomes. However, a potential beneficial effect was vidual organ.5
also noted in the outcomes of kidney transplantation. In
the initial report from the UK, although 40% of patients
EXPERIMENTAL EVIDENCE
developed DGF, the 3- and 12-month function was com-
parable with that seen in a contemporaneous cohort of In an early proof-of-concept study, the Kootstra group
DBD recipients.70 At the same time, it has previously been tested 3 hours of warm (32°C) perfusion of canine kid-
shown by several groups, that outcomes of DCD are very neys following 30 minutes hypoxia, using a complex
good at 1 year. Lower DGF rates were also noted in reports cell-free perfusate (Exsanguineous metabolic support
from two US pilot studies.77,78 In a more recent study by [EMS], Breonics Inc., US).82 Posttransplant renal func-
Minambres et al., 37 patients received a NRP-controlled tion was improved, as was graft survival.82 Recovery
DCD kidney with a 27% DGF rate and 90.9% 1-year sur- from hypoxic injury was demonstrated by successful
vival.71 Antoine et al. compared the outcomes of 92 NRP- transplantation after 120 minutes of warm ischemia
controlled DCD renal transplants in France with 846 and 18 hours of perfusion.83 The same group later dem-
matched DBD controls and noted a significantly lower onstrated the potential for delivery of gene therapy to a
DGF (9% vs. 18%) and a comparable renal function at the perfused kidney and induction of the protective protein,
time of discharge.79 hemoxygenase-1.83,84
These reports suggest a beneficial effect of NRP in The Leicester group of Nicholson demonstrated that nor-
terms of recovery rates, but more significantly in terms mothermic perfusion with oxygenated blood was able to
of renal function, with a reconditioning effect that restore depleted ATP levels and improve renal function in
requires further confirmatory studies and mechanis- a porcine reperfusion model.85 This group demonstrated
tic evaluation. It has been advocated that NRP should the feasibility of normothermic perfusion after hypothermic
become a standard method for organ recovery from DCD preservation in a porcine renal autotransplant model86 and
in the future. the benefit of leukocyte-depleted blood.87
9 • Kidney Preservation 135

The Toronto group has studied longer periods of NMP. In targeting the therapy where it is needed. One such com-
a porcine DCD (30 minutes warm ischemia) autotransplant pound is mirococept, which is an inhibitor of complement-
model, perfusion for 8 hours immediately after retrieval mediated damage. Mirococept is currently under clinical
resulted in improved early renal function.88 The issue of trial using HMP (ISRCTN49958194).95 Another class of
whether to perfuse immediately from the time of retrieval compound that can be targeted to endothelial cell mem-
or after a period of static cold storage has important logis- branes is anticoagulant molecules; HMP has been used in
tic implications, not only for the retrieval teams, but also experimental studies to deliver anticoagulant molecules,
for the specification of the perfusion device. Using the same reducing vascular resistance on reperfusion.96,97 Experi-
porcine DCD autotransplant model, evidence showed that mental studies have also investigated propofol as an addi-
early creatinine clearance (day 3) was significantly supe- tive during machine perfusion, with some promising early
rior in organs perfused immediately; this also correlated results.98
with histologic and biomarker (neutrophil gelatinase- The delivery of mesenchymal stem cells (MSCs) during
associated lipocalin) data.89 The use of perfusion metrics to perfusion is another potential avenue to explore. It has been
predict transplant outcome has been reported in the same shown that the administration of MSCs in vivo enhances
model: acid–base and flow dynamics differed significantly recovery from ischemia–reperfusion-induced acute renal
according to the degree of ischemic injury and correlated failure in rats.99 The mechanism is not yet fully under-
with posttransplant renal function.90 stood but seems to include antiinflammatory properties
and enhanced tissue repair. The use of autologous MSCs as
an induction therapy has been studied in an RCT of living-
CLINICAL EVIDENCE
related kidney transplantation.100 This study demonstrated
There is limited clinical experience of NMP in kidney trans- a lower incidence of acute rejection, opportunistic infec-
plantation to date. The first report of a clinical implemen- tion, and a better renal function at 1 year compared with
tation described 35 minutes of NMP after 11 hours cold the IL-2 receptor antibody treatment.100 Experimental and
storage of a kidney that functioned immediately.91 A sub- animal studies in transplantation have shown that MSCs
sequent paper reported 18 extended criteria kidneys, per- delivered during perfusion might reduce ischemic damage
fused for a mean of 63 minutes after static cold storage and in retrieved kidneys.101 MePEP (Mesenchymal stem cells
compared with a nonrandomized control group of 47 static in normothermic Ex-vivo PErfusion in Pigs) is an interna-
cold-stored extended criteria kidneys.92 Of the NMP-treated tional consortium that is investigating porcine MSC ther-
kidneys 5.6% developed DGF, compared with 36.2% of con- apy. For this study, MSCs are delivered to injured kidneys
trols (p = 0.014). There was no graft survival difference. during NMP to determine whether MSCs have the potential
NMP criteria are now being tested clinically as a method reduce injury and/or trigger regeneration.102
for viability assessment. After an initial exploratory phase The delivery of genes to improve organ quality during
to establish grading criteria based on macroscopic appear- extracorporeal machine perfusion could possibly reduce
ance, urine output, and vascular flow, five organs were the recipient’s exposure to the necessary viral vectors and
transplanted on the basis of NMP criteria out of eight DCD target the treatment to the donor organ.103 This method
kidneys that had been declined because of poor in situ flush- of delivering a gene and targeting the renal vascular endo-
ing; four kidneys had immediate function.93 thelium has been tested, as has the delivery of small inter-
fering RNA (SiRNA) to block gene expression and reduce
IRI.104,105
Future Directions The effect of mild hypothermia in the deceased donor
(34°C–35°C) was starkly shown in a recent clinical trial
Increasing evidence is pointing to the benefit of NMP in the that was discontinued early because of overwhelming effi-
assessment, preservation, and reconditioning of high-risk cacy.106 Active or passive cooling of the donor reduced DGF
donor kidneys. Once the technical and logistic challenges rates from 39% to 28%.106 The Consortium for Organ Pres-
have been resolved, such as the necessary duration to pro- ervation in Europe (COPE) is conducting three clinical tri-
vide maximal benefit, data from rigorous controlled clini- als alongside translational studies to advance and develop
cal trials will be essential to establish the role of this new organ preservation technologies (Table 9.6). These clini-
technology in clinical practice. One such trial is currently cal trials accompany experimental models investigating a
underway in the UK,94 and it is likely that other studies will number of novel scientific approaches to organ repair and
drive this developing field. regeneration and the development of new objective meth-
ods to measure and predict the viability and outcome of
donated organs.
TAILORING PRESERVATION STRATEGIES
It may be the case that tailored preservation techniques
During active preservation it is possible to deliver to the require a bespoke combination of mechanisms for each
organ drugs, cells, or genes, which may recondition or stage of the organ retrieval, storage, and implantation pro-
repair damaged organs. One option may be to deliver com- cess, dependent on the characteristics of each donor, organ,
pounds that localize to endothelial cell membranes, thereby and recipient.
136 Kidney Transplantation: Principles and Practice

TABLE 9.6 Consortium for Organ Preservation in Europe (COPE) Clinical Trials
Study Primary Outcome Status and Registration
A multicenter randomized controlled trial to compare the Peak AST in 7 days after liver transplant Completed, published
efficacy of normothermic machine perfusion with static ISRCTN39731134
cold storage in human liver transplantation
Cold Oxygenated Machine Preservation of Aged Renal DCD Creatinine clearance at 1 year after kidney transplant Completed
Transplants (COMPARDT)107 ISRCTN32967929
“In house” preimplantation hypothermic machine perfusion 1-year graft survival after kidney transplant Completed
reconditioning after cold storage versus cold storage alone ISRCTN63852508
in ECD kidneys from brain-dead donors (POMP)

AST, aspartate transaminase; DCD, donation after circulatory death; ECD, expanded criteria donation.

References 19. Quiroga I, McShane P, Koo DD, et al. Major effects of delayed graft
function and cold ischaemia time on renal allograft survival. Nephrol
1. Calne RY, Pegg DE, Brown FL. Renal preservation by ice cooling: an Dial Transplant 2006;21:1689–96.
experimental study relating to kidney transplantation from cadav- 20. Ojo AO, Wolfe RA, Held PJ, Port FK, Schmouder RL. Delayed graft
ers. Br Med J 1963;2(5358):651–5. function: risk factors and implications for renal allograft survival.
2. Collins GM, Bravo-Shugarman M, Terasaki P. Kidney preservation Transplantation 1997;63(7):968–74.
for transportation: initial perfusion and 30 hours’ ice storage. Lancet 21. Levy MN. Oxygen consumption and blood flow in the hypothermic,
1969;294(7632):1219–22. perfused kidney. Am J Physiol 1959;197(5):1111–4.
3. Summers DM, Johnson RJ, Hudson A, Collett D, Watson CJ, Bradley 22. Downes G, Hoffman R, Huang J, Belzer FO. Mechanism of action
JA. Effect of donor age and cold storage time on outcome in recipients of washout solutions for kidney preservation. Transplantation
of kidneys donated after circulatory death in the UK: a cohort study. 1973;16(1):46–53.
Lancet 2013;381(9868):727–34. 23. Belzer FO, Southard JH. Principles of solid-organ preservation by
4. A definition of irreversible coma. Report of the Ad Hoc Committee of cold storage. Transplantation 1988;45(4):673–6.
the Harvard Medical School to examine the definition of brain death. 24. Belzer F, Ashby BS, Dunphy JE. 24-hour and 72-hour preservation of
JAMA 1968;205(6):337–40. canine kidneys. Lancet 1967;290(7515):536–9.
5. Watson CJ, Johnson RJ, Birch R, Collett D, Bradley JA. A simplified 25. Jamieson NV, Sundberg R, Lindell S, et al. Preservation of the canine
donor risk index for predicting outcome after deceased donor kidney liver for 24–48 hours using simple cold storage with UW solution.
transplantation. Transplantation 2012;93(3):314–8. Transplantation 1988;46(4):517–22.
6. Annual Report on Kidney Transplantation 2016/17, NHS 26. Hart NA, Leuvenink HGD, Ploeg RJ. New solutions in organ preser-
Blood and Transplant. June 2017. Available online at: htt- vation. Transplant Rev 2002;16:131–41.
ps://nhsbtdbe.blob.core.windows.net/umbraco-assets- 27. Sumimoto R, Jamieson NV, Kamada N. Examination of the role of
corp/4607/kidney-annual-report-2016-17.pdf. the impermeants lactobionate and raffinose in a modified UW solu-
7. U.S. Department of Health and Human Services, Organ Procure- tion. Transplantation 1990;50(4):573–6.
ment and Transplantation Network. Data Reports 2017 [October 28. Ramella-Virieux SG, Steghens JP, Barbieux A, Zech P, Pozet N, Hadj-
25, 2017]. Available online at: https://optn.transplant.hrsa.gov/da Aissa A. Nifedipine improves recovery function of kidneys preserved
ta/view-data-reports/. in a high-sodium, low-potassium cold-storage solution: study with
8. Snoeijs MGJ, Winkens B, Heemskerk MBA, et al. Kidney transplanta- the isolated perfused rat kidney technique. Nephrol Dial Transplant
tion from donors after cardiac death: a 25-year experience. Trans- 1997;12(3):449–55.
plantation 2010;90(10):1106–12. 29. Rauen U, de Groot H. New insights into the cellular and molecular
9. Bos EM, Leuvenink HGD, van Goor H, Ploeg RJ. Kidney grafts from mechanisms of cold storage injury. J Investig Med 2004;52(5):299–
brain dead donors: inferior quality or opportunity for improvement? 309.
Kidney Int 2007;72(7):797–805. 30. Boudjema K, van Gulik T, Lindell SL, Vreugdenhil P, Southard JH,
10. Nijboer WN, Schuurs TA, van der Hoeven JAB, et al. Effects of brain Belzer FO. Effect of oxidized and reduced glutathione in liver preser-
death on stress and inflammatory response in the human donor kid- vation. Transplantation 1990;50:948–51.
ney. Transplant Proc 2005;37(1):367–9. 31. Gores GJ, Nieminen AL, Wray BE, Herman B, Lemasters JJ. Intracel-
11. Koo DDH, Welsh KI, McLaren AJ, Roake JA, Morris PJ, Fuggle SV. lular pH during “chemical hypoxia” in cultured rat hepatocytes: pro-
Cadaver versus living donor kidneys: impact of donor factors on tection by intracellular acidosis against the onset of cell death. J Clin
antigen induction before transplantation. Kidney Int 1999;56(4): Invest 1989;83(2):386–96.
1551–9. 32. Bonventre JV, Cheung JY. Effects of metabolic acidosis on viability of
12. Pratschke J, Wilhelm MJ, Kusaka M, et al. Accelerated rejec- cells exposed to anoxia. Am J Physiol 1985;249(1):C149–59.
tion of renal allografts from brain-dead donors. Ann Surg 33. Bernardi P. Mithochondrial transport of cations: channels, exchang-
2000;232(2):263–71. ers, and permeability transition. Physiol Rev 1999;79:1127–55.
13. Chen EP, Bittner HB, Kendall SW, Van Trigt P. Hormonal and hemo- 34. Claesson K, Lindell S, Southard JH, Belzer FO. Chlorpromazine,
dynamic changes in a validated animal model of brain death. Crit quinacrine, and verapamil as donor pretreatment in liver pres-
Care Med 1996;24(8):1352–9. ervation, tested in the isolated perfused rat liver. Cryobiology
14. Bittner HB, Kendall SWH, Chen EP, Craig D, Van Trigt P. The effects 1991;28(5):422–7.
of brain death on cardiopulmonary hemodynamics and pulmonary 35. Takei Y, Marzi I, Kauffman FC, Currin RT, Lemasters JJ, Thurman
blood flow characteristics. Chest J 1995;108(5):1358–63. RG. Increase in survival time of liver transplants by protease inhibi-
15. Nagareda T, Kinoshita Y, Tanaka A, et al. Clinicopathology of kid- tors and a calcium channel blocker, nisoldipine. Transplantation
neys from brain-dead patients treated with vasopressin and epineph- 1990;50(1):14–20.
rine. Kidney Int 1993;43(6):1363–70. 36. Topp SA, Upadhya GA, Strasberg SM. Cold preservation of iso-
16. Kootstra G, Daemen JH, Oomen AP. Categories of non-heartbeating lated sinusoidal endothelial cells in MMP 9 knockout mice:
donors. Transplant Proc 1995;27:2893–4. effect on morphology and platelet adhesion. Liver Transplant
17. Thuong M, Ruiz A, Evrard P, et al. New classification of donation 2004;10:1041–8.
after circulatory death donors definitions and terminology. Transpl 37. Upadhya GA, Strasberg SM. Glutathione, lactobionate, and histi-
Int 2016;29(7):749–59. dine: cryptic inhibitors of matrix metalloproteinases contained in
18. Opelz G, Dohler B. Multicenter analysis of kidney preservation. University of Wisconsin and histidine/tryptophan/ketoglutarate
Transplantation 2007;83(3):247–53. liver preservation solutions. Hepatology 2000;31:1115–22.
9 • Kidney Preservation 137

38. Oberbauer R, Rohrmoser M, Regele H, Muhlbacher F, Mayer G. 63. Treckmann J, Minor T, Gallinat A, et al. Lipid peroxidation products
Apoptosis of tubular epithelial cells in donor kidney biopsies pre- and alpha glutathione-S-transferase in machine perfusate of older
dicts early renal allograft function. J Am Soc Nephrol 1999;10(9): donor kidneys are associated with post-transplant outcome. Am J
2006–13. Transplant 2012;12:424.
39. Gotoh K, Lu Z, Morita M, et al. Participation of autophagy in the initi- 64. Watson CJE, Wells AC, Roberts RJ, et al. Cold machine perfusion
ation of graft dysfunction after rat liver transplantation. Autophagy versus static cold storage of kidneys donated after cardiac death:
2009;5(3):351–60. a UK multicenter randomized controlled trial. Am J Transplant
40. Turkmen K, Martin J, Akcay A, et al. Apoptosis and autophagy in 2010;10(9):1991–9.
cold preservation ischemia. Transplantation 2011;91(11):1192–7. 65. O’Callaghan JM, Morgan RD, Knight SR, Morris PJ. Systematic
https://doi.org/10.097/TP.0b013e31821ab9c8. review and meta-analysis of hypothermic machine perfusion versus
41. McCord JM. Oxygen-derived free radicals in postischemic tissue static cold storage of kidney allografts on transplant outcomes. Br J
injury. N Engl J Med 1985;312(3):159–63. Surg 2013;100(8):991–1001.
42. Schachter M, Foulds S. Free radicals and the xanthine oxidase path- 66. Jiao B, Liu S, Liu H, Cheng D, Cheng Y, Liu Y. Hypothermic machine
way. In: Grace P, Mathie R, editors. Ischaemi-reperfusion injury. perfusion reduces delayed graft function and improves one-year graft
Hoboken, NJ: Blackwell Science; 1999. p. 137–56. survival of kidneys from expanded criteria donors: a meta-analysis.
43. Kosieradzki M, Kuczynska J, Piwowarska J, et al. Prognostic signifi- PLoS ONE 2013;8(12):e81826.
cance of free radicals: mediated injury occuring in the kidney donor. 67. O’Callaghan JM, Pall KT, Pengel LHM. Supplemental oxygen during
Transplantation 2003;75:1221–7. hypothermic kidney preservation: a systematic review. Transplant
44. Salahudeen AK. Cold ischemic injury of transplanted kidneys: new Rev (Orlando, FL) 2017;31(3):172–9.
insights from experimental studies. Am J Physiol 2004;287:F181–7. 68. Jochmans I, Akhtar MZ, Nasralla D, et al. Past, present, and future
45. Vreugdenhil PK, Belzer FO, Southard JH. Effect of cold storage on tis- of dynamic kidney and liver preservation and resuscitation. Am J
sue and cellular glutathione. Cryobiology 1991;28(2):143–9. Transplant 2016;16(9):2545–55.
46. Ploeg RJ, van Bockel JH, Langendijk PT, et al. Effect of preservation 69. Fondevila C, Hessheimer AJ, Ruiz A, et al. Liver transplant using
solution on results of cadaveric kidney transplantation. The Euro- donors after unexpected cardiac death: novel preservation protocol
pean Multicentre Study Group. Lancet 1992;340(8812):129–37. and acceptance criteria. Am J Transplant 2007;7(7):1849–55.
47. De Boer J, De Meester J, Smits JMA, et al. Eurotransplant randomized 70. Oniscu GC, Randle LV, Muiesan P, et al. In situ normothermic regional
multicenter kidney graft preservation study comparing HTK with perfusion for controlled donation after circulatory death—the United
UW and Euro-Collins. Transplant Int 1999;12(6):447–53. Kingdom experience. Am J Transplant 2014;14(12):2846–54.
48. Stewart ZA, Lonze BE, Warren DS, et al. Histidine-tryptophan-keto- 71. Minambres E, Suberviola B, Dominguez-Gil B, et al. Improving the
glutarate (HTK) is associated with reduced graft survival of deceased outcomes of organs obtained from controlled donation after circu-
donor kidney transplants. Am J Transplant 2009;9(5):1048–54. latory death donors using abdominal normothermic regional perfu-
49. O’Callaghan JM, Knight SR, Morgan RD, Morris PJ. Preservation sion. Am J Transplant 2017;17(8):2165–72.
solutions for static cold storage of kidney allografts: a systematic 72. Gonzalez FX, Garcia-Valdecasas JC, Lopez-Boado MA, et al. Adenine
review and meta-analysis. Am J Transplant 2012;12(4):896–906. nucleotide liver tissue concentrations from non-heart-beating donor
50. O’Callaghan JM, Knight SR, Morgan RD, Morris PJ. A national reg- pigs and organ viability after liver transplantation. Transplant Proc
istry analysis of kidney allografts preserved with Marshall’s solution 1997;29(8):3480–1.
in the United Kingdom. Transplantation 2016;100(11):2447–52. 73. Aguilar A, Alvarez-Vijande R, Capdevila S, Alcoberro J, Alcaraz A.
51. Zaouali MA, Ben Abdennebi H, Padrissa-Altes S, Mahfoudh-Boussaid Antioxidant patterns (superoxide dismutase, glutathione reductase,
A, Rosello-Catafau J. Pharmacological strategies against cold ischemia and glutathione peroxidase) in kidneys from non-heart-beating-
reperfusion injury. Expert Opin Pharmacother 2010;11(4):537–55. donors: experimental study. Transplant Proc 2007;39(1):249–52.
52. Baumert H, Faure JP, Zhang K, et al. Evidence for a mitochondrial 74. Valero R, Cabrer C, Oppenheimer F, et al. Normothermic recircu-
impact of trimetazidine during cold ischemia and reperfusion. Phar- lation reduces primary graft dysfunction of kidneys obtained from
macology 2004;71(1):25–37. non-heart-beating donors. Transplant Int 2000;13(4):303–10.
53. Faure JP, Petit I, Zhang K, et al. Protective roles of polyethylene gly- 75. Demiselle J, Augusto JF, Videcoq M, et al. Transplantation of kidneys
col and trimetazidine against cold ischemia and reperfusion injuries from uncontrolled donation after circulatory determination of death:
of pig kidney graft. Am J Transplant 2004;4(4):495–504. comparison with brain death donors with or without extended crite-
54. Durigutto P, Sblattero D, Biffi S, et al. Targeted delivery of neutral- ria and impact of normothermic regional perfusion. Transplant Int
izing Anti-C5 antibody to renal endothelium prevents complement- 2016;29(4):432–42.
dependent tissue damage. Front Immunol 2017;8:1093. 76. Reznik ON, Skvortsov AE, Reznik AO, et al. Uncontrolled donors with
55. De Vries B, Matthijsen RA, Wolfs TG, Van Bijnen AA, Heeringa P, controlled reperfusion after sixty minutes of asystole: a novel reliable
Buurman WA. Inhibition of complement factor C5 protects against resource for kidney transplantation. PLoS ONE 2013;8(5):e64209.
renal ischemia-reperfusion injury: inhibition of late apoptosis and 77. Magliocca JF, Magee JC, Rowe SA, et al. Extracorporeal support for
inflammation. Transplantation 2003;75(3):375–82. organ donation after cardiac death effectively expands the donor
56. Uhlmann D, Gaebel G, Armann B, et al. Attenuation of proinflamma- pool. J Trauma 2005;58(6):1095–101; discussion 101-2.
tory gene expression and microcirculatory disturbances by endothe- 78. Rojas-Pena A, Sall LE, Gravel MT, et al. Donation after circulatory
lin A receptor blockade after orthotopic liver transplantation in pigs. determination of death: the University of Michigan experience with
Surgery 2006;139(1):61–72. extracorporeal support. Transplantation 2014;98(3):328–34.
57. Gu M, Takada Y, Fukunaga K, et al. Pharmacologic graft protec- 79. Antoine C, Videcoq M, Riou B, et al. Controlled donation after circula-
tion without donor pretreatment in liver transplantation from non- tory death (cDCD) donors may become similar to brain death donors
heart-beating donors. Transplantation 2000;70(7):1021–5. (DBD). [abstract]. Am J Transplant 2017;17(Suppl. 3). Available
58. Lindbergh CA. An apparatus for the culture of whole organs. J Exp online at: https://atcmeetingabstracts.com/abstract/controlled-
Med 1935;62(3):409–31. donation-after-circulatory-death-cdcd-donors-may-become-
59. Belzer FO, Ashby BS, Gulyassy PF, Powell M. Successful seventeen- similar-to-brain-death-donors-dbd/.
hour preservation and transplantation of human-cadaver kidney. 80. Le Gallois CJJ. Expériences sur le Principe de la Vie: Notamment sur
N Engl J Med 1968;278(11):608–10. Celui des Mouvements du Coeur, et sur le Siége de ce Principe. Paris:
60. Moers C, Smits JM, Maathuis MHJ, et al. Machine perfusion or cold D’Hautel; 1812.
storage in deceased-donor kidney transplantation. N Engl J Med 81. Carrel A, Lindbergh CA. The culture of whole organs. Science
2009;360(1):7–19. 1935;81(2112):621–3.
61. Moers C, Jochmans I, Treckmann J, et al. Better graft survival with 82. Stubenitsky BM, Booster MH, Brasile L, Araneda D, Haisch CE, Koot-
machine perfusion than cold storage after three years: follow-up stra G. Exsanguinous metabolic support perfusion—a new strategy
analysis of the European multicentre RCT in deceased-donor kidney to improve graft function after kidney transplantation. Transplanta-
transplantation. Transplant Int 2011;24(S2):93. tion 2000;70(8):1254–8.
62. Treckmann J, Moers C, Smits J, et al. Machine perfusion versus cold 83. Brasile L, Stubenitsky BM, Booster MH, et al. Overcoming severe
storage for preservation of kidneys from expanded criteria donors renal ischemia: the role of ex vivo warm perfusion. Transplantation
after brain death. Transplant Int 2011;24(6):548–54. 2002;73(6):897–901.
138 Kidney Transplantation: Principles and Practice

84. Brasile L, Buelow R, Stubenitsky BM, Kootstra G. Induction of heme 96. Sedigh A, Larsson R, Brannstrom J, et al. Modifying the ves-
oxygenase-1 in kidneys during ex vivo warm perfusion. Transplan- sel walls in porcine kidneys during machine perfusion. J Surg Res
tation 2003;76(8):1145–9. 2014;191(2):455–62. https://doi.org/10.1016/j.jss.2014.04.006.
85. Bagul A, Hosgood SA, Kaushik M, Kay MD, Waller HL, Nicholson 97. Hamaoui K, Gowers S, Boutelle M, et al. Organ pretreatment with
ML. Experimental renal preservation by normothermic resuscitation cytotopic endothelial localising peptides to ameliorate microvas-
perfusion with autologous blood. Br J Surg 2008;95(1):111–8. cular thrombosis and perfusion deficits in ex-vivo renal haemo-
86. Hosgood SA, Barlow AD, Yates PJ, Snoeijs MGJ, van Heurn ELW, reperfusion models. Transplantation 2016;100(12):e128–39.
Nicholson ML. A pilot study assessing the feasibility of a short period 98. Snoeijs MG, Vaahtera L, de Vries EE, et al. Addition of a water-soluble
of normothermic preservation in an experimental model of non propofol formulation to preservation solution in experimental kid-
heart beating donor kidneys. J Surg Res 2011;171(1):283–90. ney transplantation. Transplantation 2011;92(3):296–302.
87. Harper S, Hosgood S, Kay M, Nicholson M. Leucocyte depletion 99. Lange C, Togel F, Ittrich H, et al. Administered mesenchymal stem
improves renal function during reperfusion using an experimen- cells enhance recovery from ischemia/reperfusion-induced acute
tal isolated haemoperfused organ preservation system. Br J Surg renal failure in rats. Kidney Int 2005;68(4):1613–7.
2006;93(5):623–9. 100. Tan J, Wu W, Xu X, et al. Induction therapy with autologous mesen-
88. Kaths JM, Echeverri J, Goldaracena N, et al. Eight-hour continu- chymal stem cells in living-related kidney transplants: a randomized
ous normothermic ex vivo kidney perfusion is a safe preservation controlled trial. JAMA 2012;307(11):1169–77.
technique for kidney transplantation: a new opportunity for the 101. Gregorini M, Corradetti V, Pattonieri EF, et al. Perfusion of isolated
storage, assessment, and repair of kidney grafts. Transplantation rat kidney with mesenchymal stromal cells/extracellular vesicles
2016;100(9):1862–70. prevents ischaemic injury. J Cell Mol Med 2017;21(12):3381–93.
89. Kaths JM, Echeverri J, Chun YM, et al. Continuous normothermic https://doi.org/10.1111/jcmm.13249.
ex vivo kidney perfusion improves graft function in donation after 102. Sierra-Parraga JM, Eijken M, Hunter J, et al. Mesenchymal stromal
circulatory death pig kidney transplantation. Transplantation cells as anti-inflammatory and regenerative mediators for donor
2017;101(4):754–63. kidneys during normothermic machine perfusion. Stem Cells Dev
90. Kaths JM, Hamar M, Echeverri J, et al. Normothermic ex vivo kidney 2017;26(16):1162–70.
perfusion for graft quality assessment prior to transplantation. Am J 103. Sandovici M, Deelman LE, de Zeeuw D, van Goor H, Henning RH.
Transplant 2018;18(3):580–9. Immune modulation and graft protection by gene therapy in kidney
91. Hosgood SA, Nicholson ML. First in man renal transplantation after transplantation. Eur J Pharmacol 2008;585(2-3):261–9.
ex vivo normothermic perfusion. Transplantation 2011;92(7):735–8. 104. Brasile L, Stubenitsky B, Booster M, Green E, Haisch C, Kootstra G.
92. Nicholson ML, Hosgood SA. Renal transplantation after ex vivo Application of exsanguineous metabolic support to human kidneys.
normothermic perfusion: the first clinical study. Am J Transplant Transplant Proc 2001;33(1–2):964–5.
2013;13(5):1246–52. 105. Zheng X, Zhang X, Feng B, et al. Gene silencing of complement C5a
93. Hosgood SA, Thompson E, Moore T, Wilson CH, Nicholson ML. receptor using siRNA for preventing ischemia/reperfusion injury.
Normothermic machine perfusion for the assessment and trans- Am J Pathol 2008;173(4):973–80.
plantation of declined human kidneys from donation after circu- 106. Niemann CU, Feiner J, Swain S, et al. Therapeutic hypothermia
latory death donors. Br J Surg 2018;105(4):388–94. https://doi. in deceased organ donors and kidney-graft function. N Engl J Med
org/10.1002/bjs.10733. 2015;373(5):405–14.
94. Hosgood SA, Saeb-Parsy K, Wilson C, Callaghan C, Collett D, 107. Nasralla D, Coussios CC, Mergental H, et al. A randomized trial
Nicholson ML. Protocol of a randomised controlled, open-label trial of normothermic preservation in liver transplantation. Nature
of ex vivo normothermic perfusion versus static cold storage in 2018;557(7703):50–6. https://doi.org/10.1038/s41586-018-0047-9.
donation after circulatory death renal transplantation. BMJ Open
2017;7(1):e012237.
95. Kassimatis T, Qasem A, Douiri A, et al. A double-blind randomised
controlled investigation into the efficacy of Mirococept (APT070)
for preventing ischaemia reperfusion injury in the kidney allograft
(EMPIRIKAL): study protocol for a randomised controlled trial. Tri-
als 2017;18(1):255.
10 Histocompatibility in Renal
Transplantation
SUSAN V. FUGGLE and CRAIG J. TAYLOR

CHAPTER OUTLINE Historical Background Complement-Dependent Lymphocytotoxic


The HLA System Crossmatch
HLA Genes and Their Products B Cell Crossmatch
HLA Class I Crossmatch Serum Sample Selection (Timing)
HLA Class II Immunoglobulin Class and Specificity
HLA on the Web Flow Cytometric Crossmatch Test
HLA Matching Organ Allocation and Pretransplant Donor
HLA-Specific Allosensitization Crossmatch Testing
HLA-Specific Antibody Detection and The Virtual Crossmatch
Characterization Immunologic Risk Stratification
Complement-Dependent Strategies for Transplanting Sensitized
Lymphocytotoxicity and Highly Sensitized Patients
Solid-Phase Binding Assays for HLA-Specific Antibody Removal
Antibody Detection and Specification Paired Exchange
Antibody Screening Strategies Posttransplant Monitoring
Patient Sensitization Profile and Definition Future Directions
of Unacceptable Specificities Concluding Remarks
Donor Crossmatch
Donor Crossmatch Techniques and Their
Clinical Relevance

Historical Background by cytotoxicity, were found to be associated with hyper-


acute rejection (HAR).3,4 HLA was quickly recognized
The study of histocompatibility accelerated during the as the human equivalent of the major histocompatibility
1960s when the pioneers of clinical kidney transplantation complex (MHC), previously identified in inbred rodents, the
recognized that graft destruction was mediated through products of which control the recognition of self and foreign
immunologic mechanisms. In 1961 the introduction of antigens.5
chemical immunosuppression, first 6-mercaptopurine fol-
lowed soon after by azathioprine and steroids, enabled
short and medium-term success. However, 40% to 50% of The HLA System
deceased donor transplants were lost because of immediate
or early graft failure due to irreversible rejection in the first The HLA system encoded on the short arm of chromo-
year and thereafter there was an insidious decline in graft some 6 is the most intensively studied region of the human
function. These early experiences severely limited the suc- genome. The region spans over four megabases and con-
cess of human allotransplantation and led to the study of tains in excess of 250 expressed genes, making it the most
compatibility of transplanted tissue, which, over the next gene-dense region characterized to date.6 Approximately
40 years gave rise to the specialty of “histocompatibility 28% of these genes encode proteins with immune-related
and immunogenetics” (H&I). functions, making the region of particular relevance to
The first human leukocyte antigens (HLA) were discov- transplant clinicians and immunologists.
ered in 1958 and subsequent years by Jean Dausset, Rose HLA has a central role in immune recognition for defense
Payne, and Jon van Rood.1 Later, many more HLA antigens against foreign pathogens and neoplasia, mediating T cell
were characterized using antibodies in sera obtained from signaling through the presentation of self and foreign anti-
multiparous women and from patients after multiple blood gens in the form of short protein fragments (peptides) recog-
transfusions. Such antibodies were also demonstrated in nized by self-HLA restricted T lymphocytes (see Chapter 2).
recipient sera after transplant rejection,2 and antibod- Recognition of nonself peptides in the context of self-HLA
ies reactive with donor lymphocytes present before renal (i.e., altered self) is the function of the T cell antigen recep-
transplantation, either detected by leukoagglutination or tor and elicits a powerful immune response. The extensive
139
140 Kidney Transplantation: Principles and Practice

polymorphism of HLA has evolved to enable efficient bind- HLA Class I


ing of peptides from the vast array of potentially pathogenic HLA class I genes span two megabases at the telomeric end
organisms that invade and colonize our bodies. Therefore of the 6p21.3 region of chromosome 6. This region encodes
the evolutionary pressures to develop and maintain diver- the classical “transplantation antigens” (HLA-A, -B, and
sity vary with time and geographic area. As a consequence, -C) that are expressed on virtually all nucleated cells.7
HLA has adapted differently according to geographic region Genes of the HLA class I loci encode the 44 kD heavy chains
and ethnic group, and HLA phenotypes differ across popu- which associate with intracellular peptides present within
lations throughout the world. the cytoplasm. The tertiary structure is stabilized on the cell
surface by noncovalent association with β2-microglobulin,
HLA GENES AND THEIR PRODUCTS a nonpolymorphic 12 kD protein encoded on chromo-
some 15. The heavy chain consists of three extracellular
The HLA system is a complex multigene family consisting immunoglobulin-like domains (α1, α2, α3), a hydrophobic
of more than 10 loci. HLA types are codominantly inherited transmembrane region, and a cytoplasmic tail. The two
on a maternal and paternal haplotype and transmitted as a extracellular domains distal to the cell membrane (α1 and
single Mendelian trait (Fig. 10.1), and therefore an individ- α2) are highly polymorphic and fold to form a peptide-bind-
ual can express two alleles at each locus. The genes encod- ing cleft consisting of eight strands forming an antiparallel
ing HLA and their corresponding glycoprotein products are beta pleated sheet, overlaid by two alpha helices. The cleft
divided into two classes according to their biochemical and accommodates peptides of 8 to 10 amino acids in length,
functional properties: HLA class I and HLA class II. mostly derived from “endogenous” proteins present within

-F
-G
C -H
L -K
A -A
S -J
25 25 S -L
I -E
24 24
-C
23 23 -B
22.3 22.3
22.2 22.2
22.1 22.1 -LTA
-TNF
21.3 21.3 C -LTB
p p
21.2 21.2 L -Hsp70
A
21.1 21.1 S -C2
S -Bf
12 12
III -C4A
11.2 11.2 -C4B
11.1 11.1
-CYP21B
11 11
12 12
13 13
14 14 -DRA
-DRB4
15 15 -DRB5
16.1 16.1
16.2 16.2 -DRB3
16.3 16.3 -DRB1
-DQA1
21 21
-DQB1
q q C
22.1 22.1 L -DQB3
22.2 22.2 A -DQA2
S -DQB2
22.3 22.3 S -DOB
23.2 23.1 23.2 23.1 II -DMB
23.3 23.3 -DMA
24 24 -DOA
-DPA1
25.2 25.1 25.2 25.1 -DPB1
25.3 25.3 -DPA2
26 26 -DPB2
27 27

Fig. 10.1 Genomic organization of the HLA region on chromosome 6. HLA antigens are codominantly inherited en bloc as a haplotype from mater-
nal and paternal chromosomes. Red: pseudogenes. Green: “nonclassical” HLA genes with no known role in clinical solid-organ transplantation. Blue:
genes encoding HLA products with clinical relevance to solid-organ transplantation.
10 • Histocompatibility in Renal Transplantation 141

the cell cytoplasm. The major areas of amino acid polymor- TABLE 10.1 HLA Genes and Their Products
phism line the sides and base of the cleft and thereby gov-
ern the peptide-binding repertoire of the HLA molecule. In Name Molecular Characteristics
contrast, the α3 domain (proximal to the cell membrane) is HLA-A Class I α-chain
highly conserved and acts as a ligand for CD8 expressed on HLA-B Class I α-chain
T lymphocytes. This interaction confers HLA class I restric- HLA-C Class I α-chain
HLA-E Associated with Class I 6.2kB Hind III fragment
tion on CD8 positive T lymphocytes, which have a predomi- HLA-F Associated with Class I 5.4kB Hind III fragment
nantly cytotoxic function and form the basis for cellular HLA-G Associated with Class I 6.0kB Hind III fragment
immunity to intracellular pathogens such as viruses. HLA-H Class I pseudogene
There are other class I loci and knowledge about their HLA-J Class I pseudogene
expression and function has emerged (Table 10.1). HLA- HLA-K Class I pseudogene
HLA-L Class I pseudogene
H, -J, -K, and -L are pseudogenes and HLA-N, -P, -S, -T, HLA-N Class I gene fragment
-U, -V, -W, -X, -Y, and -Z are gene fragments that are not HLA-P Class I gene fragment
transcribed or translated. HLA-G is expressed on placen- HLA-S Class I gene fragment
tal trophoblast cells, implicating a possible involvement HLA-T Class I gene fragment
HLA-U Class I gene fragment
in fetal–maternal development. HLA-E, -F, and -G have HLA-V Class I gene fragment
limited polymorphism and are known to act as ligands for HLA-W Class I gene fragment
natural killer (NK) cell inhibitory receptors (e.g., CD94). HLA-X Class I gene fragment
These loci may prove to be important in certain experimen- HLA-Y Class I gene fragment
tal xenograft models and in bone marrow transplantation HLA-Z Class I gene fragment (located in the HLA
class II region)
(where NK cells are involved in the rejection process), but HLA-DRA DR α-chain
their clinical relevance in solid-organ transplantation has HLA-DRB1 DR β1-chain determining specificities DR1 to
not been firmly established. There is, however, an emerging DR18
role for these molecules in innate immunity to persistent HLA-DRB2 Pseudogene with DR β-like sequences
HLA-DRB3 DR β3-chain determining DR52 found on
viruses such as cytomegalovirus (CMV) and they may prove DR17, DR18, DR11, DR12, DR13, and DR14
to have an important role in post transplant viral defense. haplotypes
HLA-DRB4 DR β4-chain determining DR53 found on DR4,
HLA Class II DR7, DR9 haplotypes
The HLA class II region consists of three main loci: HLA- HLA-DRB5 DR β5-chain determining DR51 found on
DR15 and DR16 haplotypes
DR, -DQ, and -DP. The glycoprotein products are heterodi- HLA-DRB6 DRB pseudogene found on DR1, DR2, and
mers with noncovalently associated alpha and beta chains DR10 haplotypes
of molecular weight approximately 33 and 28 kD, respec- HLA-DRB7 DRB pseudogene found on DR4, DR7, and
tively. Both chains consist of two extracellular immuno- DR9 haplotypes
HLA-DRB8 DRB pseudogene found on DR4, DR7, and
globulin-like domains, a transmembrane region and a DR9 haplotypes
cytoplasmic tail. The membrane distal domains α1 and β1 HLA-DRB9 DRB pseudogene, probably found on all
form a peptide-binding cleft similar to, but less rigid than haplotypes
that of HLA class I, accommodating peptides of 10 to 20 HLA-DQA1 DQ α-chain
amino acids derived predominantly from ingested (endocy- HLA-DQB1 DQ β-chain determining specificities DQ1 to
DQ9
tosed or phagocytosed) extracellular (exogenous) proteins. HLA-DQA2 DQ α-chain-related sequence, not known to
The β1 domains of HLA-DR, -DQ, and -DP are highly poly- be expressed
morphic and govern the peptide-binding repertoire. They HLA-DQB2 DQ β-chain-related sequence, not known to
are constitutively expressed on cells with immune function be expressed
HLA-DQB3 DQ β-chain-related sequence, not known to
such as B lymphocytes, activated T lymphocytes and anti- be expressed
gen-presenting cells (monocytes, macrophages, and cells of HLA-DOA DO α-chain
dendritic lineage). HLA class II expression can be induced HLA-DOB DO β-chain
on most cell types during inflammatory responses (includ- HLA-DMA DM α-chain
ing allograft rejection) by cytokines such as gamma-inter- HLA-DMB DM β-chain
HLA-DPA1 DP α-chain
feron and tumor necrosis factor alpha.8–10 The conserved HLA-DPB1 DP β-chain
membrane proximal domain associates with CD4 on T lym- HLA-DPA2 DP α-chain-related pseudogene
phocytes with predominately helper/inducer function and HLA-DPB2 DP β-chain-related pseudogene
thereby confers HLA class II restriction and forms the basis HLA-DPA3 DP α-chain-related pseudogene
of cellular and humoral immunity to circulating pathogens
such as bacteria.
19 HLA-DR specificities defined by serologic methods, com-
HLA Polymorphism and Nomenclature. Early inves- pared with more than 2000 HLA-DRB sequence variants
tigations into HLA polymorphism used relatively crude (alleles) detected using DNA-based typing methods. The
alloantisera able to distinguish only a limited number of number of newly defined alleles identified is still increasing
antigens. Nearly half a century later, these simple tech- and has now surpassed even the highest expectations of the
niques have been superseded by molecular methods capa- early pioneers.
ble of resolving HLA variants at the DNA sequence level and Concomitant with the ever-increasing complexity of
identifying single amino acid polymorphisms that are indis- the HLA region, a nomenclature system has been devel-
tinguishable by serology. For example, there are currently oped to accurately assign HLA loci and their alleles.11 This
142 Kidney Transplantation: Principles and Practice

nomenclature system encompasses the methodology (serol- TABLE 10.2 Resolution of HLA Typing Methods and
ogy, biochemistry, and DNA sequencing) and level to which Their Application to Renal Transplantation
the HLA genes and their products have been resolved. The
nomenclature is complex and, to those outside the field, can HLA Typing Resolution Method
appear confusing. HLA allele matching High-resolution (2-field) DNA
sequence-based typinga
Resolution of HLA Typing Methods. Serologically based Split HLA specificity matching Serology and low resolution
(generic) DNA typingb
HLA typing uses alloantisera and monoclonal antibodies Broad HLA specificity matching Serology and low resolution
that bind to tertiary epitopes of the cell surface HLA glyco- (generic) DNA typing
proteins. There is a high degree of sequence homology be- HLA-B, -DR matching Serology and low resolution
tween HLA specificities and identical amino acid sequence (generic) DNA typing
Epitope matching Serologically defined cross-reac-
motifs (epitopes) are often shared between groups of anti- tive groups
gens.12–14 Serologically defined motifs/
The degree of HLA compatibility between transplant determinants
donors and recipients can be considered at many different Single amino acid residues
levels of resolution, depending on the HLA typing meth- Linear peptides and conforma-
tional epitopes
odology (Table 10.2). This can range from matching for Supertypic antigen matching
serologically defined epitopes to matching for single amino Triplet/Eplet amino acid mis-
acid differences detected through high-resolution DNA matches (HLAMatchmaker)
sequence-based methods (allele matching). aHigh-resolution DNA typing can be translated into low-resolution serologic
equivalents (allele families).
World Health Organization (WHO) Nomenclature for bLow resolution HLA typing by PCR utilizes DNA primers designed to identify
HLA. HLA genes and their polymorphic products have now polymorphisms at a level comparable to serology.
been characterized and cloned and have been given official Adapted from Taylor CJ, Dyer PA. Maximizing the benefits of HLA matching
designations using the following principles. The genes are for renal transplantation: alleles, specificities, CREGs, epitopes or residues?
Transplantation 1999;68:1093–4.
prefixed by the letters HLA followed by the loci or region,
for example, HLA-A, HLA-B, or HLA-D. The HLA-D region
has several subregions denoted HLA-DR, -DQ, -DP, -DO, and but the level of expression has not been confirmed for the
-DM (see Fig. 10.1). These are followed by the letters A or B to particular allele.
define the gene encoding the alpha and beta chain gene prod- The HLA-DR and HLA-DP alpha chains have only lim-
uct of that subregion, respectively (e.g., HLA-DRB genes code ited polymorphism and therefore the HLA-DRB1 or -DPB1
for the DRβchain protein product). Where there is more than allele (which code for the main polymorphic amino acid
one A or B gene within a subregion, a corresponding number determinants present on the beta chain) is usually anno-
is given (e.g., HLA-DRB1; see Fig. 10.1 and Table 10.1). tated alone. In contrast both the HLA-DQ alpha and beta
A new nomenclature system was launched by the chains are polymorphic. To describe one of these alleles pre-
WHO in 2010 to accommodate the growing number of cisely, definition of both the A and B alleles may be required
HLA alleles being discovered.11 In this system each allele (e.g., HLA-DQA1*01:01 and DQB1*05:01). Although the
is uniquely identified by up to four sets of digits separated alpha and beta chain protein products of the A and B gene
by colons (termed fields), prefixed by an asterisk (*). The pairs associate preferentially, there is also the possibility of
digits in the first field usually correlate with the serologic the formation of novel hybrid molecules. A complete list of
specificity, for instance HLA-B*27 correlates with the sero- recognized HLA genes and their expressed products can be
logic specificity HLA-B27. However, for most serologically found at www.bmdw.org (Bone Marrow Donors World-
defined antigens there is further polymorphism detectable wide; HLA information).
at the DNA and amino acid sequence level. The second field
denotes the subtype, listed in the order the alleles were dis- Extended HLA Haplotypes. The HLA region displays
covered. Alleles with different numbers at this level have strong linkage disequilibrium whereby certain HLA alleles
nucleotide substitutions that alter the amino acid sequence are inherited together as a conserved HLA haplotype.
(e.g., HLA-B*27:01, HLA-B*27:02) and so forth. The third Therefore extended HLA haplotypes involving HLA-A,
field indicates synonymous substitutions within the coding -B, -C, -DR, and -DQ commonly exist within and between
region (i.e., there is no change the amino acid sequence of ethnic groups. This greatly improves the probability of
the expressed protein) and the fourth field indicates poly- finding an HLA-matched unrelated donor as common
morphism in noncoding regions. HLA haplotypes are frequently found within a population
Some alleles or genes contain sequence defects prevent- (e.g., HLA-A*01:01, -B*08:01, -C*07:01, -DRB1*03:01,
ing normal antigen expression at the cell surface. Nonex- -DRB3*01:01, -DQA1*05:01, -DQB1*02:01).15 There
pressed alleles (null alleles) are indicated using the suffix is only relatively weak linkage centromeric to HLA-DQ
“N” (e.g., HLA-DRB4*01:03:01N) whereas alleles with low because of a recombination “hot-spot” between HLA-DQ
expression or soluble (secreted) alleles carry the suffix “L” or and -DP.
“S,” respectively. The suffix “C” indicates a protein detected
within the cytoplasm and not on the cell surface; “A” aber- HLA on the Web
rant expression, where there is doubt whether the protein is Information concerning the HLA system is rapidly expand-
expressed; and “Q,” questionable expression, where a given ing and articles such as this are always out of date by the
mutation has been previously shown to affect expression, time they go to print. However, there are a number of Internet
10 • Histocompatibility in Renal Transplantation 143

websites with useful links that are regularly updated. These (death with function treated as failure). The best transplant
provide contemporary articles and information concerning survival was achieved in transplants with no mismatches
HLA genes, nomenclature, polymorphism, DNA and amino at HLA-A, -B, and -DR (“000” HLA-A, -B, -DR mismatch
acid sequences for both lay and professional readers: grade). Other well-matched transplants, termed “favor-
  
   ably matched transplants,” with a maximum of one HLA-A
www.bmdw.org and one HLA-B antigen mismatched in the absence of mis-
www.ashi-hla.org/index.htm matches at HLA-DR (110, 100, 010 mismatch grades) had
www.bshi.org.uk a significantly improved survival over transplants of all
www.efiweb.org other match grades.26,27 An analysis of factors influencing
www.anthonynolan.org.uk the long-term outcome of these transplants revealed that
www.sanger.ac.uk for patients with transplants functioning after 6 years, only
www.epregistry.com.br older donor age and diabetes had a significant detrimental
  
influence on survival.
The influence of HLA mismatch on outcome of first
HLA Matching deceased donor transplant was investigated in a cohort of
patients, transplanted in the UK between 1995 and 2001.
It was more than 40 years ago that HLA matching between As a result of the allocation policy the recent transplants
donor and recipient was found to be associated with bet- were significantly better matched than the previously
ter transplant and patient survival.2,16–19 Matching for analyzed cohort (1986–1993), where 46% transplants
the class I HLA-A and -B antigens influenced survival, but were 0-DR mismatched and 10% had 2-DR mismatches,
matching for the class II HLA-DR antigens was shown to compared with 60% 0-DR mismatched and only 3% 2-DR
have the most powerful effect.19,20 A beneficial effect of HLA mismatches in the 1995 to 2001 cohort. In a multivariate
matching on graft survival was demonstrated in analyses of analysis there was no effect of HLA-A mismatching, but a
large data sets and national and international databases.21 significant effect of two mismatches at HLA-B and an incre-
Over the years there has been an overall improvement in mental effect of mismatching at HLA-DR.28 In recent mul-
transplant survival and a decrease in the survival advantage tivariate analyses of the outcome of 7837 adult patients
conferred by HLA matching.16,21 The improvement can be transplanted in the UK between 2009 to 2014, there is still
attributed to a number of factors, but one of the most power- a significant beneficial effect of HLA matching on graft out-
ful is advancements in the effectiveness of immunosuppres- come at 1 and 5 years posttransplant.
sion. This was clearly demonstrated in a local comparison There have been several publications from the Collab-
of transplant survival in patients receiving azathioprine orative Transplant Study revealing associations between
and prednisolone, cyclosporine and prednisolone, and HLA mismatching and clinical events posttransplant.
triple therapy (cyclosporine, azathioprine, prednisolone) Increasing numbers of HLA mismatches were shown to be
where 1-year transplant survival rates were 65%, 69%, significantly associated with an increased requirement for
and 81%, respectively.22 In this analysis HLA-DR compat- antirejection treatment29 and with the cumulative inci-
ibility still had a marked effect on the posttransplant clinical dence of death with a functioning graft resulting from car-
course, with an increased incidence of rejection in HLA-DR diovascular disease or infection, but not from malignant
mismatched grafts, the socioeconomic effects of which were neoplasm.30 The number of HLA-DR mismatches was
increased use of immunosuppressive drugs, longer hospital shown to be associated with increased incidence of non-
stays, and higher 3-month creatinine levels.23 Hodgkin’s lymphoma31,32 and hip fractures.33 It is pos-
Despite further improvements in immunosuppres- sible that these associations result from the higher levels
sion and patient management, a beneficial effect of HLA of immunosuppression used in the management of poorly
matching is still apparent in data from recent transplants HLA-matched transplants.
published from the Collaborative Transplant Study24 In solid-organ transplantation the effects of HLA match-
(Fig. 10.2) and from the United Network of Organ Shar- ing reported are generally based on matching at the HLA-A,
ing (UNOS). The UNOS data revealed a linear relationship -B, and -DR loci. The effect of matching other HLA loci has
between mismatches at HLA-A, -B, -DR, and allograft sur- been analyzed, but demonstrating an independent effect is
vival, independent of HLA locus, in 189,141 transplants difficult because of linkage disequilibrium.
performed between 1987 and 2013. Although the trans- Matching for HLA-DQ has been variously reported as
plants analyzed were performed over a long time period, having either a beneficial effect34 or no effect on transplant
the HLA effect was present in the transplants performed in outcome35,36 However, in a recent analysis of data from
recent periods.25 the Australia and New Zealand Dialysis and Transplant
In analyzing the effect of HLA on transplant outcome, Registry, HLA-DQ mismatches were found to be associ-
it is important that other factors known to have a strong ated with an increased incidence of acute rejection, inde-
influence on outcome are taken in account. In a rigorous pendent of HLA-A, -B, -DR mismatches. This effect was
multivariate analysis of factors influencing the outcome of further increased when donors and recipients were mis-
primary deceased donor transplants in a cohort of trans- matched for HLA-DR.37 Registry analysis has shown that
plants performed in the UK between 1986 and 1993, the HLA-DPB matching has an effect on the transplant survival
year of transplant, donor and recipient age, waiting time of regrafts, but not of first transplants.38 It is possible that
to transplant, diabetes in the recipient, donor cause of this effect results from matching for certain immunogenic
death, exchange of kidneys, cold ischemia time, and HLA HLA-DPB epitopes39 and undetected immunologic priming
mismatching were found to influence transplant survival against donor-DP mismatches of the failed graft.
144 Kidney Transplantation: Principles and Practice

HLA-A+B+DR Mismatches
Deceased Donor, First Kidney Transplants 2008–2016

100

95

90 0 MM n = 2,934
1 MM n = 3,517

Graft survival (%)


85 2 MM n = 10,005
3 MM n = 15,564
80 4 MM n = 15,241
5 MM n = 9,344
75 6 MM n = 3,155
70

65

0
0 1 2 3
A Post-transplant time (years)

Collaborative Transplant Study K-21121-0218

HLA-DR Mismatches
Deceased Donor, First Kidney Transplants 2008–2016

100

95

90
Graft survival (%)

85 0 MM n = 22,044
1 MM n = 28,738
80 2 MM n = 9,138
75

70

65

0
0 1 2 3
B Post-transplant time (years)

Collaborative Transplant Study K-21641-0218

Fig. 10.2 The influence of human leukocyte antigen (HLA)-A, -B, -DR mismatch on 3-year graft survival. (A) HLA-A+B+DR mismatches, deceased donor, first
kidney transplants, 2008 to 2016. (B) HLA-DR deceased donor, first kidney transplants, 2008 to 2016. (Reproduced from the Collaborative Transplant Study.)

The definition of an HLA match between donors and HLA-Specific Allosensitization


recipients can be considered at different levels depending
on the resolution of the HLA type and consideration of The single most important function of an H&I laboratory
areas of similarity and differences in the HLA molecules. is to detect recipient immunologic priming against foreign
The influence of compatibility for the broad serologic speci- HLA (allosensitization) and to avoid transplantation of
ficities, serologic split specificities, epitopes common within recipients with preexisting donor HLA-specific antibodies
serologic cross reactive groups (CREGs), single amino acid (DSA). After an encounter with HLA alloantigens, naïve
differences, or epitopes has been considered in deceased T and B lymphocytes elicit immune effector functions
donor renal transplantation,40 and matching at these vari- to isolate and destroy allogeneic tissue. In the context of
ous levels has been reported to be associated with improved transplantation, however, calcineurin-based immunosup-
outcomes.41–44 pressive regimes are effective in blocking naïve T and B cell
There is now considerable interest in epitopes on the HLA activation and, in the modern era, early allograft loss in
molecules and the opportunity to reduce the immunogenic- nonsensitized recipients due to irreversible allograft rejec-
ity of the transplant by matching donor and recipient for tion is uncommon. In contrast, memory T and B cells that
immunodominant epitopes, with the aim of reducing the have been primed by previous exposure to foreign HLA are
potential for the immune response and the development of often resistant to conventional immunosuppressive therapy
donor-specific HLA antibodies after transplantation.45,46 and are likely to cause irreversible cellular and/or humoral
This topic will be covered in a later section. allograft rejection. Although HLA matching is effective in
10 • Histocompatibility in Renal Transplantation 145

avoiding transplantation between “incompatible” allosen- COMPLEMENT-DEPENDENT


sitized donor–recipient pairs, because of the high level of LYMPHOCYTOTOXICITY
heterogeneity between HLA types within and between pop-
ulations, most organ transplants between genetically unre- The complement-dependent lymphocytotoxicity (CDC) test
lated individuals are mismatched at one or more HLA loci. was the first technique applied routinely for HLA-specific
HLA-specific allosensitization can occur at both the T antibody screening; it is also used for the donor lymphocyte
cell (cellular) and B cell (humoral) level but current tech- crossmatch test. In this assay, lymphocyte target cells are
nology to detect alloreactive memory T and B cells is lim- used to detect complement-fixing IgM and IgG antibodies
ited to translational research in specialist laboratories.47,48 present in patient’s serum, as indicated by cell lysis after the
In the absence of large scale routinely applicable assays to addition of rabbit complement (Fig. 10.3). IgM antibodies
detect cellular sensitization, allosensitization is determined can be differentiated from IgG antibodies by pretreatment
indirectly by the presence of circulating IgG HLA-specific of patient serum with dithiothreitol (DTT) that reduces the
antibodies in recipient serum.49 HLA-specific IgG is secreted disulfide bonds in the IgM pentamer, but leaves IgG rela-
by long-lived bone marrow resident plasma cells that are tively intact. Patient serum is tested against lymphocyte
formed after B cell/alloantigen engagement and cognate panels obtained from volunteer blood donors that can either
interaction with CD4 (helper) T cells. This results in B be “random” or alternatively “selected” to represent the
cell maturation, clonal expansion, immunoglobulin class HLA types in the potential organ donor population (termed
switch (from IgM to IgG), and the formation of antigen-spe- panel reactive antibodies; PRA). The CDC assay is used to indi-
cific memory T and B cells. On repeat exposure to the same cate the presence (and provide limited specificity analysis)
antigen, memory T and B cells elicit a rapid and aggressive of recipient lymphocytotoxic antibodies with potential to
immune response with potential to cause irrevocable graft cause a positive donor lymphocyte crossmatch.
damage. Therefore circulating IgG HLA-specific antibodies There are a number of limitations of the CDC technique.
are an effective but incomplete surrogate for detecting allo- Only complement-fixing antibodies are detected, viable
reactive T and/or B cell memory. donor lymphocytes are required, and the assay sensitiv-
The most common route of patient allosensitization is by ity is dependent on the particular batch of rabbit comple-
exposure to foreign (nonself) HLA after blood transfusion, ment used. Characterization of complex antibody profiles
pregnancy, and/or a previous allograft. Approximately 20% in patient serum is limited, and both HLA and non-HLA
of pregnant women produce HLA-specific antibodies to pater- lymphocytotoxic antibodies are detected. Although the
nally inherited fetal HLA antigens and pregnancy followed use of DTT can differentiate IgM from IgG antibodies, this
by blood transfusion is a potent stimulus for the formation of does not indicate the specificity of the antibody and poten-
high-level and broadly reactive IgG HLA-specific antibodies. tially clinically relevant weak IgG HLA-specific antibodies
Similarly, HLA-specific antibody formation is common after may also be rendered negative after the addition of DTT to
a failed allograft, and many such patients become “highly patient serum. Reactivity resulting from an IgM HLA-spe-
sensitized” with antibodies that react to HLA alloantigens cific antibody is indistinguishable from reactivity of an IgM
present in the majority of the potential donor population. The non-HLA-specific (often auto-reactive) antibody. However,
extent of sensitization in patients awaiting a second or subse- such antibodies are frequently weak or nonreactive with
quent transplant is related to the number of donor-recipient lymphocytes from patients with B cell chronic lymphatic
HLA mismatches and continuation or withdrawal of main- leukemia (CLL), and therefore including these cells in the
tenance immunosuppression after graft failure and return to screening panel can be useful in elucidating a patient’s anti-
the transplant waiting list.50,51 Therefore, to avoid the devel- body profile.54
opment of high levels of sensitization, clinical management There have been a number of approaches used to increase
of patients with a failing graft should include consideration the sensitivity and specificity of the CDC test to detect low-
of the risks associated with maintaining immunosuppression level antibodies that are potentially harmful. These include
together with future options and a timescale for repeat trans- increasing the incubation times, the wash (Amos) technique
plantation.52 In addition to these classical routes of allosensi- and augmentation with antihuman globulin (AHG). In the
tization there is increasing awareness of naturally occurring Amos technique, unbound antibody is washed from the cell
HLA-specific antibodies that may result from cross-reactivity suspension before the addition of rabbit complement, thus
with infectious agents or autologous tissue damage. Such removing the anticomplementary factors and low-affinity
“idiopathic” antibodies are reactive with specific epitopes IgM antibodies in the serum to preferentially detect clini-
expressed on denatured HLA proteins (considered clinically cally relevant IgG antibodies. In the AHG augmentation
benign) or native (conformationally folded) HLA proteins CDC test, antikappa light chain is added to the washed cells
that have potential to cause graft rejection.53 before the addition of complement, thus increasing assay
sensitivity.
HLA-SPECIFIC ANTIBODY DETECTION AND
CHARACTERIZATION SOLID-PHASE BINDING ASSAYS FOR HLA-
SPECIFIC ANTIBODY DETECTION AND
Over the past 15 years new technologies for the detection SPECIFICATION
and characterization of HLA-specific antibodies have been
developed enabling a more accurate assessment of complex It is now recognized that humoral rejection caused by HLA
antibody populations present in patient serum and compre- class I- and/or class II-specific antibodies present at levels
hensive elucidation of a patient’s sensitization profile. The below the detection threshold of the CDC assay is a major
available technologies are outlined next. cause of allograft rejection and year-on-year graft attrition.
146 Kidney Transplantation: Principles and Practice

Panel/donor Recipient 1 serum Viable cells


lymphocytes (negative crossmatch)

Complement

Recipient 2 serum Cell lysis


(positive crossmatch)

Complement

B
Fig. 10.3 (A) Lymphocytotoxic crossmatch test: Panel or donor lymphocytes are incubated with recipient serum in the wells of a microtiter (Terasaki)
tray, followed by the addition of rabbit complement. After a second incubation period vital stains (e.g., acridine orange and ethidium bromide) are
added and the wells are viewed using fluorescent (ultraviolet) microscopy to determine cell viability. (B) Lymphocytotoxic (CDC) crossmatch results.
Panel A: viable lymphocytes take up acridine orange and appear a yellow-orange color (negative crossmatch). Panel B: lysed cells (have pores in the
lymphocyte cell membrane caused by antibody binding and complement activation) take up ethidium bromide and appear a brown color (positive
crossmatch).

This stimulated the development of new technology (solid- antibody binding is detected using a fluorescent-labeled
phase binding assays; SPA) that provide increased test antihuman-globulin antibody. Luminex single-antigen
sensitivity (to detect low-level antibodies) and specific- beads (SAB) enable rapid and simultaneous elucidation
ity (to identify antibodies likely to damage a transplanted of multiple antibody populations in a patient’s serum and,
organ).55 Various commercially available SPA kits are all for the first time, facilitates accurate specification of com-
based on the common principle—incubation of patient plex antibody profiles in highly sensitized patients.56,57
serum with purified HLA protein bound to a polystyrene Although commercially available Luminex-based HLA-
surface and detection of HLA-specific antibody binding specific antibody detection and specification kits are
using a fluorochrome- or enzyme-labeled antihuman IgG currently licensed for qualitative use only, the semiquan-
conjugate. titative readout (Median Fluorescence Intensity; MFI) has
The first SPA techniques used for HLA-specific anti- been used to indicate antibody levels that form an impor-
body detection and specification were enzyme-linked tant function in informing a pretransplant immunologic
immunosorbent assay (ELISA) and flow cytometry, but risk assessment. However, the Luminex assay is subject
more recently these have been largely superseded by to a number of in vitro artifacts, and interpretation of
“Luminex” technology that uses purified HLA proteins MFI values requires caution and expert knowledge. SAB
bound to polystyrene microbeads (Fig. 10.4). The micro- express variable levels of denatured HLA to which anti-
beads are impregnated with two fluorescent dyes in differ- body binding is common, giving rise to a false-positive
ent ratios to enable differentiation of up to 100 different DSA. In addition, inhibitory factors in patient sera may
bead populations using a dedicated dual-laser flow cytom- block HLA-specific antibody detection and give rise to a
eter (Luminex platform). Each bead population is coated misleadingly low assessment of DSA. Such inhibition,
with purified HLA proteins of multiple or single HLA class however, is easily overcome by serum dilution, heat inac-
I or HLA class II alleles which allow comprehensive detec- tivation, or the addition of EDTA.58,59
tion and specification of HLA-A, -B, -C, and -DR, -DQ, and Furthermore, Luminex SAB screening enables real-time
-DP specific antibodies, respectively. The antigen-coated posttransplant antibody monitoring of DSA to support the
beads are incubated with patient serum and HLA-specific diagnosis and response to treatment for antibody-mediated
10 • Histocompatibility in Renal Transplantation 147

Flow cytometry/Luminex

2. Patient serum 3. Labeled secondary antibody (e.g.,


containing antigen- FITC conjugated anti-human IgG) to
specific alloantibody detect alloantibody binding
1. Antigen-coated micro-
particle or plastic surface

ELISA
A

B
Fig. 10.4 (A) Schematic representation of antibody screening using Luminex solid-phase binding assay: (1) Purified HLA proteins (either pooled HLA
specificities or single-antigen specificities) coated onto a solid phase (e.g., microparticles [Flow cytometry/Luminex]) are incubated with patient serum;
(2) HLA-specific antibodies bind to the antigen coated beads and nonspecific antibodies are washed off; (3) IgG HLA-specific antibodies bound to the
antigen coated beads are detected using a conjugated (e.g., FITC antihuman IgG) secondary antibody and fluorescent signal under excitation by a laser.
(B) An example of HLA-specific antibody specification using Luminex Single Antigen beads, illustrating binding to a common HLA epitope (HLA-Bw4)
present on certain HLA-B antigens and Bw4-expressing HLA-A antigens (HLA-A23, A24, A25, A32).

rejection.55 These assays are more sensitive than CDC, strategy but may use different approaches and technolo-
and they primarily detect IgG, but can also be modified gies. One common strategy is to perform an initial screen
to detect IgM60 and complement fixing (C1q/C4a/C3d) to detect the presence of HLA class I and class II antibodies
antibodies.61–63 (e.g., Luminex “mixed beads”) and then to perform anti-
body specificity analysis using single-antigen HLA-anti-
body detection beads (SAB). To perform effective antibody
ANTIBODY SCREENING STRATEGIES
screening, patient serum samples should be obtained at
The aim of an antibody screening strategy is to determine regular intervals while on the transplant waiting list to pro-
whether the patient has developed HLA-specific antibod- vide a historical and contemporary assessment of antibody
ies and if so, the antibody level, immunoglobulin class, and levels and specificity.
specificity. The results facilitate the generation of a list of
“acceptable” (antibody negative or antibody levels below PATIENT SENSITIZATION PROFILE AND
a predefined threshold) and “unacceptable” (antibody DEFINITION OF UNACCEPTABLE SPECIFICITIES
positive) donor HLA mismatches for each patient to guide
organ allocation and the selection of suitable antibody- Comprehensive knowledge of a patient’s allosensitiza-
compatible donor–recipient pairs. All laboratories sup- tion events, the HLA antibody specificities, levels, immu-
porting renal transplantation have an antibody screening noglobulin class (IgM/IgG), and timing of appearance/
148 Kidney Transplantation: Principles and Practice

disappearance of HLA antibodies enables the definition of DONOR CROSSMATCH TECHNIQUES AND THEIR
“unacceptable HLA mismatches” where DSA levels above CLINICAL RELEVANCE
a given threshold are likely to cause uncontrolled anti-
body-mediated rejection and constitute a veto to trans- The purpose of a pretransplant donor crossmatch is to
plantation. The antibody level considered as unacceptable detect donor-specific sensitization predictive of hyperacute,
for a given donor HLA mismatch varies between patients acute, and chronic rejection (cellular and humoral) and to
and centers, and is dependent on the clinically acceptable ensure appropriate therapeutic strategies are in place that
level of immunologic risk, patient clinical status, donor are effective at controlling the ensuing rejection response.
type (i.e., living donor, donor after circulatory death, Therefore the crossmatch strategy must define the immu-
or donor after brain death), the likelihood of alternative nologic risk by distinguishing antibodies that are likely to
(lower risk) options, and the local clinical management be harmful, the type of rejection response that is likely to
policy for undertaking HLA-specific antibody incompat- occur, and therapeutic strategies to treat and control the
ible (HLAi) kidney transplantation. HLA mismatches from rejection response. Because of the intricate relationship
a previous transplant and mismatched paternal specifici- between this and the clinical program, crossmatch strate-
ties after pregnancy may also be considered unacceptable. gies vary between centers, dependent on laboratory and
In countries or regions where there is exchange of kidneys, clinical facilities and expertise.
these unacceptable HLA specificities are registered with
the organ exchange organization to prevent unnecessary COMPLEMENT-DEPENDENT
shipping of organs to HLA antibody incompatible patients LYMPHOCYTOTOXIC CROSSMATCH
with a predicted positive donor lymphocyte crossmatch
(termed a “virtual crossmatch”). The list of unacceptable The donor lymphocytotoxic crossmatch test was estab-
HLA specificities to which a patient is sensitized can be lished in the 1960s and has remained a cornerstone for
compared with the HLA types of the organ donor popula- determining donor and recipient compatibility. Recipi-
tion to determine the proportion of donors that would be ent cytotoxic antibodies (predominantly IgM, IgG3, and
antibody incompatible with each patient on the transplant IgG1) that bind donor cells cause activation of the clas-
waiting list. The terms “calculated HLA antibody reaction sical complement pathway and a high percentage of cell
frequency” (cRF) and “calculated panel reactive antibod- death above background levels is interpreted as a “posi-
ies” (cPRA) are used synonymously to describe the level of tive crossmatch” with the potential to damage a trans-
allosensitization, where 0% cRF is nonsensitized, 50% cRF planted kidney. Ensuring a negative pretransplant donor
is antibody incompatible with half of a random donor pool, T cell lymphocytotoxic crossmatch (detecting HLA class
and 90% cRF would be antibody incompatible with 9 out I donor-specific antibodies) has virtually eliminated the
of 10 random donors. occurrence of hyperacute rejection, but in its simplest
form, the CDC crossmatch has several major drawbacks
and has therefore been subject to many modifications (see
Donor Crossmatch previous discussion).

High levels of donor-specific HLA antibodies detected in the B CELL CROSSMATCH


pretransplant DTT-modified CDC donor lymphocytotoxic
crossmatch have a detrimental effect upon graft outcome, Early studies of the clinical relevance of a positive pretrans-
with the majority of transplants succumbing to hyperacute plant donor B-cell crossmatch were hampered by the inabil-
or acute humoral rejection. In the absence of preemptive ity to distinguish potentially damaging HLA class I and class
antibody removal strategies such antibodies constitute a II specific antibodies from nonharmful “non-HLA” antibod-
veto to transplantation. Recipient antibodies against the ies. Contradictory findings ranged between no effect, an
mismatched donor HLA bind to the vascular endothelium of enhanced graft survival and poor graft survival. These find-
the transplanted organ, disrupt the intercellular junctions, ings can now be explained by the heterogeneous antibodies
and cause release of cell surface heparin sulfate and loss of that can cause a positive B cell crossmatch. In studies where
the antithrombotic state, thereby leading to rapid, uncon- antibody specificity was defined, it was clear that the major-
trollable activation of the thrombotic and complement ity of CDC-positive B cell lymphocytotoxic crossmatches are
cascades. The resultant intravascular coagulation and caused by non-HLA specific, usually B cell autoreactive,
interstitial hemorrhage can lead to graft destruction within antibodies that are not harmful to a transplant. A minority
minutes or hours after revascularization of the organ. of positive B cell crossmatches are caused by IgG HLA class
More recently, the clinical importance of low-level donor II specific antibodies that can be deleterious to transplant
HLA-specific antibodies, only detectable using more sensitive outcome, but are unlikely to cause HAR. The presence of
Luminex-SPA and/or flow cytometric crossmatch assay (but unusually high-titer HLA-DR specific antibodies can, how-
CDC negative), have also become apparent as a cause of anti- ever, cause HAR and such antibodies are more common in
body-mediated rejection. Unlike a positive IgG-CDC donor patients with previous graft rejection.65–68
crossmatch, weak donor HLA-specific sensitization does not The introduction of Luminex-based antibody screening
necessarily constitute a veto to transplantation but instead has now enabled precise specification of donor HLA class-II
informs an individualized immunologic risk stratification. It specific antibodies which, when applied together with the
is therefore necessary to consider the choice of crossmatch pretransplant donor B-cell crossmatch has confirmed the
technique(s) used in conjunction with the antibody screen- importance of HLA-DR, -DQ, and -DP antibodies in kidney
ing strategy and the patient’s sensitization status.64 allograft outcome.69–73
10 • Histocompatibility in Renal Transplantation 149

FS (1) vs. SS (2) FITC (3) vs. CD3-PE


104 50
1023
0
103

Events
102

101

0 100 0
0 1023 100 101 102 103 100 101 102 103
FITC
A Forward vs. side scatter B RPE (CD3 vs. FITC) C Fluorescence histogram

55
Strong
Positive positive
crossmatch crossmatch
Negative
control
Events

0
100 101 102 103
FITC
D Fluorescence intensity
Fig. 10.5 Flow cytometric crossmatch test. (A) Cells pass through a laser beam and “forward and side light scatter” is detected by photomultiplier
tubes. An “electronic gate” is used to select cells of morphologic interest (in this case lymphocytes). (B) T lymphocytes are identified using a recombinant
phycoerythrin (RPE)-labeled CD3-specific antibody and HLA-specific IgG bound to cells is identified with FITC labeled antihuman IgG. (C) Light emission
is detected and displayed as a “fluorescence histogram.” (D) Increased FITC fluorescence (a shift to the right on the fluorescence histogram) is a measure
of HLA-specific IgG bound to T lymphocytes above that of the negative control indicating a positive crossmatch.

CROSSMATCH SERUM SAMPLE SELECTION


(TIMING) cyclosporine-based immunosuppression, whereas second-
ary responses (denoted as IgG positive) that commonly occur
An essential feature of the immune system is immunologic after pregnancy and previous transplant rejection are indic-
memory and its ability to produce a rapid and vigorous ative of immunologic priming accompanied by T and B cell
secondary response on reexposure to antigens to which an memory that is poorly controlled by immunosuppression.
individual is already primed. To avert the risk of rejection
caused by an anamnestic memory response, crossmatch
regimens take account of the patient’s current antibody
FLOW CYTOMETRIC CROSSMATCH TEST
status and historic serum samples obtained throughout a Although the CDC crossmatch was effective at averting
recipient’s time on the transplant waiting list, which are HAR, it was apparent that a number of transplants still suf-
selected to represent peak periods of sensitization. fered primary nonfunction or delayed graft function, and
this was particularly prevalent in sensitized patients and
IMMUNOGLOBULIN CLASS AND SPECIFICITY regrafts. This indicated that early graft dysfunction in sen-
sitized recipients may be caused by low levels of antibody,
The findings of acceptable primary graft survival but poor below the sensitivity threshold of the conventional CDC
regraft survival associated with a historic positive (but cur- crossmatch. Garovoy and colleagues75 addressed this ques-
rent negative) crossmatch prompted further modification tion using a flow cytometric crossmatch (FC-XM) test (Fig.
of the CDC crossmatch assay to identify the immunoglob- 10.5) capable of detecting low-level IgG HLA specific anti-
ulin class and specificity of antibodies causing a positive bodies that were undetectable by CDC. In this retrospective
crossmatch. Taylor and colleagues74 reported acceptable study there was a higher incidence of delayed graft func-
primary and regraft survival associated with historic IgM tion and graft failure in the presence of a pretransplant flow
HLA-­ specific sensitization but poor graft survival with cytometric positive (but CDC negative) donor crossmatch,
historic IgG HLA specific antibodies. These results indi- indicating a pathogenic role for weak, sublytic HLA-specific
cated that past allo-sensitization events that only resulted antibodies. Others quickly corroborated this finding, but a
in a transient primary response and IgM allo-antibody significant proportion of patients had an uneventful clinical
production could be readily controlled by conventional course, despite a positive FC-XM. These data demonstrated
150 Kidney Transplantation: Principles and Practice

a high sensitivity but lower specificity of a positive FC-XM can be completed before patient admission. In cases, how-
in predicting early graft dysfunction caused by antibody- ever, where there have been recent allosensitization events,
mediated rejection. Many centers were concerned that particularly in patients undergoing repeat transplantation
“false-positive” crossmatches would unnecessarily deny (e.g., changes in immunosuppression), it is necessary to
patients the opportunity of a transplant and were deterred undertake a prospective pretransplant donor lymphocyte
from adopting the technique in routine clinical practice.76 crossmatch using patient serum obtained within 24 hours
Nevertheless, the predictive value of a positive result was before the operation.
higher in sensitized patients and regrafts that carry an
increased immunologic risk of rejection and the increased THE VIRTUAL CROSSMATCH
assay sensitivity is widely used in such scenarios.77,78
The specificity of antibodies causing a positive donor T In selected cases where a patient’s antibody profile has been
and/or B cell flow cytometric crossmatch test for predicting completely characterized and comprehensive data con-
graft outcome has been further enhanced by the applica- cerning allosensitization events is known, deceased donor
tion of Luminex-based antibody screening. It is now widely organ allocation can be undertaken based on knowledge of
accepted that a positive donor FC-XM in patients with the donor HLA type and a negative “virtual crossmatch.” A
proven DSA, determined using single-antigen HLA-specific negative virtual crossmatch is ascertained by comparing the
antibody detection beads (Luminex-SAB), is associated with HLA mismatches of a donor with the patient’s recent and his-
increased graft loss compared with a negative FC-XM or a toric antibody-defined unacceptable mismatches, and when
positive FC-XM in the absence of DSA.79 no donor HLA-specific antibodies are present, organs can be
The realization of the important role of DSA in chronic allocated to prospective recipients that have a high probabil-
allograft loss is implicated by the deposition of complement ity of a negative donor lymphocyte crossmatch. Such prac-
activation products on peritubular capillaries together tice has been commonplace in the UK and Eurotransplant
with circulating IgG DSA (detected by Luminex SAB) as for many years and is effective at reducing the incidence of
a diagnostic marker for AMR. Loupy et al. showed that shipping deceased donor kidneys to patients that are subse-
DSA detected using the C1q modified Luminex SAB assay quently found to have a positive crossmatch. The efficacy of
is associated with allograft rejection, suggesting that com- this approach is, however, critically dependent on compre-
plement-fixing IgG isotypes (IgG1 and IgG3) confer high hensive donor HLA typing that includes a minimum resolu-
immunologic risk.80 It is not clear, however, if the addi- tion to define all clinically relevant HLA specificities to which
tional information obtained using the Luminex C1q-SAB a patient may develop antibodies. Donor HLA typing for the
assay is independent of antibody strength as the Luminex HLA-C, -DQ, and -DP loci is not routine in all countries or
C1q-SAB and IgG-SAB MFI values are highly correlated centers and under such circumstances a virtual crossmatch
(see reviews81,82). cannot be reliably performed.84
In carefully selected donor–recipient combinations
ORGAN ALLOCATION AND PRETRANSPLANT where recipient sensitization is either absent or clearly
defined low-level nondonor HLA-specific antibodies are
DONOR CROSSMATCH TESTING
present, a negative virtual crossmatch can be predicted
Prolonged cold ischemia time (CIT) is a significant and con- with sufficient confidence to omit the prospective pretrans-
trollable factor that has an adverse effect on deceased donor plant donor lymphocyte crossmatch and proceed directly to
kidney transplant outcome. There is a progressive detrimen- transplantation.85 The adoption and stringent adherence to
tal effect of CIT on transplant outcome with 90% survival at these and similar crossmatch policies removes delays to the
1 year for organs transplanted within 20 hours compared transplant operation associated with donor–recipient histo-
with 83% for organs transplanted at >30 hours (relative risk compatibility testing resulting in lower cold ischemia time
1.9).83 It is therefore essential that deceased donor organ and reduced incidence of delayed graft function.64,86
allocation and crossmatch policies are designed to ensure a
safe decision-making process and minimize delays in trans-
plantation associated with the allocation process. The tech- Immunologic Risk Stratification
nical advances afforded by molecular (PCR-based) methods
and Luminex-SAB antibody screening have facilitated HLA The role of histocompatibility laboratories has progressed
typing, organ allocation, and donor crossmatch strategies from providing a simple binary decision to permit or veto
capable of identifying suitable recipients before completion transplantation, to the development of immunologic risk
of the organ retrieval operation and thus removing delays stratification policies that inform the clinical decision
caused by histocompatibility testing. whether to proceed or not to transplantation and to selec-
Many histocompatibility laboratories receive donor tion of appropriate therapeutic options.
peripheral blood obtained early in the donation process, The antibody screening and donor crossmatch tech-
before commencing the retrieval operation. This enables niques described earlier (CDC, FC-XM, and Luminex-SAB)
prospective donor HLA typing and completion of local and provide different levels of sensitivity and specificity. This
national allocation algorithms to identify potential recipi- information, used together, can inform an individualized
ents before organ donation. In addition, modern cell separa- pretransplant immunologic risk assessment.71 This ranges
tion techniques using immunomagnetic particles enable the from high immunologic risk of HAR in the presence of cur-
recipient crossmatch to be performed using donor periph- rent high-level IgG donor HLA-specific antibodies that con-
eral blood. In selected cases (e.g., nonsensitized patients stitute a veto to transplantation in the absence of effective
with low immunologic risk) archived sera collected within antibody reduction therapy; to intermediate immunologic
the last 3 months are used in the crossmatch test, which risk, in which HAR is unlikely, but an increased incidence
10 • Histocompatibility in Renal Transplantation 151

Fig. 10.6 Pretransplant immunologic risk assessment. Relationship between pretransplant donor crossmatch (determined using CDC and Flow
Cytometry), DSA levels (determined using Luminex single-antigen beads), the immunologic risk and decision to transplant.

of humoral rejection necessitates proactive application TABLE 10.3 Generic Risk Assessment for Antibody-
of effective treatment strategies; to low immunologic risk Mediated Rejection
caused by low-level DSA associated with an increased inci- Immunologic
dence of rejection but little evidence of overall poor graft Risk CDC-XM (IgG) FC-XM Luminex-SAB DSA
outcome. Such generalized risk stratification is not absolute
High Positive Positive Positive (MFI >5000)
because of additional factors such as immunologic memory, Intermediate Negative Positive Positive (MFI >2000)
peak antibody level, antibody priming source, and timing. Lowa Negative Negative Positive (MFI 500–2000)
As a guide Fig. 10.6 and Table 10.3 illustrate the level of Standarda Negative Negative Negative (MFI <500)
immunologic risk based on differences in sensitivity and spec- aThe immunologic risk may vary for individual patients depending on past
ificity of the antibody screening and crossmatch techniques. (historic) allogeneic priming events that occurred before listing for trans-
CDC-XM detects only high levels of complement fixing anti- plantation, when there is no serologic record of possible HLA-specific
bodies, FC-XM provides more sensitive assessment for detec- antibody formation. For example, past pregnancies where the mother
tion of lower level antibodies (below the threshold detectable formed HLA-specific antibodies against the paternal mismatched HLA
by CDC), and Luminex-SAB provides the highest level of sen- and antibody level has declined over time. Long-lived memory T and/or
B cells may become reactivated and cause an unexpected anamnestic
sitivity for antibody detection but when applied alone, pro- response, with acute cellular and/or humoral rejection.
vides the lowest level of specificity to predict graft rejection.

transplant with an HLA antibody compatible donor from


Strategies for Transplanting the national deceased donor pool and special strategies are
required to find suitable kidneys for these patients. This
Sensitized and Highly Sensitized information can be used by clinicians to inform decisions
Patients about the best therapeutic option for a patient.
Patients are often described as highly sensitized, or HSP,
One of the benefits of defining a patient’s HLA antibody if they have an IgG %PRA value or %cRF of 85%, and such
profile is that it is possible to determine the frequency of patients are given priority in national allocation algorithms.
the unacceptable HLA specificities in a donor pool and In the UK, in the 1998 national kidney allocation scheme,
thus estimate the chance of receiving crossmatch-negative HSPs were prioritized for well-matched transplants with
transplant. In the UK a calculated HLA antibody reac- no HLA-A, -B, or -DR mismatches between the donor and
tion frequency or “%cRF” can be determined for patients recipient (000 HLA-A, -B, -DR mismatch grade).87 Patients
on the transplant waiting list by using a tool that com- with completely defined antibody profiles were also eligible
pares a patient’s unacceptable HLA specificities and blood for “favorably matched” transplants. These were trans-
group against a file of 10,000 UK donors. Similar tools are plants where there was a maximum of one HLA-A and one
available within Eurotransplant and in other organiza- HLA-B mismatches in the absence of mismatches at HLA-
tions. Patients with a high %cRF are likely to be difficult to DR (denoted 100, 010, 110 HLA-A, -B, -DR mismatches).
152 Kidney Transplantation: Principles and Practice

This policy resulted in a three-fold increase in the number of living donation and the outcome of such transplants is
of HSP transplanted, 62% of these transplants having a 000 an important consideration. A recent analysis of the UK
mismatch grade.88,89 National Registry for Antibody Incompatible Transplants
The basis for national kidney allocation in the UK showed that the 5-year transplant survival of HLA-incom-
changed in 2006 but the scheme retained the priority given patible transplants was 71% (95% confidence interval [CI]
to HSP.90 The scheme was modified in 2013 to give further 66%–75%) compared with 78% (95% CI, 77%–79%) for
priority for long waiting patients (>7 years), and most of standard living donor transplants and 88% (95% CI, 87%–
these patients are HSP. In the absence of a clinically urgent 89%) for standard deceased donor transplants. This shows
pediatric patient, long waiting patients have absolute pri- that the outcomes are acceptable when faced with a choice
ority when a suitable antibody-compatible kidney becomes between waiting for an antibody-compatible deceased
available. This policy has enabled transplantation of many donor transplant and living donor antibody-incompatible
of these long waiting, HSPs. In future the aim is to identify transplantation.98
patients with a very high %cRF at the time of listing and In considering patients for antibody removal, it is impor-
ensure that suitable kidneys are allocated to them, regard- tant that the HLA antibody specificities and DSA titers
less of waiting time. are determined before the commencement of antibody
Eurotransplant adopted a different approach and intro- removal. This will help inform whether this approach is
duced the Acceptable Mismatch Program for HSPs. In this appropriate for a particular patient. Furthermore, during
program extensive antibody screening was performed to antibody removal it is important to monitor antibody lev-
identify “windows” in the patient’s immune repertoire. els to determine the effectiveness of the treatment regimen.
The HLA specificities of cells unreactive with the patient’s Most centers use Luminex single-antigen beads for antibody
serum were identified as “acceptable mismatches.” The monitoring and advocate that a final crossmatch against
Acceptable Mismatch Program includes minimal mis- the potential donor is performed. After transplantation,
matching criteria of full HLA-DR compatibility or matching antibody rebound usually occurs and monitoring antibody
for one HLA-B and one HLA-DR specificity. This has been levels provides valuable information to indicate whether
a highly successful program, and 43% of patients entered additional antibody removal is required. Experience in per-
into the program are transplanted within 6 months and forming transplants after desensitization is mounting but
58% within 21 months.91 When this system was first intro- because of the complexity in the management before and
duced the antibody screening was performed on carefully after transplantation, it may be that in the longer term,
selected cells with only one mismatched antigen with the patients for transplantation after antibody removal are
HSP. This approach was extremely labor intensive and referred to specialist centers.
would only be possible in a laboratory with access to very
large panels of HLA-typed cells. The advent of solid-phase
assays with single-antigen preparations, and alternatively, Paired Exchange
cell lines expressing single HLA antigens,92 greatly expe-
dites this type of approach. A computer algorithm “HLA Paired exchange, or living donor exchange, is another
Matchmaker” developed by Duquesnoy may assist in defin- option for patients who have a potential living donor, but
ing acceptable mismatches.14,93 In the algorithm each HLA for reasons of HLA or ABO antibody incompatibility the
specificity is represented as a string of amino acid triplets transplant cannot proceed. In such schemes reciprocally
and it is possible to compare HLA specificities and thereby compatible donors and recipients are paired through an
identify mismatched triplets. The theory is that if there are allocation algorithm and exchange transplants are under-
no triplets mismatched, then the specificity will not be rec- taken. Simple systems pair two recipients and their respec-
ognized, and an immune response will not be generated. tive donors and the transplants occur simultaneously, but
Clearly HLA antigens are not linear sequences, nor are it is possible that multiple exchanges can be undertaken
amino acids in a protein “triplets”; nevertheless, the algo- when exchanges are undertaken simultaneously, although
rithm has been shown to assist in the process of specifying a logistically complex, the process maximizes the chances
patient’s sensitization profile.94–96 that all transplants will proceed. There are other modifi-
A recent analysis of the 10-year outcome of transplants cations of the process that facilitate chains of transplants.
performed in highly sensitized patients within Eurotrans- The first is “domino-paired donation” (DPD) whereby a
plant showed that patients who were transplanted through chain of simultaneous living donor transplants is initi-
the Acceptable Mismatch Program on the basis of proven ated by a kidney from a nondirected altruistic donor and
acceptable mismatches had superior graft survival to ends with transplantation of the last donor kidney into a
patients transplanted on the basis of unacceptable mis- recipient registered for a deceased donor transplant.99 The
matches.97 This demonstrates the importance of careful second type of chain is termed nonsimultaneous extended
HLA antibody specification on transplant outcome. altruistic donor (NEAD) chains. In the NEAD chains
the transplants are not performed simultaneously and
“bridge donors” are created, whose incompatible recipient
Antibody Removal is already transplanted and the donor waits to donate to
another recipient. This process removes the requirement
One of the options for transplanting sensitized patients is for complex logistics, but adds in a risk that the “bridge
to use antibody removal techniques to reduce donor-spe- donor” may renege.100 There are well-established kidney
cific HLA antibody before an HLA incompatible transplant exchange programs in the US and in Europe101–103 (see
(see Chapter 22). This is usually considered in the context Chapter 23).
10 • Histocompatibility in Renal Transplantation 153

Of relevance to histocompatibility, to achieve efficient of MICA antibodies in transplanted patients and a higher
exchange it is crucial that the patients’ HLA antibody pro- incidence of antibodies in patients whose transplants
files are accurately defined to avoid positive crossmatches failed.126,127 However, data from a large multicenter study
and disruption of planned exchanges. Most of the trans- showed that the presence of MICA posttransplant was not
plants performed by this route are antibody compatible, but an independent risk factor for graft loss.128
in patients who are highly sensitized and thus difficult to
transplant with a compatible donor from a relatively small
donor pool, antibody incompatible transplantation can also Future Directions
be facilitated. By review the patient’s HLA antibody profile
and unacceptable HLA specificities, it is possible to “de- The traditional approach to assessing HLA compatibility is
list” unacceptable specificities and achieve more favorable to count the number of mismatched donor HLA specificities
antibody incompatible transplants than would have been either for all loci together (i.e., 0–6 mismatches for HLA-A,
achieved with direct donation. -B, -DR) or at individual loci (i.e., 1.1.0., see previous). Since
its inception, however, it has been recognized that HLA poly-
morphism has evolved from common ancestral genes under-
Posttransplant Monitoring going equal and unequal recombination, gene duplication,
DNA insertion, and point mutation. As a result many HLA
There is extensive literature on the development of donor specificities share common amino acid sequence motifs (epit-
HLA-specific antibodies after renal transplantation.104–106 opes) that give rise to extensive serologic cross-reactivity.129
DSA together with histologic and immunohistological (C4d Some “families” of HLA molecules are very similar, whereas
staining) are features in the Banff scheme for the diagno- others are highly divergent130 and are therefore likely to dif-
sis of antibody-mediated rejection ABMR.107 These fac- fer in their ability to evoke allograft rejection, depending on
tors are known to have an important role in transplant the amino acid sequence of mismatched donor HLA alleles in
failure.108–111 The proportion of recipients developing relation to the recipient HLA type.
antibodies posttransplantation has been reported to range Historical attempts to identify “immunodominant” mis-
between 12% and 60%.112 This figure is not only influ- matches (e.g., taboo mismatches) that are more likely to be
enced by the sensitivity of the assay system used to detect the target of a host rejection response have not withstood
DSA, but also by clinical factors such as the nature and the test of time. However, modern DNA sequence-based
degree of HLA mismatching between the donor and recipi- technology now facilitates HLA typing at allele (2-field)
ent, immunosuppressive regimens and compliance with level, which together with knowledge of tertiary protein
immunosuppression. structure enables a new approach to assess mismatched
The appearance of donor HLA-specific antibodies has alloantigen immunogenicity. The amino acid sequence of
been shown to be associated with a poorer outcome and donor HLA alleles are compared with the recipient HLA type
with the occurrence of acute and chronic rejection, and is and assessed using computer algorithms that are effective
now recognized as the leading pathology of graft attrition to predict alloantigen posttransplant alloantibody forma-
and a major cause of graft loss.113,114 Analysis of serial tion and graft outcome.131–133 Furthermore, atomic resolu-
posttransplant serum samples showed that donor-specific tion homology modeling of HLA protein tertiary structure
HLA antibodies are strongly predictive of allograft failure obtained by x-ray crystallography has been applied to
being detected before chronic rejection or transplant fail- compare surface physicochemical properties (electrostatic
ure.115,116 The results of a large international prospective charge and hydrophobicity) that govern protein–protein
trial that included more than 4500 patients from 36 units (i.e., antibody–antigen) interaction, to provide a more pre-
also concluded that HLA antibody production precedes cise assessment of donor alloantigen immunogenicity.130
transplant failure.117 The temporal relationship between It is now clear that assessing donor and recipient HLA
the appearance of DSA and onset of graft dysfunction can, mismatches at the amino acid sequence level, together with
however, range from months to many years and it is not yet patient compliance and/or clinical reduction/withdrawal
clear that clinical intervention is effective to attenuate the of immunosuppression are strong independent predictors
clinical outcome which is dependent on antibody level and of alloantibody formation and graft rejection.134,135 Such
specificity.118,119 algorithms are widely accepted to provide superior assess-
Mismatched HLA antigens are important stimuli for ment of donor–recipient HLA compatibility and can be
an alloimmune response, but antibodies to nonclassi- used to assist pretransplant organ allocation and posttrans-
cal polymorphic MHC antigens may also contribute. The plant immunologic risk assessment, to identify patients at
MHC-related chain A and B antigens (MICA and MICB) increased risk of humoral rejection. In the future such algo-
are expressed on epithelia in response to cellular stress and rithms will be a helpful guide for donor selection, and the
on endothelium in vitro. In the kidney MICA and MICB choice and level of maintenance immunosuppression, with
expression has been reported on tubular epithelia.120,121 the aim of improving long-term allograft survival.
Antibodies to MICA were reported in the sera of transplant
recipients,122,123 but as the antigens are not expressed on
lymphocytes124,125 MICA and MICB antibodies are not Concluding Remarks
detected in standard crossmatch tests. Furthermore, donors
and recipients are not routinely typed genotyped for MICA HLA matching, definition of allosensitization, donor
and therefore donor specificity of the MICA antibodies can- crossmatching, and posttransplant antibody monitor-
not be determined. Studies have demonstrated the presence ing all make important contributions to successful renal
154 Kidney Transplantation: Principles and Practice

transplant programs. Growing knowledge of immunologic 16. Mickey MR. HLA matching in transplants from cadaver donors. In:
risk stratification together with accurate information about Terasaki PI, editor. Clinical Kidney Transplants 1985. Los Angeles,
CA: UCLA Tissue Typing Laboratory; 1985. p. 45–56.
the availability and likelihood of alternative (lower risk) 17. Persijn GG, Cohen B, Lansbergen Q, et al. Effect of HLA-A and -B
donor options can guide the clinical decision to proceed matching on survival of grafts and recipients after renal transplanta-
or not to transplantation and initiate preemptive therapy tion. N Engl J Med 1982;307:905.
to optimize posttransplant clinical course and long-term 18. Terasaki PI, Thrasher DL, Hauber TH. Serotyping for homotransplan-
tations: XIII. Immediate kidney rejection and associated pre-formed
graft outcome. Rigid adherence to unacceptable HLA mis- antibodies. In: Dausset J, Hamburger J, Mathe G, editors. Advances in
matches defined by Luminex SAB alone may be unhelpful, Transplantation. Copenhagen: Munksgaard; 1968. p. 225–9.
but when applied together with CDC-XM and/or FC-XM, the 19. Ting A, Morris PJ. Matching for the B-cell antigens of the HLA-DR
information provides improved sensitivity and specificity to series in cadaver renal transplantation. Lancet 1978;1:575.
prospectively identify donor-recipient pairs with low, inter- 20. Ting A, Morris PJ. Powerful effect of HLA-DR matching on survival of
cadaveric renal allografts. Lancet 1980;2:282.
mediate, and high risk of humoral rejection. High immu- 21. Cecka JM. The UNOS scientific renal transplant registry—ten years
nologic risk usually constitutes a veto to transplantation, of kidney transplants. In: Cecka M, Terasaki PI, editors. Clinical
low and intermediate risk HLAi transplantation should be Transplants 1997. Los Angeles, CA: UCLA Tissue Typing Labora-
avoided if alternative options are likely, and should only be tory; 1998. p. 1–16.
22. Taylor CJ, Welsh KI, Gray CM, et al. Clinical and socio-economic ben-
undertaken with appropriate clinical caution. The immu- efits of serological HLA-DR matching for renal transplantation over
nologic risk of proceeding to transplantation should be three eras of immunosuppression regimens at a single unit. In: Tera-
assessed together with the clinical risk of not proceeding saki PI, Cecka M, editors. Clinical Transplants 1993. Los Angeles,
and the patient remaining on dialysis. CA: UCLA Tissue Typing Laboratory; 1994. p. 233–41.
The recent realization that donor HLA-specific alloanti- 23. Taylor CJ, Bayne AM, Welsh KI, et al. HLA-DR matching is effec-
tive in reducing post transplant costs in renal allograft recipients on
bodies are the leading cause of graft attrition has led to a triple therapy. Transplant Proc 1993;25:210.
reexamination of pre- and posttransplant allosensitization 24. Susal C, Opelz G. Current role of human leukocyte antigen match-
and the role of HLA matching. It is an exciting time as many ing in kidney transplantation. Curr Opin Organ Transplant
of the traditional boundaries are being challenged to enable 2013;18:438.
25. Williams RC, Opelz G, McGarvey CJ, et al. The risk of transplant fail-
transplantation of patients who previously would have ure with HLA mismatch in first adult kidney allografts from deceased
been unlikely to be transplanted. donors. Transplantation 2016;100:1094.
26. Morris PJ, Johnson RJ, Fuggle SV, et al. Analysis of factors that affect
References outcome of primary cadaveric renal transplantation in the UK. Lan-
1. Terasaki PI, editor. History of HLA: Ten Recollections. Los Angeles, cet 1999;354:1147.
CA: UCLA Tissue Typing Laboratory; 1990. p. 90024. 27. Fuggle SV, Johnson RJ, Rudge CJ, et al. Human leukocyte antigen
2. Morris PJ, Williams GM, Hume DM, et al. Serotyping for homotrans- and the allocation of kidneys from cadaver donors in the United
plantation. XII. Occurrence of cytotoxic antibodies following kidney Kingdom. Transplantation 2004;77:618.
transplantation in man. Transplantation 1968;6:392. 28. Johnson RJ, Fuggle SV, O’Neill J, et al. Factors influencing outcome
3. Kissmeyer-Nielsen F, Olsen S, Petersen VP, et al. Hyperacute rejec- after deceased heartbeating donor kidney transplantation in the UK:
tion of kidney allografts, associated with pre-existing humoral anti- an evidence base for a new national kidney allocation policy. Trans-
bodies against donor cells. Lancet 1996;2:662. plantation 2010;89:379.
4. Williams GM, Hume DM, Hudson Jr RP, et al. “Hyperacute” renal- 29. Opelz G, Dohler B. Ceppellini lecture 2012: collateral damage from
homograft rejection in man. N Engl J Med 1968;279:611. HLA mismatching in kidney transplantation. Tissue Antigens
5. Gorer PA, Lyman S, Snell GD. Studies on the genetic and antigenic 2013;8:235.
basis of tumour transplantation. Linkage between a histocompatibil- 30. Opelz G, Dohler B. Association of HLA mismatch with death with a
ity gene and ‘fused’ in mice. Proc Roy Soc B 1948;151:57. functioning graft after kidney transplantation: a collaborative trans-
6. Horton R, Wilming L, Rand V, et al. Gene map of the extended plant study report. Am J Transplant 2012;12:3031.
human MHC. Nat Rev Genet 2004;5:889. 31. Opelz G, Döhler B. Impact of HLA mismatching on incidence of post-
7. Daar AS, Fuggle SV, Fabre JW, et al. The detailed distribution of transplant non-Hodgkin lymphoma after kidney transplantation.
HLA-A, B, C antigens in normal human organs. Transplantation Transplantation 2010;89:567.
1984;38:287. 32. Opelz G, Döhler B. Pediatric kidney transplantation: analysis of donor
8. Daar AS, Fuggle SV, Fabre JW, et al. The detailed distribution of age, HLA match and post-transplant non-Hodgkin ­ lymphoma.
MHC Class II antigens in normal human organs. Transplantation Transplantation 2010;90:292.
1984;38:293. 33. Opelz G, Döhler B. Association of mismatches for HLA-DR with inci-
9. Fuggle SV, McWhinnie DL, Chapman, et al. Sequential analysis of dence of post-transplant hip fracture in kidney transplant recipients.
HLA-class II antigen expression in human renal allografts. Induction Transplantation 2011;91:65.
of tubular class II antigens and correlation with clinical parameters. 34. Tong JY, Hsia S, Parris GL, et al. Molecular compatibility and
Transplantation 1986;42:144. renal graft survival- the HLA-DQB1 genotyping. Transplantation
10. Fuggle SV, McWhinnie DL, Morris PJ. Precise specificity of induced 1993;55:390.
tubular HLA-class II antigens in renal allografts. Transplantation 35. Bushell A, Higgins RM, Wood KJ, et al. HLA-DQ mismatches
1987;44:214. between donor and recipient in the presence of HLA-DR compatibil-
11. Marsh SGE, Albert ED, Bodmer WF, et al. Nomenclature for factors of ity do not influence the outcome of renal transplants. Hum Immunol
the HLA system 2010. Tissue Antigens 2010;75:291. 1989;26:179.
12. Akkoc N, Scornik JC. Intramolecular specificity of anti-HLA alloanti- 36. Freedman BI, Thacker L, Heise ER, et al. HLA-DQ matching in cadav-
bodies. Hum Immunol 1991;30:91. eric renal transplantation. Clin Transpl 1997;11:480.
13. Marsh SGE, Bodmer JG. HLA-DR and -DQ epitopes and monoclonal 37. Lim WH, Chapman JR, Coates PT, et al. HLA-DQ mismatches and
antibody specificity. Immunol Today 1989;10:305. rejection in kidney transplant recipients. Clin J Am Soc Nephrol
14. Duquesnoy RJ. HLA matchmaker: a molecularly based algorithm 2016;11:875.
for histocompatibility determination I Description of the algorithm. 38. Mytilineos J, Deufel A, Opelz G. Clinical relevance of HLA-DPB locus
Hum Immunol 2002;63:339. matching for cadaver kidney retransplants: a report of the Collabora-
15. Taylor CJ, Bolton EM, Pocock S, et al. Banking on human embryonic tive Transplant Study. Transplantation 1997;63:1351.
stem cells: estimating the number of donor cell lines needed for HLA 39. Laux G, Mansmann U, Deufel A, et al. A new epitope-based HLA-DPB
matching. Lancet 2005;366:2019. matching approach for cadaver kidney retransplants. Transplanta-
tion 2003;75:1527.
10 • Histocompatibility in Renal Transplantation 155

40. Taylor CJ, Dyer PA. Maximising the benefits of HLA matching for of immunoglobulin G strength on single antigen beads. Hum Immu-
renal transplantation; alleles, specificities, CREGs, epitopes or resi- nol 2011;72:849.
dues? Transplantation 1999;68:1093. 63. Sicard A, Ducreux A, Rabeyrin M, et al. Detection of C3d-binding
41. Thompson JS, Thacker LR. CREG matching for first cadaveric kidney donor-specific anti-HLA antibodies at diagnosis of humoral rejection
transplants performed by SEOPF centers between October 1987 and predicts renal graft loss. J Am Soc Nephrol 2015;6:457.
September 1995. Southeastern Organ Procurement Foundation. 64. Taylor CJ, Kosmoliaptsis V, Sharples LD, et al. Ten year experience of
Clin Transplant 1996;10:586. selective omission of the pre-transplant crossmatch test in deceased
42. Takemoto SK. HLA amino acid residue matching. Clin Transplants donor kidney transplantation. Transplantation 2010;89:185.
1996;397. 65. Berg B, Moller E. Immediate rejection of a HLA-A, B compatible,
43. Wujciak T, Opelz G. Evaluation of HLA matching for CREG antigens HLA-DR incompatible kidney with a positive donor-recipient B-cell
in Europe. Transplantation 1999;68:1097. crossmatch. Scand J Urol Nephrol Suppl 1980;54:36.
44. Kosmoliaptsis V, Sharples LD, Chaudhry A, et al. HLA class I amino 66. Mohanakumar T, Rhodes C, Mendez-Picon G, et al. Renal allograft
acid sequence based matching following inter-locus subtraction and rejection associated with presensitization to HLA-DR antigens.
long-term outcome after deceased donor kidney transplantation. Transplantation 1981;31:93.
Hum Immunol 2010;71:851. 67. Ahern AT, Artruc SB, DellaPelle P, et al. Hyperacute rejection of
45. Wiebe C, Pochinco D, Blydt-Hansen TD, et al. Class II HLA epi- HLA-AB-identical renal allografts associated with B lymphocyte and
tope matching-A strategy to minimize de novo donor-specific endothelial reactive antibodies. Transplantation 1982;33:103.
antibody development and improve outcomes. Am J Transplant 68. Scornik JC, LeFor WM, Cicciarelli JC, et al. Hyperacute and acute kid-
2013;13:3114. ney graft rejection due to antibodies against B cells. Transplantation
46. Lim WH, Wong G, Heidt S, et al. Novel aspects of epitope match- 1992;54:61.
ing and practical application in kidney transplantation. Kidney Int 69. Billen EV, Christiaans MH, Doxiadis II , et al. HLA-DP antibodies before
2017;93:314. and after renal transplantation. Tissue Antigens 2010;75:278.
47. Karahan GE, de Vaal YJH, Roelen DL, Buchli R, Claas FHJ, Heidt S. 70. Dunn TB, Noreen H, Gillingham K, et al. Revisiting traditional risk
Quantification of HLA class II-specific memory B cells in HLA-sensi- factors for rejection and graft loss after kidney transplantation. Am J
tized individuals. Hum Immunol 2015;76:129. Transplant 2011;11:2132.
48. Karahan GE, de Vaal YJH, Krop J, et al. A memory B cell crossmatch 71. Dyer PA, Claas FHJ, Doxiadis II , et al. Minimising the clinical impact
assay for quantification of donor-specific memory B cells in the of the alloimmune response through effective histocompatibility test-
peripheral blood of HLA-immunized individuals. Am J Transplant ing for organ transplantation. Transplant Immunol 2012;27:83.
2017;17:2617. 72. Jolly EC, Key T, Rasheed H, et al. Pre-formed donor HLA-DP specific
49. British Transplantation Society and British Society for Histocom- antibodies mediate acute and chronic antibody-mediated rejection
patibility and Immunogenetics. Guidelines for the detection and following renal transplantation. Am J Transplant 2012;12:2845.
characterisation of clinically relevant antibodies in solid organ 73. Otten HG, Verhaar MC, Borst HP, et al. Pretransplant donor-specific
transplantation. 2014. Available online at: https://www.bts.org uk/ HLA Class-I and -II antibodies are associated with an increased risk
guidelines-standards/archived-guidelines. for kidney graft failure. Am J Transplant 2012;12:1618.
50. Kosmoliaptsis V, Gjorgjimajkoska O, Sharples LD, et al. Impact of 74. Taylor CJ, Chapman JR, Ting A, et al. Characterisation of lymphocy-
donor mismatches at individual HLA-A, -B, -C, -DR, and -DQ loci totoxic antibodies causing a positive crossmatch in renal transplan-
on the development of HLA-specific antibodies in patients listed for tation: relationship to primary and regraft outcome. Transplantation
repeat renal transplantation. Kidney Int 2014;86:1039. 1989;48:953.
51. Wiebe C Nevins TE, Robiner WN, Thomas W, Matas AJ, Nickerson 75. Garovoy MR, Rheinschmidt MA, Bigos M, et al. Flow cytometric
PW. The synergistic effect of class II HLA epitope-mismatch and non- analysis: a high technology crossmatch technique facilitating trans-
adherence on acute rejection and graft survival. Am J Transplant plantation. Transplant Proc 1983;15:1939.
2015;15:2197. 76. Lazda VA, Pollak R, Mozes MF, et al. The relationship between flow
52. Andrews PA. On behalf of the standards committee of the British cytometer crossmatch results and subsequent rejection episodes in
Transplantation Society. Summary of the British Transplantation cadaver renal allograft recipients. Transplantation 1988;45:562.
Society Guidelines for Management of the Failing Kidney. Trans- 77. Cook DJ, Terasaki PI, Iwaki Y, et al. The flow cytometry crossmatch
plant Transplantation 2014;98:1130. in kidney transplantation. Clin Transpl 1987:409.
53. Otten HG, Verhaar MC, Borst HP, et al. The significance of pretrans- 78. Karpinski M, Rush D, Jeffery J, et al. Flow cytometric crossmatching
plant donor-specific antibodies reactive with intact or denatured in primary renal transplant recipients with a negative anti-human
human leucocyte antigen in kidney transplantation. Clin Exp Immu- globulin enhanced cytotoxicity crossmatch. J Am Soc Nephrol
nol 2013;173:536. 2001;12:2807.
54. Ting A, Morris PJ. Reactivity of autolymphocytotoxic antibodies 79. Couzi L, Araujo C, Guidicelli G, et al. Interpretation of positive flow
from dialysis patients with lymphocytes from chronic lymphocytic cytometric crossmatch in the era of the single-antigen bead assay.
leukaemia (CLL) patients. Transplantation 1978;25:31. Transplantation 2011;91:527.
55. Taylor CJ, Kosmoliaptsis V, Summers DM, et al. Back to the future: 80. Loupy A, Lefaucheur C, Vernerey D, et al. Complement-binding
application of contemporary technology to long-standing questions anti-HLA antibodies and kidney-allograft survival. N Engl J Med
about the clinical relevance of HLA-specific alloantibodies in renal 2013;369:1215.
transplantation. Hum Immunol 2009;70:563. 81. Filippone EJ, Farber JL. The humoral theory of transplantation: epit-
56. Fulton RJ, McDade RDC, Smith PL, et al. Advanced multiplexed anal- ope analysis and the pathogenicity of HLA antibodies. J Immunol Res
ysis with the FlowMetrix system. Clin Chem 1997;43:1749. 2016;2016:5197396.
57. Morales-Buenrostro LE, Terasaki PI, Marino-Vazquez LA, et al. ‘Nat- 82. Taylor CJ, Kosmoliaptsis V, Martin J, et al. Technical limitations of
ural’ human leukocyte antigen antibodies found in nonalloimmu- the C1q single antigen bead assay to detect complement binding
nized healthy males. Transplantation 2008;86:1111. HLA-specific antibodies. Transplantation 2017;101:1206.
58. Peacock S, Kosmoliaptsis V, Bradley AJ, et al. Questioning the added 83. Summers DM, Johnson RJ, Allen J, et al. Analysis of factors that
value of Luminex single antigen beads to detect C1q binding donor determine outcome following transplantation with kidneys donated
HLA-specific antibodies. Transplantation 2014;98:384. after cardiac death in the UK. Lancet 2010;376:1303.
59. Schaub S, Hönger G, Koller MT, et al. Determinants of C1q binding in 84. Bryan CF, Luger AM, Smith JL, et al. Sharing kidneys across donor-
the single antigen bead assay. Transplantation 2014;98:387. service area boundaries with sensitized candidates can be influenced
60. Khan N, Robson AJ, Worthington JE, et al. The detection and defi- by HLA-C. Clin Transplant 2010;24:56.
nition of IgM alloantibodies in the presence of IgM autoantibodies 85. Taylor CJ, Smith SI, Morgan CH, et al. Selective omission of the donor
using Flow PRA beads. Hum Immunol 2003;64:593. crossmatch before renal transplantation; efficacy, safety and effects
61. Smith JD, Hamour IM, Banner MR, et al. C4d fixing, Luminex bind- on cold storage time. Transplantation 2000;69:719.
ing antibodies - a new tool for prediction of graft failure after heart 86. Shrestha S, Bradbury L, Boal M, et al. Logistical factors influencing
transplantation. Am J Transplant 2007;7:2809. cold ischemia times in deceased donor kidney transplants. Trans-
62. Chen G, Sequeira F, Tyan DB. Novel C1q assay reveals a clinically plantation 2016;100:422.
relevant subset of human leukocyte antigen antibodies independent 87. Fuggle SV, Belger MA, Johnson RJ, et al. A new national scheme for
the allocation of adult kidneys in the UK. In: Cecka JM, Terasaki P,
156 Kidney Transplantation: Principles and Practice

editors. Clinical Transplants 1998. Los Angeles, CA: UCLA Tissue 112. McKenna RM, Takemoto S, Terasaki PI. Anti-HLA antibodies after
Typing Laboratory; 1999. p. 107–13. solid organ transplantation. Transplantation 2000;69:319.
88. Fuggle SV, Martin S. Towards performing transplantation in highly 113. Loupy A, Hill GS, SuberBielle C, et al. Significance of C4d Banff
sensitized patients. Transplantation 2004;78:186. scores in early protocol biopsies of kidney transplant recipients
89. Fuggle SV, Johnson RJ, Bradley JA, et al. Impact of the 1998 UK with pre-formed donor-specific antibodies (DSA). Am J Transplant
national allocation scheme for deceased heartbeating donor kidneys. 2011;11:56.
Transplantation 2010;89:372. 114. Wiebe C, Nickerson P. Posttransplant monitoring of de novo human
90. Johnson RJ, Fuggle SV, Mumford L, et al. A new UK (2006) national leukocyte antigen donor-specific antibodies in kidney transplanta-
kidney allocation scheme (2006 NKAS) for deceased heartbeating tion. Curr Opin Organ Transplant 2013;18:470.
donor kidneys. Transplantation 2010;89:387. 115. Lee PC, Terasaki PI, Takemoto SK, et al. All chronic failures of kidney
91. Claas F, Witvliet MD, Duquesnoy RJ, et al. The acceptable mismatch transplants were preceded by the development of HLA antibodies.
program as a fast tool to transplant highly sensitized patients await- Transplantation 2002;74:1192.
ing a post-mortal kidney: short waiting time and excellent graft out- 116. Worthington JE, Martin S, Al-Husseini DM, et al. Post-transplan-
come. Transplantation 2004;78:190. tation production of donor HLA-specific antibodies is a predictor of
92. Zoet YM, Eijsink C, Kardol MJ, et al. The single antigen expressing renal transplant outcome. Transplantation 2003;75:1034.
lines (SALs) concept: an excellent tool for the screening for HLA spe- 117. Terasaki PI, Ozawa M. Predicting kidney graft failure by HLA anti-
cific antibodies. Hum Immunol 2005;66:519. bodies: a prospective trial. Am J Transplant 2004;4:438.
93. Duquesnoy RJ, Witvliet M, Doxiadus II , et al. HLA-matchmaker 118. Terasaki PI, Cai J. Human leukocyte antigen antibodies and
based strategy to identify acceptable HLA class I mismatches for chronic rejection: from association to causation. Transplantation
highly sensitized kidney transplant candidates. Transplant Int 2008;86:377.
2004;17:22. 119. Everly MJ, Everly JJ, Arend LJ, et al. Reducing de novo donor-specific
94. Duquesnoy RJ, Takemoto S, de Lange P, et al. HLA-matchmaker: a antibody levels during acute rejection diminishes renal allograft loss.
molecularly based algorithm for histocompatibility determination. Am J Transplant 2009;9:1063.
III. Effect of matching at the HLA-A,B amino acid triplet level on kid- 120. Hankey KG, Deachenberg CB, Papadimitriou JC, et al. MIC expression
ney transplant survival. Transplantation 2003;75:884. in renal and pancreatic allografts. Transplantation 2002;73:304.
95. Goodman RS, Taylor CJ, O’Rourke CM, et al. Utility of HLAMatch- 121. Quiroga I, Salio M, Koo DDH, et al. Expression of MHC class I-related
maker and single-antigen HLA-antibody detection beads for iden- chain B (MICB) molecules on renal transplant biopsies. Transplanta-
tification of acceptable mismatches in highly sensitized patients tion 2005;81:1196.
awaiting kidney transplantation. Transplantation 2006;81:1331. 122. Zwirner NW, Marcos CY, Mirbaha F, et al. Identification of MICA as
96. Kosmoliaptsis V, Bradley JA, Sharples LD, et al. Predicting the a new polymorphic alloantigen recognized by antibodies in sera of
immunogenicity of HLA class I alloantigens using structural epi- organ transplant recipients. Hum Immunol 2000;61:917.
tope analysis determined by HLAMatchmaker. Transplantation 123. Sumitran-Holgersson SS, Wilczek HE, HolgerssonJ, et al. Identifica-
2008;85:1817. tion of the nonclassical HLA molecules, MICA, as targets for humoral
97. Heidt S, Haasnoot GW, van Rood JJ, et al. Kidney allocation based immunity associated with irreversible rejection of kidney allografts.
on proven acceptable antigens results in superior graft survival in Transplantation 2002;74:269.
highly sensitized patients. Kidney Int 2017;93:491. 124. Zwirner NW, Fernandez-Vina MA, Stastny P. MICA, a new poly-
98. Pankhurst L, Hudson A, Mumford L, et al. The UK National Regis- morphic HLA-related antigen, is expressed mainly by keratinocytes,
try of ABO and HLA antibody incompatible renal transplantation: endothelial cells, and monocytes. Immunogenetics 1998;47:139.
pretransplant factors associated with outcome in 879 transplants. 125. Zwirner NW, Dole K, Stastny P. Differential surface expression of
Transplant Direct 2017;3:181. MICA by endothelial cells, fibroblasts, keratinocytes and monocytes.
99. Montgomery RA, Zachery AA, Ratner LE, et al. Domino aired kidney Hum Immunol 1999;60:323.
donation: a strategy to make best use of live nondirected donation. 126. Mizutani K, Teraski PI, Rosen A, et al. Serial ten year follow-up of
Lancet 2006;368:419. HLA and MICA antibody production prior to graft failure. Am J
100. Aslagi I, Gichrist DS, Roth AE, et al. Non simultaneous chains and Transplant 2005;5:2265.
dominos in kidney paired donation-revisited. Am J Transplant 127. Mizutani K, Terasaki P, Bignon JD, et al. Association of kidney
2011;11:984. transplant failure and antibodies against MICA. Hum Immunol
101. Segev DL, Genrty SE, Warren DS, et al. Kidney paired donation and 2006;67:683.
optimizing the use of live donor organs. JAMA 2005;293:1883. 128. Lemy A, Andrien M, Lionet A. Posttransplant major histocompat-
102. Johnson RJ, Allen JE, Fuggle SV, et al. Early experience of paired ibility complex class i chain-related gene a antibodies and long-term
donation in the United Kingdom. Transplantation 2008;86:1672. graft outcomes in a multicenter cohort of 779 kidney transplant
103. De Klerk M, Kal-van Gestel JA, Haase-Kromwijk BD, et al. Eight years recipients. Transplantation 2012;93:1258.
of the Dutch living donor kidney exchange program. Clin Transpl 129. Tambur AR, Claas FH. HLA epitopes as viewed by antibodies: what is
2011;287. it all about? Am J Transplant 2015;15:1148.
104. Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Evolution and clinical 130. Mallon DH, Bradley JA, Taylor CJ, et al. Structural and electrostatic
pathologic correlations of de novo donor-specific HLA antibody post analysis of HLA B-cell epitopes: inference on immunogenicity and
kidney transplant. Am J Transplant 2012;12:1157. prediction of humoral alloresponses. Curr Opin Organ Transplant
105. Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Rates and determinants 2014;19:420.
of progression to graft failure in kidney allograft recipients with de 131. Dankers MK, Witvliet MD, Roelen DL, et al. The number of amino
novo donor-specific antibody. Am J Transplant 2015;15:2921. acid triplet differences between patient and donor is predictive for the
106. Tambur AR, Wiebe C. HLA diagnostics: evaluating DSA strength by antibody reactivity against mismatched human leukocyte antigens.
titration. Transplantation 2018;102:S23. Transplantation 2004;77:1236.
107. Haas M. An updated Banff Schema for diagnosis of antibody medi- 132. Kosmoliaptsis V, Sharples LD, Chaudhry AN, et al. Predicting HLA
ated rejection in renal allografts. Curr Opin Organ Transplant class II alloantigen immunogenicity from the number and physio-
2014;19:315. chemical properties of amino acid polymorphisms. Transplantation
108. Feucht HE, Schneeberger H, Hillebrand G, et al. Capillary deposition 2011;91:183.
of C4d complement fragment and early renal graft loss. Kidney Int 133. Duquesnoy RJ. HLA epitope based matching for transplantation.
1993;43:1333. Transplant Immunol 2014;31:1.
109. Terasaki PI. Humoral theory of transplantation. Am J Transplant 134. Wiebe C, Nickerson P. Strategic use of epitope matching to improve
2003;3:665. outcomes. Transplantation 2016;100:2048.
110. Koo DDH, Roberts ISD, Quiroga I, et al. C4d deposition in early renal 135. Kosmoliaptsis V, Mallon DH, Chen Y, et al. Alloantibody responses
allograft protocol biopsies. Transplantation 2004;78:398. after renal transplant failure can be better predicted by donor-recip-
111. Cai J, Terasaki PI. Humoral theory of transplantation-mechanism, ient HLA amino acid sequence and physicochemical disparities than
prevention, and treatment. Hum Immunol 2005;66:334. conventional HLA matching. Am J Transplant 2016;16:2139.
11 Surgical Techniques of
Kidney Transplantation
CHRISTOPHER J.E. WATSON, PETER J. FRIEND and
LORNA P. MARSON

CHAPTER OUTLINE Preparation of Recipient Double Ureters


Preparation of Kidney Augmented Bladder
Site Pyelopyelostomy
Incision Pyeloureterostomy and
Preparation of Operative Bed Ureteroureterostomy
Revascularization Pyelovesicostomy
Arterial Anastomosis Ureteroenterostomy
External Iliac Artery Ureteric Stents
Internal Iliac Artery Management of Catheter and Stent
Venous Anastomosis Closure
Reperfusion of the Kidney Pediatric Recipient
Reconstruction of the Urinary Tract Pediatric Donor
Ureteroneocystostomy (Anastomosis of the Double Kidney Transplant
Transplant Ureter Directly to the Bladder) Transplant Nephrectomy
Transvesical Ureteroneocystostomy
Extravesical Ureteroneocystostomy
Parallel Incision Ureteroneocystostomy

Kidney transplantation is a major surgical procedure that Preparation of Recipient


involves both vascular and ureteric anastomoses, and it
is usually performed by a dedicated transplant surgeon, The general preparation and selection of recipients for
although in the past it was performed predominantly transplantation is discussed in Chapter 4. Potential kidney
by urologists or vascular surgeons. Most recipients are transplant recipients are carefully assessed before being
already established on dialysis, although some may avoid placed on the waiting list. Medical and surgical risk factors
dialysis by having the transplant preemptively, and many will have been identified and evaluated, and periodically
consider this the gold standard treatment. Preemptive reassessed while waiting. On admission for transplanta-
transplantation confers a modest benefit over transplan- tion, a further careful history and physical examination
tation after the start of dialysis, but not if the duration of are required to ensure that there is no immediate contra-
dialysis is less than a year. There are concerns about equity indication to major surgery. For example, have there been
of access to preemptive transplants, as it tends to disad- changes since the patient was last assessed? Particular
vantage individuals who are less educated and from lower attention should be paid to the patient’s fluid and electro-
socioeconomic groups. In addition there is a suggestion lyte status. In addition a history of recent sensitizing events
that patients transplanted preemptively may have lower should be elicited (blood transfusions, immunosuppression
adherence to immunosuppression.1–3 Preemptive trans- withdrawal in someone awaiting a retransplant). These
plantation is particularly advantageous in children.4,5 should be brought to the attention of the histocompat-
Renal transplant recipients are frequently elderly, with ibility laboratory. The patient may require dialysis before
other comorbidity (e.g., diabetes, cardiovascular disease, going to surgery because of fluid overload or a high serum
obesity), which increases the surgical and anesthetic chal- potassium concentration; this will depend on the nature of
lenges. In addition most have impaired platelet function dialysis and when this was last carried out. Potassium often
through a combination of uremia and antiplatelet ther- rises as a consequence of anesthesia, blood transfusion, and
apy (aspirin or clopidogrel), and some will be on warfarin reperfusion of the kidney; it is essential to ensure that the
for previous thromboembolic disease or prosthetic heart patient has a normal serum potassium pretransplant. It is
valves. Thus this group of patients carries a relatively high much easier and safer to dialyze a patient before transplant
operative risk. than immediately posttransplant.

157
158 Kidney Transplantation: Principles and Practice

The potential risks of a renal transplantation should After induction of anesthesia, a central venous cath-
be discussed with patients at the time of listing and eter (CVC) may be inserted into the internal jugular vein
must include general risks of surgery and specific risks (preferably under ultrasound guidance) to allow cen-
of the procedure. These risks include technical compli- tral venous pressure monitoring to ensure optimal fluid
cations such as arterial or venous thrombosis, bleeding replacement and postoperative dialysis, although in some
or urinary complications, risk of delayed graft function centers, the use of CVC may be reserved for those patients
(which can occur in up to 50% deceased donor kidney who will receive antithymocyte globulin (ATG) induction.
transplants), and of acute rejection. Careful explanation In patients without hemodialysis access the selected cath-
of the possible requirement for biopsy should be given. eter should enable postoperative hemodialysis if required.
The need for immunosuppression, with its attendant Subclavian vein cannulation should be avoided if possible
drug-specific and immunosuppression-associated side because of the risk of causing subclavian venous stenosis,
effects, should also be discussed at the time of listing. Any which would prejudice future upper-limb vascular access
recipient choices as to the nature of donors considered when the kidney has failed. Other aspects of the induction
acceptable or not acceptable should be recorded (e.g., of anesthesia and monitoring during the operative proce-
donors with a history of cancer or high risk behavior for dure are discussed in Chapter 13.
hepatitis or HIV). At the time of admission for transplant, Once the patient is anesthetized, a urinary balloon cath-
specific risks pertaining to the donor should be discussed eter is inserted aseptically into the recipient’s bladder (see
with the patient where appropriate. Informed consent to later). The skin should be prepared carefully in the operat-
proceed can then be obtained. ing room; body hair is removed and the skin of the abdomi-
Immunosuppression may be commenced before the nal wall prepared with an antimicrobial agent, typically
patient goes to surgery. Although there is no hard evi- chlorhexidine gluconate in alcohol.10 It is wise to prepare
dence that preoperative immunosuppression is necessary, the entire abdomen from nipples to midthighs, especially
many centers prefer a loading dose of a calcineurin inhibi- in a recipient with vascular disease, because occasionally
tor or antimetabolite to ensure a better blood level in the the original incision may need to be extended or abandoned
first hours posttransplant. Induction agents (most typi- and the opposite iliac fossa opened, or saphenous vein har-
cally basiliximab) are also started preoperatively. Where vested to manage a vascular problem.
the recipient is receiving an antibody-incompatible graft Immediately before surgery, the World Health Organi-
(typically from a living donor), he or she will usually have zation (WHO) surgical safety check should be undertaken,
received several days of preoperative immunosuppression giving additional consideration to checking blood group
in addition to undergoing antibody removal. compatibility of donor and recipient and confirming the
Prophylaxis against deep vein thrombosis and pul- HLA crossmatch status. It is also important to ensure that
monary embolus should be undertaken with low dose the correct donor kidney is in the operating room.
molecular weight heparin according to hospital protocol,
thromboembolic deterrent (TED) stockings, and periopera-
tive intermittent calf compression. Preparation of Kidney
Although the transplant operation is a clean one, the
patient will be immunosuppressed and is at high risk for The preparation of the deceased donor kidney should be
wound infection. In addition it is possible for the deceased done in advance of the transplant procedure in case some
donor kidney to be contaminated during retrieval or for anomaly (for example, a previously unrecognized tumor) is
there to be a urinary tract infection as a consequence of the present that would preclude transplantation, or damage is
donor having a urethral catheter while on the intensive care found that requires repair. Good preparation will ensure an
unit before death. Infection in the vicinity of the vascular adequate length of vessels with good hemostasis at the time
anastomosis may result in secondary hemorrhage, which of reperfusion, both in living and deceased donor kidneys.
is an uncommon but catastrophic complication, resulting A varying degree of dissection of the kidney is required
in loss of the kidney, compromise of distal circulation, and when the kidney is removed from cold storage. In the case
significant mortality. Prophylactic antimicrobial therapy, of a deceased donor kidney removed as part of an en bloc
with a spectrum to cover common skin organisms as well procedure, considerable dissection needs to be performed,
as possible urinary tract contaminants, has been shown to and this should be done carefully and with a good light on a
reduce the risk of developing sepsis with bacteremia and back table with the kidney in a bowl of ice slush.
for developing bactiuria6; a single dose may be sufficient.7 The kidney should be oriented as it is in the body. The
Antimicrobial cover may also be required if the donor was renal vein should be picked up and dissected toward the
known to be infected, such as donors dying from meningo- kidney, dividing small tributaries, and ligating and divid-
coccal meningitis; the advice of a specialist in microbiology ing the gonadal and adrenal veins. Dissection should not
is valuable in these situations. continue close to the renal pelvis. If there is more than one
Consideration must be given at this stage to the cold renal vein, smaller veins can be ligated, assuming that there
ischemia time, which should be minimized. Prolonged cold is one large renal vein. If two renal veins are of equal size
ischemia is associated with increased rates of delayed graft and are not arising from a single caval patch, there is a risk
function and worse long-term outcomes, particularly in of subsequent venous infarction if one vein is ligated; it is
transplants from donors after circulatory death.8 Logisti- preferable to implant both veins separately or to join the
cal factors such as preoperative dialysis and the need for veins to form a common trunk for a single anastomosis. A
a prospective crossmatch, contribute significantly to cold short right renal vein can be extended with donor inferior
ischemic times.9 vena cava or external iliac vein.
11 • Surgical Techniques of Kidney Transplantation 159

Once the venous preparation is complete, the renal vein Where combined pancreas and kidney transplantation is
may then be folded over the kidney and the artery dissected performed via a vertical midline transperitoneal approach,
out from adherent fat. Care must be taken to define the arte- the pancreas is usually placed in the right iliac fossa and
rial anatomy, ensuring that no polar arteries are damaged the kidney in the left iliac fossa. To prevent torsion of the
in the process. In particular, the right renal artery typically renal pedicle, the kidney is best placed in the retroperitoneal
gives a branch to the adrenal which may occasionally pass space, which is accessed by inserting an index finger into
on to supply the upper pole of the kidney. The ureter can now the prevesical space just lateral to the midline and develop-
be dissected free, with care not to divide the tissue between ing the plane laterally.16
the ureter and lower pole of kidney (the so-called golden tri-
angle, see Chapter 29) and to retain a small amount of tis-
sue around the ureter to preserve its blood supply. Incision
Once the kidney has been prepared, the perinephric fat
can be removed. The kidney may then be flushed with 100 There are two common incisions used to expose the exter-
to 200 ml of kidney perfusion fluid, as this may remove nal iliac vessels and bladder. The oblique Rutherford Mori-
products of metabolism and allows detection of defects in son or curvilinear incision is made in the right or left lower
the artery or vein, which should be repaired at this stage. quadrant of the abdomen beginning almost in the midline
2 cm above the pubic tubercle and curving upward and 2
cm parallel to the inguinal ligament and ending just above
Site the anterior superior iliac spine of the iliac crest. Caution
is taken to avoid the lateral cutaneous nerve of the thigh,
Although traditionally the right iliac fossa was used for which emerges through external oblique 1 cm medial to
implantation of the kidney,11–15 in reality there is little to the anterior superior iliac spine. In a child or small adult,
choose between sides. It has been suggested that the left kid- this incision can be carried up to the costal margin to
ney is best placed in the right iliac fossa and the right kid- increase exposure.17 The external oblique muscle and fas-
ney in the left iliac fossa, an approach that places the pelvis cia are divided in the line of the incision and split to the
and ureter medially to facilitate future ureteric reconstruc- lateral extent of the wound. This incision is carried medi-
tion, should it be required. In contrast, placing the deceased ally on to the rectus sheath to permit retraction or division
donor kidney on the ipsilateral side with anastomoses to the of part of the rectus muscle for later exposure of the blad-
external iliac vessels avoids crossing the renal artery and der. To expose the peritoneum, the internal oblique and
vein, and may be facilitated by the use of a subrectus pouch transverse muscles are divided with cautery in the line of
(see later); similarly, live donor kidneys implanted using the the incision.
internal iliac artery may be placed contralaterally to avoid The Alexandre or pararectal incision is slightly more
vessels crossing. In general, however, it is reasonable to use vertical than that of Rutherford Morison. It starts 2 cm
either iliac fossa for placement of the kidney. above the pubic symphysis and passes laterally and crani-
Other factors which may dictate the optimal site of place- ally along the edge of the rectus sheath, two finger breadths
ment include the existence of previous abdominal incisions, medial to the anterior superior iliac spine. The confluence
particularly if placement of a transplant incision would of the oblique abdominal muscles just lateral to the rectus
result in devitalizing an area of abdominal wall. Previous sheath (the Spigelian fascia) is divided to expose peritoneum
venous thrombosis in one leg is an indication to use the beneath.
opposite side lest the thrombus has obliterated the ipsilat- Once the peritoneum is exposed, the inferior epigastric
eral iliac veins. A history of femoral vein cannulation is also vessels may be ligated and divided to improve access, but
a relative contraindication to use that side for fear of iliac if there are multiple renal arteries, the inferior epigastric
venous thrombosis or partial thrombosis. vessels should be preserved in the first instance in case the
If a colostomy or ileostomy were emerging from one side inferior epigastric artery is required for anastomosis to a
of the abdomen, the contralateral side would usually be lower polar renal artery. It may also be wise to preserve the
chosen, although it would be preferable to use the same side vascular supply to the rectus muscle if the muscle has been
as an ileal conduit (urostomy) to facilitate ureteric implan- divided in previous surgery, for example, during a subcos-
tation. The presence of large polycystic kidneys may dictate tal incision to remove an ipsilateral kidney, gallbladder, or
which side is chosen, as there may only be room to implant spleen. Although division of the spermatic cord was advo-
a kidney on one side. Occasionally the polycystic kidneys cated in early descriptions of the procedure and was com-
are too large to permit placement of a transplant. Such a mon practice for many years, it should not be done and is
situation should be picked up as part of the assessment pro- rarely required for adequate exposure. The spermatic cord
cess and one or both polycystic kidneys should be removed may be freed laterally, which allows it to be retracted medi-
before the patient is activated on the transplant list. Poly- ally. In females the round ligament can be divided between
cystic nephrectomy is best done either laparoscopically or ligatures.
through a midline incision; a transverse incision is contra-
indicated to avoid later concerns regarding skin viability
when performing the transplant incision. Preparation of Operative Bed
In children, in whom the vascular anastomoses of the
renal vessels may be to the aorta and vena cava because After exposure of the transversalis fascia and peritoneum,
of the size of the kidney, the right side is preferred because the transversalis fascia is divided, and the peritoneum is
the kidney is placed behind the cecum and ascending colon. reflected upward and medially to expose the psoas muscle
160 Kidney Transplantation: Principles and Practice

and the iliac vessels. This is best done in a caudal to cranial


direction. At this stage, a self-retaining retractor is inserted
to provide good exposure while allowing the assistant to
have both hands free to assist with the anastomosis. Dis-
section proceeds in the first instance to expose the external,
common, and/or internal iliac arteries, depending on cir-
cumstance: the external iliac artery is the site of first prefer-
ence if there is a healthy Carrel patch on the donor renal
artery; the internal iliac artery may be considered if not,
and the common iliac artery if it is a second transplant or
if there is significant arterial disease affecting the external
iliac vessels. Considerations in children are discussed later.
The lymphatics that course along the vessels are preserved
where possible and separated from the artery without divi-
sion. It has been suggested that lymphatics should be ligated
and not cauterized when being divided, because this is said
to prevent the later occurrence of a lymphocele, although
what evidence there is does not support that suggestion (see
Chapter 28).18 The surgeon must be careful not to mistake
the genitofemoral nerve for a lymph vessel. It lies on the
medial edge of the psoas muscle, and a branch may cross
the distal external iliac artery. If the internal iliac artery is to
be used, it is important to mobilize a length of the common Fig. 11.1 Iliac vessels dissected free.
and external iliac arteries so that the internal iliac artery
can be rotated laterally without kinking at its origin and so
that the vascular clamps can be applied to the common and
external iliac arteries when the internal iliac artery is short. Revascularization
Care is taken to inspect the origin of the internal iliac artery,
if this is to be used, for any evidence of atheroma and, simi- Anomalies of the renal artery or vein are common,
larly, any atheromatous disease in the common or exter- amounting to 30% deceased donor kidneys retrieved.20
nal iliac artery should be noted. If there are two or more In living donation, whereas kidneys are selected pref-
renal arteries not on a Carrel patch of aorta, the dissection erentially to have a single artery, multiple arteries are
of the internal iliac artery is extended distally to expose the common. Because the renal arterial inflow comprises end
initial branches of the internal iliac artery, which may be arteries with no intrarenal communication, all arteries
suitable for anastomosis to individual renal arteries. This need to be perfused, but particularly the lower-pole artery,
can be done either in situ, or the bifurcation of the inter- because it is likely to give rise to the ureteric blood sup-
nal iliac removed and a back table anastomosis of the two ply. If multiple arteries are present and separate (i.e., not
renal arteries onto the divisions of the internal iliac artery on a common Carrel patch), there are several surgical
performed, before the kidney is implanted using the resected techniques that can be used: The vessels can be spatu-
portion of recipient internal iliac as an interposition graft.19 lated together to form a common trunk (Fig. 11.2),17 the
Having completed the exposure of the appropriate iliac arter- internal iliac artery can be removed from the recipient and
ies, dissection of the external iliac vein is performed (Fig. 11.1). its branches used to anastomose to the renal arteries on
If a left kidney with a long renal vein is available, dissection the back table,19 a smaller artery may be anastomosed
of the external iliac vein alone generally allows a satisfactory end-to-side to the larger main renal artery, a small acces-
anastomosis without tension. Uncommonly, if the kidney has sory artery can be anastomosed to the inferior epigastric
a very short renal vein, such as with a right kidney or occa- artery, or the renal arteries can be implanted separately
sionally a left kidney whose vein has been shortened, or if the into the external iliac artery. A small upper polar artery, if
recipient is obese, the internal iliac vein and usually one or thought to be too small to anastomose safely to the major
two gluteal veins can be ligated and divided. This technique renal artery, may be ligated, provided that it supplies less
allows the common and external iliac veins to be brought well than one-eighth of the kidney (this should be evident on
up into the wound, particularly if the internal iliac artery is perfusion of the kidney after removal).
divided, and this facilitates the performance of a tension-free A deceased donor kidney usually has a renal artery or
anastomosis. However, division of the internal iliac and glu- arteries arising from a single aortic patch, and this patch
teal veins is not without risk, because slippage of the ligature should be trimmed to an appropriate size and used for
may result in hemorrhage that is difficult to control. Alter- anastomosis to the external iliac artery. If two renal arter-
native means of managing short renal veins are preferred, ies are widely separated on the aortic patch, the patch may
including use of the parachute technique for venous anasto- be divided to allow separate implantation into the external
mosis, a more distal placement on the external iliac vein, or iliac artery, or the two separate patches joined together to
use of a segment of donor inferior vena cava to lengthen the form a shorter patch, or one may be implanted end-to-side
renal vein. Temporary placement of the cold kidney graft into to the external iliac artery and the other to the internal iliac
the wound assists in the selection of the sites for anastomosis artery, or both may be implanted to separate branches of
on the recipient artery and vein. the internal iliac artery.
11 • Surgical Techniques of Kidney Transplantation 161

End-to-end End-to-side

Internal
Iliac Artery

Two renal arteries Two renal arteries


on a patch separate anastomoses

Ligated trunks
of artery
Fig. 11.3 Anastomosis of two renal arteries to the external iliac artery. (From
CIA Lee HM. Surgical techniques of renal transplantation. In: Morris PJ, editor. Kidney
transplantation. London: Academic Press/Grune & Stratton; 1979. p.149.)
EA

the side of the glove.22 This technique not only keeps the
B kidney cool during the anastomosis, but also facilitates
A
handling of the kidney.

Two arteries joined Interposition y-graft of ARTERIAL ANASTOMOSIS


as a pantaloon native internal iliac artery
Fig. 11.2 Variations of renal artery anastomoses. (From Lee HM. Surgical
External Iliac Artery
techniques in renal transplantation. In: Morris PJ, ed. Kidney Transplanta- An end-to-side anastomosis of the renal artery to the exter-
tion. London: Academic Press/Grune & Stratton; 1979. p.150.) nal iliac artery (or common iliac artery) usually is performed
using an appropriately trimmed cuff of aorta attached to the
Before making the arteriotomy or venotomy, the sur- renal artery (the Carrel patch) (Fig. 11.3). Vascular clamps
geon should mentally visualize the kidney in situ in its are applied to the external iliac artery proximally and dis-
final resting place, as well as picturing the course that the tally if an end-to-side anastomosis is to be performed, with
renal artery and vein would take to ensure the optimal site care taken to avoid clamping diseased segments of artery
for the anastomosis. Where the renal artery is much lon- wherever possible. An arteriotomy appropriately placed
ger than the vein, it may either be electively anastomosed is performed in the external iliac artery, and the lumen is
on the internal iliac artery or, more simply, the artery can flushed out again with heparinized saline; where the donor
be anastomosed to the external iliac artery but the kidney artery has no Carrel aortic patch, a hole punch is used to
placed in a subrectus pouch fashioned by dissecting the create a suitably sized hole for anastomosis.
peritoneum from the underside of the rectus muscle.21 In The anastomosis is done with a continuous 5-0 or 6-0
such a position, the longer artery tends to run a smooth monofilament vascular suture (see Fig. 11.2),17 although
course. an interrupted technique may be necessary where no Car-
When the kidney has been prepared and is ready for rel patch exists. In older patients and those who have been
implantation, the vessels are now ready for clamping. Hep- on dialysis some time, the intima may be calcified and may
arin may be administered in a modest dose (e.g., 30–60 be easily displaced from the wall of the artery. Particular
IU/kg), although many surgeons simply cross-clamp the care should be taken to ensure that all the intima on the
recipient vessels without heparinization in patients recipient artery is secured back in position during the anas-
already on dialysis. The kidney should be kept cold during tomosis to prevent a dissection propagating along the distal
the implantation phase. This may be achieved in a num- artery on reperfusion. In very severe cases of calcification
ber of ways, such as wrapping in a surgical gauze swab of the recipient artery, it may be necessary to carry out a
filled with crushed frozen saline. Another technique uses a formal endarterectomy of the iliac artery, with the distal
surgical glove to contain the kidney together with crushed intima stitched in place to prevent formation of a flap and
ice, the vessels being brought out through a small cut in subsequent dissection.
162 Kidney Transplantation: Principles and Practice

Internal Iliac Artery proximally and distally with vascular clamps or a Satin-
The internal iliac artery should not be used for anastomosis if sky side clamp is used. The venotomy is flushed out with
the contralateral internal iliac artery has already been used heparinized saline. Where possible the site of venotomy
for a previous transplant or if the contralateral limb relies on should be proximal or distal to a valve, and if a valve is
collaterals from the ipsilateral internal iliac for its perfusion present at the site of the venotomy, it should be removed
(for example, where the contralateral common iliac artery is carefully. The anastomosis is fashioned using a continuous
occluded). If the internal iliac artery is to be used, its distal 5-0 monofilament vascular suture, with the initial sutures
branches are ligated. A vascular clamp is applied to the inter- placed at either end of the venotomy (Fig. 11.5).17 A use-
nal iliac artery close to its origin (to reduce the chances of clot ful aid when doing the venous anastomosis is to place an
forming in the occluded stump) or to both the common and anchor suture at the midpoint of the lateral wall, which
external iliac arteries. The internal iliac artery is then divided allows the external iliac vein and the renal vein on the lat-
close to or at the bifurcation maximizing its length, and the eral side of the anastomosis to be drawn clear of the medial
lumen is flushed out with heparinized saline. wall of the anastomosis (Fig. 11.6). This technique reduces
The internal iliac artery is anastomosed end-to-end to the the risk of the back wall being caught up in the suture while
renal artery with 5-0 or 6-0 monofilament vascular suture the medial wall is being sutured. An alternative, and one
using a three-point anastomosis technique, as described by suited to larger patients, is to use the parachute technique,
Carrel in 1902,23 or a two-point anastomosis (Fig. 11.4)17; placing several sutures at the cranial aspect of the medial
alternatively, the parachute technique may be used, only suture line before parachuting the anastomosis down. This
tying the sutures after first placing all the sutures individually. has the benefit of distributing the tension over a wider area
If there is a disparity between the renal artery and the inter- of vein, so there is less likelihood of the suture pulling out.
nal iliac artery, the renal artery being considerably smaller in The renal vein is usually anastomosed to the external
diameter, the renal artery may be spatulated along one side iliac vein medial to the external iliac artery, although on
to broaden the anastomosis. If one side of the renal artery is occasion it may be lateral to the artery. Wherever the anas-
spatulated, care should be taken to place the spatulation of tomosis is positioned, it is important to ensure that the renal
the renal artery appropriately, taking into consideration the vein is under no tension, and care should be taken that the
final curve of the internal iliac artery and the renal artery vein is not twisted before starting the anastomosis. When a
to avoid kinking when the kidney is placed in its final posi- small child receives an adult kidney, it is sometimes neces-
tion.17 If both arteries are small, the anastomosis should be sary to shorten the renal vein to prevent kinking, especially
performed with interrupted sutures to allow for expansion. In when the vein is anastomosed to the inferior vena cava.
a child or a small adult with small arteries, the whole anasto- If the venous anastomosis is fashioned before the arterial
mosis should be performed with interrupted sutures. anastomosis (such as when the external iliac artery is to be
used), it may be desirable to remove the venous clamps to per-
mit return of blood from the leg, so shortening the duration
VENOUS ANASTOMOSIS of venous stasis. This is best achieved by placing a separate
The renal vein is anastomosed end-to-side, usually to fine bulldog clamp close to the anastomosis on the renal vein
the external iliac vein. The external iliac vein is clamped before removing the iliac vein clamps, so preventing reflux of

Fig. 11.4 Internal iliac (hypogastric) artery ligated and divided, the lumen flushed with heparinized saline. (From Lee HM. Surgical techniques of renal
transplantation. In: Morris PJ, editor. Kidney transplantation. London: Academic Press/Grune & Stratton; 1979. p.148.)
11 • Surgical Techniques of Kidney Transplantation 163

blood into the kidney. It is important that the bulldog clamp There is no intimal dissection of the proximal recipient
□ 

is not traumatic to the vein, and does not slip off the vein— artery or the donor renal artery, the latter being a conse-
two clamps are often better than one to ensure the latter. This quence of traction in the donor or extreme donor hyper-
maneuver also allows any bleeding from the venous anasto- tension during coning.
  
mosis to be managed before the kidney is revascularized.
Finally, if concern still exists, the Hume test can be reas-
The question of whether the arterial anastomosis or the
suring. When the renal vein is occluded between finger and
venous anastomosis should be done first depends on the
thumb, the kidney should swell and throb. When the vein
final position of the kidney and the ease with which the sec-
is released the kidney palpably softens as the turgor goes.
ond anastomosis may or may not be done. If the renal artery
is to be anastomosed end-to-side to the external iliac artery
(usually with a Carrel patch of aorta), it is preferable to do Reconstruction of the Urinary
the venous anastomosis first, then the end-to-side arterial
anastomosis can be positioned correctly. If the renal artery Tract
is to be anastomosed to the internal iliac artery, the arte-
Once the kidney is perfused with recipient blood and
rial anastomosis may be done first because this enables the
hemostasis has been secured, reconstruction of the uri-
renal vein to be positioned appropriately.
nary tract is carried out. Transplantation of the left kid-
ney into the right iliac fossa and the right kidney into the
REPERFUSION OF THE KIDNEY left iliac fossa reverses the normal anterior-to-posterior
relationship of the vein, artery, and collecting system and
Vascular clamps are removed sequentially, starting either
positions the renal pelvis and ureter of the kidney trans-
with the venous clamps (proximal first then distal), or the
plant so that they are the most medial and superficial of
arterial clamps (distal first then proximal), depending on
the hilar structures.13 This positioning simplifies primary
surgeon preference. Once the kidney is reperfused, atten-
(and secondary) urinary tract reconstruction if pyeloure-
tion should be paid to controlling significant bleeding points
terostomy, ureteroureterostomy, or pyelovesicostomy
on the anastomoses and ligating any tributaries that were
need to be done. The factors that determine the type of
missed during the back table preparation. The quality of
urinary tract reconstruction are the length and condi-
reperfusion is variable. Live donor kidneys and kidneys that
tion of the donor ureter, the condition of the recipient’s
have been subject to machine preservation reperfuse evenly
bladder or bladder substitute, the condition of the recipi-
and become pink very quickly. Deceased donor kidneys,
ent’s ureter, and the familiarity of the surgeon with the
particularly those with prolonged cold ischemia or those
technique.
donated after circulatory death, tend to be patchy for some
Suture material is an individual choice. Although uri-
time. Although this usually resolves over time, it is impor-
nary tract reconstruction with nonabsorbable sutures has
tant to ensure the following:
   been described,24,25 it leaves the recipient with the risk of
All the clamps have been removed.
□  stone formation. Modern synthetic absorbable monofila-
The artery is not twisted.
□  ment sutures (e.g., polyglyconate and polydioxanone) have
The recipient has a good blood pressure.
□  characteristics suitable for the immunocompromised kid-
ney transplant recipient in whom delayed wound healing
is possible.

URETERONEOCYSTOSTOMY (ANASTOMOSIS OF
THE TRANSPLANT URETER DIRECTLY TO
THE BLADDER)
This is the usual form of urinary tract reconstruction. Its
advantages are as follows:
  

It can be performed regardless of the quality or presence


□ 

of the recipient ureter.


It is several centimeters away from the vascular anasto-
□ 

moses.
The native ureter remains untouched and therefore
□ 

available for the future management of ureteric compli-


cations.
Native nephrectomy is unnecessary.
□ 
  

The goal is to anastomose the ureter to the mucosa of the


bladder, with the distal ureter surrounded in a 2- to 3-cm
tunnel so that, when the bladder contracts, there is a valve
mechanism to prevent reflux of urine up the ureter.26–28
The efficiency of this antireflux mechanism is variable.
The urinary catheter is connected to a Y connector with
Fig. 11.5 Vein anastomosis with triangular stay sutures in place. (From
Lee HM. Surgical techniques of renal transplantation. In: Morris PJ, editor. Kid- a bag filled with saline and an antibiotic and/or methy-
ney transplantation. London: Academic Press/Grune & Stratton; 1979. p.151.) lene blue dye on one line and a urinary collection bag on
164 Kidney Transplantation: Principles and Practice

A B
Fig. 11.6 Color photo of (A) preparation of external iliac vein and (B) venous anastomosis.

of a proposed vertical midline incision. The urinary blad-


der is drained, and an incision is made through all layers
of the anterior bladder wall. A retractor is placed into the
dome of the bladder to expose the trigone. A point clear
of the native ureter is selected, and a transverse incision
is made in the mucosa. A submucosal tunnel is created
with a right-angle clamp or small scissors for about 2
cm. The clamp or scissors is pushed through the blad-
der from inside to outside, and the muscular opening is
enlarged to accept the kidney transplant ureter. The ure-
ter is drawn into the bladder, where it is transected at a
length that prevents tension or redundancy. The cut end
of the ureter is incised for 3 to 5 mm and approximated
to the bladder mucosa with fine absorbable sutures. The
inferior suture includes the bladder muscle to fix the ure-
ter distally and to prevent its movement in the submuco-
sal tunnel. The retractor is removed, and the cystotomy
is closed with a single or double layer of 3-0 absorbable
suture. The bladder can be refilled to check for leakage,
and points of leakage can be repaired with interrupted
sutures. Some surgeons use two bladder mucosal inci-
sions about 2 cm apart33; when this technique is used,
the proximal bladder mucosal incision is closed with a
Fig. 11.7 Y-tube system for rinsing, filling, and draining bladder or fine absorbable suture.
bladder substitute. (From Kostra JW. Kidney transplantation. In: Kremer
B, Broelsch CE, Henne-Bruns D, editors. Atlas of liver, pancreas, and kidney Extravesical Ureteroneocystostomy
transplantation. Stuttgart: Georg Thieme Verlag; 1994. p.128.)
This is the most common technique used. Although not
producing such a “physiologic” antireflux mechanism
the other (Fig. 11.7).29 The use of an antibiotic in the solu- as the transvesical method, extravesical techniques are
tion reduces the risk of postoperative urinary infection,30,31 faster, do not require a separate cystotomy, and require
whereas the dye reassures the surgeon it is the bladder that less ureteric length, are associated with fewer urinary
has been opened and not another viscus. With this system, tract infections, leaks, and less hematuria than intravesi-
the bladder can be filled, drained, and, if necessary, refilled cal techniques (Fig. 11.9).34,35 Extravesical techniques are
during the procedure. It is especially helpful when the based on the procedure described by Lich and colleagues.26
bladder is difficult to identify because of pelvic scar tissue, Extravesical ureteroneocystostomy was adapted for renal
recipient obesity, or reduced capacity. After initially accom- transplantation by Woodruff in 1962,36 and it is well illus-
modating a small volume, the defunctioned bladder often trated by Konnak and colleagues (see Fig. 11.9).37 A sub-
accepts more fluid 1 or 2 hours into the transplantation sequent modification was the addition of a stitch to anchor
procedure.32 the toe of the spatulated ureter to the bladder to prevent
proximal slippage of the ureter in the submucosal tunnel
Transvesical Ureteroneocystostomy with loss of the antireflux valve and disruption of the ure-
The traditional technique for transvesical ureteroneo- teric anastomosis.38,39
cystostomy is similar to that described by Merrill and col- A double-pigtail (double-J) ureteric stent reduces the
leagues13 in the first successful kidney transplant from a incidence of leak and stenosis40–42 and is widely used.
twin (Fig. 11.8). The dome of the bladder is identified, and This is passed retrograde up the donor ureter, after first
stay sutures or Babcock clamps are placed on either side cutting the ureter to a suitable length and spatulating its
11 • Surgical Techniques of Kidney Transplantation 165

Fig. 11.8 (A–D) Transvesical ureteroneocystostomy. (From Lee DM. Surgical techniques of renal transplantation. In: Morris PJ, editor. Kidney transplantation.
London: Academic Press/Grune & Stratton; 1979. p.153.)

end. The bladder is distended with an antibiotic/dye solu- wound, and the mucosal sutures passed through the toe
tion through the urethral catheter. The lateral surface of and heel of the spatulated end, and the ureter parachuted
the bladder is cleared of fat and the peritoneal reflection, on to the bladder. The ureter is then anastomosed to the
a retractor is placed medially, another is placed infero- bladder mucosa with running sutures between the ureter
laterally, and a third retractor is placed cephalomedially and the mucosa of the bladder; some surgeons take a small
to hold the peritoneum and its contents out of the way. amount of bladder muscle in the suture, whereas others
Some authors recommend placing the ureter under the suture to mucosa alone. Some authors recommend specifi-
spermatic cord or round ligament, believing that this pre- cally anchoring the toe of the ureter with a horizontal or
vents posttransplant ureteric obstruction. A longitudinal vertical mattress suture placed in the toe of the ureter and
oblique incision is made for approximately 2 cm until the passed submucosally through the seromuscular layer of
bladder mucosa bulges into the incision. The bladder is the bladder and tied about 5 mm distal to the cystotomy
partially drained via the urethral catheter, and the mucosa (Fig. 11.10). When handling the ureter and bladder, care
is dissected away from the muscularis on both sides to should be taken to avoid crushing the delicate mucosa
facilitate later creation of a submucosal tunnel for the ure- with forceps. Once the ureteric anastomosis is complete,
ter. The bladder mucosa is incised and 5-0 monofilament the seromuscular layer is closed over the ureter with inter-
absorbable sutures (e.g., polydioxanone) placed through rupted absorbable sutures, care being taken to avoid nar-
both ends of the incision. The ureter is brought up to the rowing the ureter in the process.
166 Kidney Transplantation: Principles and Practice

Parallel Incision Ureteroneocystostomy


A variant of the extravesical ureteroneocystostomy is the
parallel incision ureteroneocystostomy.43 It involves two
parallel vertical incisions in the bladder muscle some 2 cm
apart. The ureter is tunneled submucosally through the lat-
eral incision and out of the medial one. The ureter is then
cut, spatulated, the bladder mucosa opened at the site of the
medial incision, and the heel of the ureter sutured to the
mucosa while the toe is sutured to both mucosa and muscle
wall (Fig. 11.11). The muscle is then closed over the both
incisions with an absorbable suture.44

DOUBLE URETERS
Double ureters can be managed simply by trimming them
to appropriate length, spatulating them, and either anas-
tomosing the medial edges together with a continuous
or interrupted fine absorbable suture (Fig. 11.12)45,46 or
joining them, one on top of the other, with a single stitch
from the toe of the upper one to the heel of the lower one.47
The conjoined ureters can be treated as a single ureter
by any of the previously described ureteroneocystostomy
techniques. The submucosal tunnel needs to be made a bit
wider. The alternative approach is to use a separate ure-
teroneocystostomy for each of the ureters.48 These same
techniques can be used for the en bloc transplantation of
pediatric kidneys or the transplantation of two adult kid-
Fig. 11.9 (A–C) Extravesical ureteroneocystostomy. (From Konnak JW, neys, stacked one on top of the other,49 into one recipient.
Herwig KR, Turcotte JG. External ureteroneocystostomy in renal transplan- Fjeldborg and Kim50 described a pyeloureteric anastomosis
tation. J Urol 1972;108:380.) for a kidney with double ureters in which both renal pelves
are joined after dividing the ureters at their ureteropelvic
junctions and suturing the posterior walls together, leav-
ing the anterior halves for anastomosis with the recipient
ureter (Fig. 11.13).50 If both ureters have been stented it
is important to ensure that both stents are subsequently
removed.

AUGMENTED BLADDER
In patients with congenital bladder abnormalities, the
bladder may have been augmented as part of previous
treatment or in preparation for transplantation. It is
important to know the anatomy and blood supply of an
augmentation patch so as not to interfere with it during
the kidney transplant procedure. Ideally the ureter should
be anastomosed to the bladder itself, with a submucosal
tunnel for ureteroneocystostomy. Where ileum or cecum
has been used, and is the most readily accessible compo-
nent of the reconstructed bladder, the donor ureter may be
anastomosed without a tunnel, and the anastomosis man-
aged in a similar fashion used for an ileal conduit. Ureteric
stents are usually used.

PYELOPYELOSTOMY
Pyelopyelostomy has been used for orthotopic kidney
Fig. 11.10 One or two mattress sutures to anchor toe of transplant transplantation, usually in the left flank.51 The native
ureter to full-thickness bladder. This prevents ureteric slippage in the kidney is removed, and the kidney transplant is revascu-
submucosal tunnel. (From Hinman Jr F. Ureteral reconstruction and exci-
sion. In: Hinman Jr F, editor. Atlas of urologic surgery. 2nd ed. Philadelphia: larized with the native renal artery or the splenic artery
WB Saunders; 1998. p.799.) and the native renal vein. The proximal ureter and renal
11 • Surgical Techniques of Kidney Transplantation 167

A B

C D
Fig. 11.11 (A–D) Parallel incision ureteroneocystostomy. (From Knechtle S. Ureteroneocystostomy for renal transplantation. J Am Coll Surg 1999;188(6):707–9.)

pelvis of the kidney transplant are opened medially, and


the native renal pelvis is anastomosed to the kidney trans-
plant renal pelvis with a running fine absorbable suture.
After completion of one wall, a double-pigtail ureteric
stent is passed with or over a guidewire through the native
ureter into the bladder, and the wire is withdrawn to allow
the distal end to curl within the bladder. Its position in the
bladder is confirmed by reflux of bladder irrigant up the
stent. The proximal coil is placed in the renal pelvis of the
kidney transplant, and the remaining half of the suture
line is completed. Compared with ureteroneocystostomy,
an advantage of urinary tract reconstruction with the
native renal pelvis or ureter is the ease with which sub-
sequent retrograde pyelography, stent placement, or
ureteroscopy can be accomplished through the normally
positioned ureteric orifice.
Fig. 11.12 Management of double ureters to make them into a single
ureteric orifice.
168 Kidney Transplantation: Principles and Practice

Fig. 11.13 Management of double ureters by pyelopyelostomy fol-


lowed by conjoined pyeloureterostomy. (From Fjeldborg O, Kim CH.
Double ureters in renal transplantation. J Urol 1972;108:377.)

PYELOURETEROSTOMY AND
URETEROURETEROSTOMY
Pyeloureterostomy and ureteroureterostomy usually are
done: (1) when the transplant ureter’s blood supply seems Fig. 11.14 Ureteropyelostomy and ureteroureterostomy. A double-
to be compromised, (2) when the urinary bladder is diffi- pigtail stent is placed after the back wall suture line has been com-
cult to identify because of pelvic scar, (3) when the bladder pleted.
does not distend enough for a ureteroneocystostomy, or
(4) as a result of surgeon preference.52–54 The techniques
for ureteropyelostomy and ureteroureterostomy are simi- kidney. However, with the two caveats mentioned, liga-
lar (Fig. 11.14). The posterior, or back wall, anastomosis is tion of the native ureter for ureteroureterostomy is nor-
completed between the kidney transplant pelvis or ureter mally uneventful.
and the side or to the spatulated end of the native ureter;
a double-pigtail ureteric stent is placed, and the anterior PYELOVESICOSTOMY
suture line is completed. The proximal native ureter is
managed by the following: Pyelovesicostomy has been described for urinary tract
  
reconstruction when the native ureter and the renal trans-
Leaving the native kidney in situ and using the side of the
□ 
plant ureter are unsuitable or become so (Fig. 11.15).57–59
native ureter for the anastomosis The bladder must reach the renal pelvis without tension. To
Ipsilateral nephrectomy and proximal ureterectomy
□ 
achieve this the bladder may be mobilized and hitched to
Ligation of the proximal ureter with the obstructed native
□ 
the psoas muscle or a bladder extension with a Boari flap
kidney left in situ55,56 may be needed.60 This technique may also be useful as a
  

The native ureter should not be ligated in the pres- management of posttransplant ureteric stenosis.
ence of urinary tract sepsis (when a pyonephrosis of the
native kidney may ensue) or where the recipient has pre- URETEROENTEROSTOMY
viously undergone ureteric reimplantation to treat reflux
disease (in which case the blood supply to the ureter may Ureteroenterostomy into an intestinal conduit or an intesti-
be severely compromised). By leaving the native ureter in nal pouch is indicated where the bladder has been removed
continuity with its kidney, and anastomosing the pelvis or or is unusable.61,62 It is preferable to create the conduit
ureter of the renal transplant to the side of the native ure- before the transplant, at the time of listing if it has not
ter, a good blood supply to the native ureter is guaranteed been created previously. The conduit or pouch is washed
without the risk of an obstructed, hydronephrotic native with antiseptic before surgery commences. The ureteric
11 • Surgical Techniques of Kidney Transplantation 169

MANAGEMENT OF CATHETER AND STENT


The urinary bladder or reservoir catheter usually is removed
on postoperative day 5. Some units test the urine at the bed-
side for nitrites and send for bacterial culture. If the urine is
shown to be infected, an antibiotic is chosen based on sen-
sitivity results and is prescribed for 10 to 14 days. Where
the stent has been fixed to the urinary catheter it will come
out as the catheter is withdrawn,64 otherwise the stent is
removed at 6 weeks using flexible cystoscopy. Early stent
removal has the advantage of reducing urinary tract infec-
tions while still reducing other complications.64,65 If a
ureteric stent is in situ it should be removed if infection is
present. Care should be taken to identify all patients with
stents in situ lest one be forgotten.

Closure
Many units obtain a biopsy specimen of the kidney rou-
tinely before closure of the wound (a “time zero biopsy”).
This biopsy can be used to provide baseline histology to
identify chronic changes and any unknown renal disease;
it may also show evidence of ischemia/reperfusion injury
or early antibody-mediated damage, but the time taken for
these to manifest histologically is generally longer than the
average transplant operation (see Chapter 26). Methods of
closing the wound vary, but closure of all musculofascial
layers with a nonabsorbable material such as nylon is pre-
ferred to avoid herniation. Skin closure with a subcuticular
absorbable suture gives the best cosmetic result.
Some surgeons prefer to drain the surgical bed to give
early warning of bleeding or urinary leak, whereas others
Fig. 11.15 Pyelovesicostomy. (From Firlit CF. Unique urinary diversions in argue that the drain is a portal for entry of microorgan-
transplantation. J Urol 1977;118:1043.) isms. If drainage is performed it should be a closed system
and drains should be removed at the earliest opportunity.
anastomosis is done with the spatulated end of the ureter The exit site of the drain should be cleaned and dressed daily
being anastomosed to the full thickness of the bowel wall. until the drain is removed.
If it is difficult to identify the intestinal conduit or pouch The historical practice of capsulotomy of the transplanted
because of surrounding intestines, the addition of methy- kidney, where the renal capsule is carefully split along its
lene blue dye to the irrigant stains the conduit or pouch convex border from pole to pole to minimize damage to the
and may make it easier to find,63 or placing a finger in the kidney as the parenchyma swelled in response to reperfu-
lumen may help. This topic is discussed more completely in sion injury is no longer performed.66,67
Chapter 12.

URETERIC STENTS Pediatric Recipient


Stents have been shown to reduce many of the urologic For older children, the transplant procedure is the same as
complications of ureteric anastomoses but are associated for adults if their weight is more than 20 kg.5,68,69 The renal
with an increased incidence of urinary tract infections.40–42 vessels are anastomosed end-to-side to the iliac vessels or to
Indications for their selective use, where stenting is not the aorta and vena cava.70
routine practice, include edema; a thickened bladder; and In smaller children (weight <20 kg), the right extraperi-
when a pyelopyelostomy, pyeloureterostomy, or uretero- toneal space can be developed by extending the incision to
ureterostomy have been performed; or when the ureter has the right costal margin,71 or a transperitoneal approach can
been anastomosed to an intestinal conduit or pouch. The be used.33 In the case of the latter, the abdomen is opened
ideal length of the stent is determined by the estimated dis- through the midline incision from the xyphoid to the pubis,
tance between the renal pelvis of the kidney graft and the and the retroperitoneum opened by incising the peritoneum
bladder (or its substitute). A double-pigtail 5 French stent lateral to the ascending colon, which is reflected medially.
of 12 cm in length is generally suitable for an adult trans- The terminal portion of the vena cava is dissected over 3
plant kidney located in the iliac fossa and anastomosed to to 4 cm, ligating and dividing two to three lumbar veins
the native bladder. posteriorly. The terminal aorta is also dissected free at its
170 Kidney Transplantation: Principles and Practice

is necessary. Interrupted sutures may be necessary for at


least half the circumference of the anastomosis because the
kidney will increase in size.
When pediatric kidneys are very small, double kidneys
may be transplanted en bloc into adults and bigger chil-
dren.72–76 However there is increasing evidence that even
kidneys from donors as small as 20 kg may be transplanted
singly with good outcomes.77
For en bloc transplantation, both kidneys are removed
with a segment of aorta and vena cava. The aorta and vena
cava caudal to the renal vessels are removed and anasto-
mosed to the aorta and vena cava cranial to the renal ves-
sels, closing off the caudal aorta and cava of the donor. This
technique allows the kidneys to be placed quite low over the
iliac vessels and provides a short distance for the ureters to
traverse to the bladder. Other techniques include simply
oversewing the cranial ends of the aorta and vena cava,
with anastomoses of the caudal ends of the aorta and vena
cava end-to-side to the iliac vessels. Some surgeons advo-
cate suturing the superior poles of the kidneys to the sides
of the aorta to prevent torsion or kinking of renal vascular
Fig. 11.16 Renal transplant in small children (<20 kg). (From Lee HM. pedicles. Ureters are implanted to the bladder separately
Surgical techniques of renal transplantation. In: Morris PJ, editor. Kidney using the extravesical approach or are joined together to
transplantation. London: Academic Press/Grune & Stratton; 1979. p.159.)
form a common funnel, as described earlier. Another tech-
nique is to remove segments of the recipient’s external iliac
bifurcation, as is the right common iliac artery. A partial artery and vein and anastomose the tubular aorta and
occluding clamp is used to isolate the vena cava and aorta, inferior vena cava into the defects. A fourth technique is to
in preference to full mobilization of vena cava and aorta and incise longitudinally the posterior aorta and inferior vena
cross-clamping. The renal vein is anastomosed to the vena cava and anastomose these vascular patches to the iliac
cava first in an end-to-side technique with nonabsorbable vessels.
monofilament vascular sutures (Fig. 11.16).17 The renal
artery is then anastomosed to either the right common iliac
artery or distal aorta in an end-to-side fashion using fine Double Kidney Transplant
nonabsorbable monofilament vascular sutures. Occasion-
ally a small aortic punch may be used to create a hole in the Less than optimal kidneys are increasingly being used for
aorta to which the renal artery is anastomosed. This latter transplantation, accepting less than perfect function in
technique is said to reduce the chance of the renal artery the knowledge that this is, nonetheless, superior to life on
lumen occluding if hypotension occurs. When the renal dialysis.78–80 Some centers routinely biopsy older donor kid-
artery is anastomosed to the aorta it is usually brought neys,81 and if the biopsy shows significant chronic changes
in front of the vena cava, although on occasion it may lie then both donor kidneys are transplanted into one recipi-
best if passed behind the vena cava. Careful liaison with ent, rather than transplanting one each into two separate
the anesthetist is required when clamping and declamping recipients.
the vena cava and aorta, because the perfused kidney may Double kidney transplantation can be performed in two
remove a large part of the child’s circulating volume, result- ways.82 Both kidneys can be transplanted on to the same side
ing in dramatic hemodynamic changes. through an extended pararectal incision. The first kidney
The ascending colon is placed back over the anterior sur- chosen should have the longest ureter, and is transplanted
face of the kidney. No fixation is necessary. The ureter is on to the common iliac artery and either the common iliac
brought down retroperitoneally crossing the common iliac vein or vena cava; the second kidney is then implanted cau-
artery at its midpoint and is implanted into the bladder as a dally onto the external iliac vessels. The ureters may be man-
ureteroneocystostomy. aged as described previously. It is important to transplant
End-to-end arterial anastomoses in growing children the cranial kidney first, because clamping the common iliac
should be done wholly or partially interrupted; for end- to- artery will remove the inflow to a kidney that has been trans-
side anastomoses, where there is a significant Carrel patch planted to the external vessels below. The alternative method
of donor vessel, continuous anastomoses may suffice. of transplantation is to implant kidneys bilaterally through
separate incisions, one in each iliac fossa.

Pediatric Donor
Transplant Nephrectomy
When a child’s kidney is used as a donor kidney for an adult
or child recipient, the surgical technique is essentially the Removal of a graft that has undergone chronic rejec-
same as has been described. Because of the small size of the tion and has been in place for many months or years
renal vessels, use of aortic and vena caval patches generally can be extremely difficult and should be performed by an
11 • Surgical Techniques of Kidney Transplantation 171

experienced transplant surgeon. The usual approach for 4. Cole BR. The psychosocial implications of pre-emptive transplanta-
the transplant nephrectomy is through the original trans- tion. Pediatr Nephrol 1991;5(1):158–61.
5. Fine RN, Korsch BM, Stiles Q, et al. Renal homotransplantation in
plant incision. An abdominal incision may be preferred in children. J Pediatr 1970;76(3):347–57.
small children, particularly if the implantation was per- 6. Green H, Rahamimov R, Gafter U, et al. Antibiotic prophylaxis for
formed intraabdominally. One may also use the abdominal urinary tract infections in renal transplant recipients: a systematic
approach to control the iliac vessels in case of a mycotic review and meta-analysis. Transpl Infect Dis 2011;13(5):441–7.
7. Orlando G, Manzia TM, Sorge R, et al. One-shot versus multidose peri-
aneurysm or a perinephric abscess, where there is a risk of operative antibiotic prophylaxis after kidney transplantation: a ran-
catastrophic hemorrhage. Prophylactic antibiotics should domized, controlled clinical trial. Surgery 2015;157(1):104–10.
be given to cover skin and urinary pathogens, together with 8. Summers DM, Johnson RJ, Allen J, et al. Analysis of factors that affect
coverage of any other organism known to be prevalent. outcome after transplantation of kidneys donated after cardiac death
In the early postoperative period, removal of the trans- in the UK: a cohort study. Lancet 2010;376(9749):1303–11.
9. Shrestha S, Bradbury L, Boal M, et al. Logistical factors influencing
plant in toto is simple with easy identification of the renal cold ischemia times in deceased donor kidney transplants. Transplan-
pedicle structures. The donor vessels may be simply ligated, tation 2016;100(2):422–8.
with the ligated stumps of donor vessels either left in situ in 10. Darouiche RO, Wall MJ Jr, Itani KM, et al. Chlorhexidine-alcohol
the recipient or removed. Leaving stumps runs the risk of versus povidone-iodine for surgical-site antisepsis. N Engl J Med
2010;362(1):18–26.
thrombosis or hemorrhage as the immune system attacks 11. Hume DM, Magee JH, Kauffman HM, et al. Renal homotransplanta-
the donor endothelium, although this is less likely the more tion in man in modified recipients. Ann Surg 1963;158:608–44.
time that has elapsed since transplantation. Alternatively, 12. Küss R, Teinturier J, Milliez P. Quelques essais de greffes du rein chez
removal of the donor vessel stumps may necessitate vas- l’homme. Mem Acad Chir (Paris) 1951;77(22-24):755–64.
cular repair using an autologous saphenous vein patch to 13. Merrill JP, Murray JE, Harrison JH, et al. Successful homotransplanta-
tion of the human kidney between identical twins. J Am Med Assoc
prevent narrowing of the native vessels. 1956;160(4):277–82.
The long-standing transplanted kidney is most easily 14. Michon L, Hamburger J, Oeconomos N, et al. Une tentative de trans-
removed subcapsularly. After deepening the original inci- plantation rénale chez l’homme: aspects medicaux et abiologiques.
sion the capsule is identified and incised. The kidney paren- Presse Med 1953;61(70):1419–23.
15. Starzl TE, Marchioro TL, Dickinson TC, et al. Technique of renal homo-
chyma is freed with blunt dissection all around the kidney transplantation. Experience with 42 cases. Arch Surg 1964;89:87–
in the plane within the capsule but outside the paren- 104.
chyma. The vessels and ureter enter the capsule deep to 16. West MS, Stevens RB, Metrakos P, et al. Renal pedicle torsion after
the kidney, and it is usually necessary to incise the capsule simultaneous kidney-­ pancreas transplantation. J Am Coll Surg
around the hilum so as to isolate the vascular pedicle. Care 1998;187(1):80–7.
17. Lee H. Surgical techniques of renal transplantation. In: Morris P, editor.
should be taken to ensure that the native vessels, typically Kidney transplantation. London: Academic Press/Grune & Stratton;
the external iliac artery and vein, are separate from the 1979. p. 145.
vascular pedicle and are not damaged. The pedicle is then 18. Farouk K, Afridi Z, Bano U, et al. Electrocoagulation versus suture-
mass-clamped with a Satinsky clamp and divided to remove ligation of lymphatics in kidney transplant recipient surgery. J Post-
grad Med Inst 2006;4:398–403.
the kidney. The vascular pedicle is oversewn with a mono- 19. Hiramitsu T, Futamura K, Okada M, et al. Impact of arterial recon-
filament vascular suture. The artery and the vein may be struction with recipient’s own internal iliac artery for multiple graft
dissected and transfixed separately at this time, but this can arteries on living donor kidney transplantation: strobe study. Medi-
be difficult and is usually unnecessary. Sometimes the seg- cine (Baltimore) 2015;94(43):e1811.
mental renal arteries and venous branches are ligated and 20. Watson CJ, Harper SJ. Anatomical variation and its management in
transplantation. Am J Transplant 2015;15(6):1459–71.
divided as they appear during dissection within the renal 21. Wheatley TJ, Doughman TM, Veitch PS, et al. Subrectus pouch for
hilar scar, obviating the need to control the pedicle. The renal transplantation. Br J Surg 1996;83(3):419.
ureter is usually readily identified and followed to the blad- 22. Roake JA, Toogood GJ, Cahill AP, et al. Reducing renal ischaemia dur-
der, where it is excised with any defect in the bladder wall ing transplantation. Br J Surg 1991;78(1):121.
23. Carrel A. Le technique opératoire des anastomoses vasculaires et la
oversewn with an absorbable suture. Transplant nephrec- transplantation des viscères. Lyon Med 1902;98:859–64.
tomy, therefore, leaves a small amount of donor material in 24. Jaffers GJ, Cosimi AB, Delmonico FL, et al. Experience with pyeloure-
the patient—the site of the anastomosis. Although theoreti- terostomy in renal transplantation. Ann Surg 1982;196(5):588–93.
cally at risk of rejection, this does not, in reality, appear to 25. McDonald JC, Landreneau MD, Hargroder DE, et al. External uretero-
cause a problem. neocystostomy and ureteroureterostomy in renal transplantation.
Ann Surg 1987;205(4):428–31.
If the wound is grossly contaminated or infected, it 26. Lich Jr R. Obstructive diseases of the urinary tract in children. J Ark
should be left open with packing, with secondary closure in Med Soc 1961;58:127–30.
mind.83,84 Closed suction drainage may be used, according 27. Politano VA, Leadbetter WF. An operative technique for the correc-
to the surgeon’s preference. tion of vesicoureteral reflux. J Urol 1958;79(6):932–41.
28. Stevens A, Marshall V. Reimplantation of the ureter into the bladder.
Surg Gynecol Obstet 1943;77:585–94.
References 29. Kootstra G. Kidney transplantation. In: Kremer B, Broelsch C, Henne-
1. Abramowicz D, Hazzan M, Maggiore U, et al. Does pre-emptive trans- Bruns D, editors. Atlas of liver, pancreas, and kidney transplantation.
plantation versus post start of dialysis transplantation with a kidney Stuttgart: Georg Thieme Verlag; 1994. p. 128.
from a living donor improve outcomes after transplantation? A system- 30. Salehipour M, Salahi H, Fathikalajahi A, et al. Is perioperative
atic literature review and position statement by the Descartes Working intravesically applied antibiotic solution effective in the prophy-
Group and ERBP. Nephrol Dial Transplant 2016;31(5):691–7. laxis of urinary tract infections after renal transplantation? Urol Int
2. Li B, Cairns JA, Robb ML, et al. Predicting patient survival after 2010;85(1):66–9.
deceased donor kidney transplantation using flexible parametric mod- 31. Salmela K, Eklund B, Kyllonen L, et al. The effect of intravesically
elling. BMC Nephrol 2016;17(1):51. applied antibiotic solution in the prophylaxis of infectious complica-
3. Wu DA, Robb ML, Watson CJE, et al. Barriers to living donor kidney tions of renal transplantation. Transpl Int 1990;3(1):12–4.
transplantation in the United Kingdom: a national observational 32. Barry JM, Lemmers MJ, Meyer MM, et al. Cadaver kidney transplanta-
study. Nephrol Dial Transplant 2017;32(5):890–900. tion in patients more than 65 years old. World J Urol 1996;14(4):243–8.
172 Kidney Transplantation: Principles and Practice

33. Starzl T. Experience in renal transplantation. Philadelphia: WB 60. Dunfield VM. Boari operation in case of solitary kidney; treatment
­Saunders; 1964. of obstruction of solitary ureter following hysterectomy. AMA Arch
34. Slagt IK, Dor FJ, Tran TC, et al. A randomized controlled trial compar- Surg 1955;70(3):328–32.
ing intravesical to extravesical ureteroneocystostomy in living donor 61. Hatch DA, Belitsky P, Barry JM, et al. Fate of renal allografts trans-
kidney transplantation recipients. Kidney Int 2014;85(2):471–7. planted in patients with urinary diversion. Transplantation
35. Alberts VP, Idu MM, Legemate DA, et al. Ureterovesical anastomotic 1993;56(4):838–42.
techniques for kidney transplantation: a systematic review and meta- 62. Kelly WD, Merkel FK, Markland C. Ileal urinary diversion in conjunc-
analysis. Transpl Int 2014;27(6):593–605. tion with renal homotransplantation. Lancet 1966;1(7431):222–6.
36. Woodruff MF, Nolan B, Robson JS, et al. Renal transplantation in 63. Whitehead ED, Narins DJ, Morales PA. The use of methylene blue
man. Experience in 35 cases. Lancet 1969;1(7584):6–12. in the identification of the ileal conduit during re-operation. J Urol
37. Konnak JW, Herwig KR, Turcotte JG. External ureteroneocystostomy 1972;107(6):960–2.
in renal transplantation. J Urol 1972;108(3):380–1. 64. Patel P, Rebollo-Mesa I, Ryan E, et al. Prophylactic ureteric stents in
38. Bradic I, Pasini M, Vlatkovic G. Antireflux ureterocystostomy at the renal transplant recipients: a multicenter randomized controlled trial
vertex of the bladder. Br J Urol 1975;47(5):525–30. of early versus late removal. Am J Transplant 2017;17(8):2129–38.
39. Campos Freire J, de Goes GM, de Campos Freire JG. Extravesical ureteral 65. Liu S, Luo G, Sun B, et al. Early removal of double-J stents decreases
implantation in kidney transplantation. Urology 1974;3(3):304–8. urinary tract infections in living donor renal transplantation: a prospec-
40. Benoit G, Blanchet P, Eschwege P, et al. Insertion of a double pig- tive, randomized clinical trial. Transplant Proc 2017;49(2):297–302.
tail ureteral stent for the prevention of urological complications 66. Hume D. Kidney transplantation. In: Rapaport F, Dausset J, editors.
in renal transplantation: a prospective randomized study. J Urol Human transplantation. London: Grune & Stratton; 1968. p. 110.
1996;156(3):881–4. 67. Shackman R, Dempster WJ, Wrong OM. Kidney homotransplantation
41. Pleass HC, Clark KR, Rigg KM, et al. Urologic complications after renal in the human. Br J Urol 1963;35:222–55.
transplantation: a prospective randomized trial comparing different 68. Belzer FO, Schweitzer RT, Holliday M, et al. Renal homotransplanta-
techniques of ureteric anastomosis and the use of prophylactic ure- tion in children. Am J Surg 1972;124(2):270–8.
teric stents. Transplant Proc 1995;27(1):1091–2. 69. Najarian JS, Simmons RL, Tallent MB, et al. Renal transplantation in
42. Wilson CH, Rix DA, Manas DM. Routine intraoperative ureteric stent- infants and children. Ann Surg 1971;174(4):583–601.
ing for kidney transplant recipients. Cochrane Database Syst Rev 70. Broyer M, Gagnadoux MF, Beurton D, et al. Transplantation in chil-
2013;(6):CD004925. dren: technical aspects, drug therapy and problems related to primary
43. Barry JM. Unstented extravesical ureteroneocystostomy in kidney renal disease. Proc Eur Dial Transplant Assoc 1981;18:313–21.
transplantation. J Urol 1983;129(5):918–9. 71. Nahas WC, Mazzucchi E, Scafuri AG, et al. Extraperitoneal access
44. Knechtle S. Ureteroneocystostomy for renal transplantation. J Am for kidney transplantation in children weighing 20 kg. or less. J Urol
Coll Surg 1999;188(6):707–9. 2000;164(2):475–8.
45. Conlin MJ, Lemmers MJ, Barry JM. Extravesical ureteroneocystostomy 72. Amante AJ, Kahan BD. En bloc transplantation of kidneys from pedi-
for duplicated allograft ureters. J Urol 1994;152(4):1201–2. atric donors. J Urol 1996;155(3):852–6; discussion: 856-7.
46. Prout Jr GR, Hume DM, Williams GM, et al. Some urological aspects 73. Dreikorn K, Rohl L, Horsch R. The use of double renal transplants
of 93 consecutive renal homotransplants in modified recipients. J Urol from paediatric cadaver donors. Br J Urol 1977;49(5):361–4.
1967;97(3):409–25. 74. Kinne DW, Spanos PK, DeShazo MM, et al. Double renal transplants from
47. Barry JM, Pearse HD, Lawson RK, et al. Ureteroneocystostomy in kidney pediatric donors to adult recipients. Am J Surg 1974;127(3):292–5.
transplant with ureteral duplication. Arch Surg 1973;106(3):345–6. 75. Lindstrom BL, Ahonen J. The use of both kidneys obtained from pedi-
48. Szmidt J, Karolak M, Sablinski T, et al. Transplantation of kidneys atric donors as en bloc transplants into adult recipients. Scand J Urol
with nonvascular anatomical abnormalities. Transplant Proc Nephrol 1975;(29 Suppl):71–2.
1988;20(5):767. 76. Schneider JR, Sutherland DE, Simmons RL, et al. Long-term success
49. Masson D, Hefty T. A technique for the transplantation of 2 adult with double pediatric cadaver donor renal transplants. Ann Surg
cadaver kidney grafts into 1 recipient. J Urol 1998;160(5):1779–80. 1983;197(4):439–42.
50. Fjeldborg O, Kim CH. Double ureters in renal transplantation. J Urol 77. Maluf DG, Carrico RJ, Rosendale JD, et al. Optimizing recovery, utili-
1972;108(3):377–9. zation and transplantation outcomes for kidneys from small, ≤20 kg,
51. Gil-Vernet JM, Gil-Vernet A, Caralps A, et al. Orthotopic renal trans- pediatric donors. Am J Transplant 2013;13(10):2703–12.
plant and results in 139 consecutive cases. J Urol 1989;142(2 Pt 78. Alfrey EJ, Lee CM, Scandling JD, et al. When should expanded crite-
1):248–52. ria donor kidneys be used for single versus dual kidney transplants?
52. Hamburger J, Crosnier J, Dormont J. Experience with 45 renal homo- Transplantation 1997;64(8):1142–6.
transplantations in man. Lancet 1965;1(7393):985–92. 79. Remuzzi G, Grinyo J, Ruggenenti P, et al. Early experience with
53. Lawler RH, West JW, McNulty NP, et al. Homotransplantation of the dual kidney transplantation in adults using expanded donor cri-
kidney in the human. J Am Med Assoc 1950;144(10):844–5. teria: Double Kidney Transplant Group (DKG). J Am Soc Nephrol
54. Leadbetter Jr GW, Monaco AP, Russell PS. A technique for reconstruc- 1999;10(12):2591–8.
tion of the urinary tract in renal transplantation. Surg Gynecol Obstet 80. Stratta RJ, Bennett L. Preliminary experience with double kidney
1966;123(4):839–41. transplants from adult cadaveric donors: analysis of United Network
55. Lord RH, Pepera T, Williams G. Ureteroureterostomy and pyeloure- for Organ Sharing data. Transplant Proc 1997;29(8):3375–6.
terostomy without native nephrectomy in renal transplantation. Br J 81. Remuzzi G, Cravedi P, Perna A, et al. Long-term outcome of renal trans-
Urol 1991;67(4):349–51. plantation from older donors. N Engl J Med 2006;354(4):343–52.
56. Schiff Jr M, Lytton B. Secondary ureteropyelostomy in renal trans- 82. Ekser B, Baldan N, Margani G, et al. Monolateral placement of both
plant recipients. J Urol 1981;126(6):723–5. kidneys in dual kidney transplantation: low surgical complication
57. Bennett AH. Pyelocystostomy in a renal allograft. Am J Surg rate and short operating time. Transpl Int 2006;19(6):485–91.
1973;125(5):633–5. 83. Kohlberg WI, Tellis VA, Bhat DJ, et al. Wound infections after trans-
58. Firlit CF, Merkel FK. The application of ileal conduits in pediatric renal plant nephrectomy. Arch Surg 1980;115(5):645–6.
transplantation. J Urol 1977;118(4):647–50. 84. Schweizer RT, Kountz SL, Belzer FO. Wound complications in recipi-
59. Herwig KR, Konnak JW. Vesicopyelostomy: a method for urinary ents of renal transplants. Ann Surg 1973;177(1):58–62.
drainage of the transplanted kidney. J Urol 1973;109(6):955–7.
12 Transplantation and the
Abnormal Bladder
ARMAN A. KAHOKEHR and ANDREW C. PETERSON

CHAPTER OUTLINE Assessment of Lower Urinary Tract Urinary Tract Infections


Function and Pretransplant Workup Methods to Achieve Continence
Physical Examination and Noninvasive Tests Results of Renal Transplantation into
Indications for Invasive Investigations Reconstructed and Abnormal Bladders
Pretransplant Imaging Special Considerations for the Transplant
Normal Cystometric Values Surgeon
Special Pretransplant Issues Posterior Urethral Valves
Methods to Enhance Bladder Emptying Prune-Belly Syndrome
Vesicoureteral Reflux and Abnormal Bladder Male Voiding Dysfunction, BPH/LUTS
Compliance Female Voiding Dysfunction
Timing of Bladder Reconstruction and Conclusion
“Dry-Diversion”
Posttransplant Issues
Maintenance of Lower Urinary Function

The ability of the urinary bladder to store urine at low the abnormal bladder that contributed to the damage
pressure and to empty completely at intervals with simul- of the native kidneys might adversely influence the out-
taneous relaxation of the sphincter complex is essential to come of the transplant. Many reports have shown that
preserve the integrity of the kidneys and to achieve conti- bladder dysfunction can negatively affect graft function
nence. Although an abnormal lower urinary tract is not a if left untreated. Reinberg and colleagues6 first pointed
contraindication to renal transplantation, lower urinary this out in 1988 for children with PUV, and since then
tract dysfunction (LUTD) needs to be addressed before and other authors have verified these observations.7,8 Correc-
after transplantation. In this chapter we shall discuss the tion of structural anomalies and optimization of storage
causes, evaluation, and methods of treatment of bladder and emptying functions of the bladder are often recom-
dysfunction. mended before transplantation.9 In fact, improvement of
Abnormal bladder function can be present in both bladder function can slow down the progression of renal
adults and children with end-stage renal disease (ESRD) insufficiency and allow transplantation to be postponed.10
who are transplant candidates, but the problem is more Although in general all anticipated reconstructive proce-
prevalent in the pediatric age group. About 20% to 30% of dures on the lower urinary tract are best performed before
children who develop renal failure have potential bladder transplantation, this is not always possible for several rea-
dysfunction as the etiology of the renal failure or a con- sons. For example, in children before toilet training it may
tributor to ESRD.1,2 be difficult to predict future continence. When polyuria is
Patients at risk for bladder dysfunction include those present, it may be difficult to predict how the bladder will
with a history of posterior urethral valve (PUV), prune-belly behave once normal diuresis is restored. Likewise it is dif-
syndrome (PBS), neurogenic bladder (NGB), bladder exstro- ficult to predict future bladder function after a prolonged
phy, Ochoa and Hinman syndromes, patients with anorec- period of oligo- or anuria.
tal malformations, and persistence of the cloaca. In some It should also be kept in mind that bladders that seem to
of these conditions, the development of ESRD is frequently have normal function initially may become abnormal over
a consequence of the congenital anomaly associated with time, such as is seen in cases of valve bladders or in children
renal dysplasia (PUV, PBS, persistence of the cloaca);3–5 with tethered spinal cord, and sometimes in adults with
however, in others, such as NGB, whether congenital or bladder outlet obstruction from the prostate. The opposite
acquired, renal damage results from bladder dysfunction is also true. For example, a bladder that has inadequate
and is preventable with good management. capacity or compliance in the face of polyuria may function
When renal failure results from underlying urologic adequately after transplantation when normal urine out-
anomalies (e.g., PUV, PBS, NGB), it can be assumed that put is restored.11

173
174 Kidney Transplantation: Principles and Practice

Assessment of Lower Urinary Routine office investigations include a 72-hour void-


ing diary (Fig. 12.1). This records the date, time, and vol-
Tract Function and Pretransplant ume of each void. In addition, each incontinent episode
Workup is recorded by the amount and the precipitating cause.
This is very important to gauge the magnitude and fre-
The general principles of making the bladder useful to quency of incontinence, the overall voiding pattern, and
receive a renal graft include: (1) providing a method for the daily urine output, and estimates a functional blad-
emptying when spontaneous voiding is not possible, (2) der capacity. If incontinence is present, we insist on
restoring a safe bladder capacity and compliance to protect pad weight measurement; this is the most accurate and
the graft, and (3) achieving continence. Although urinary objective measure of incontinence before any invasive
incontinence may not affect the functional result of trans- treatment. In adult male patients, an American Urologi-
plantation, restoring continence greatly enhances quality cal Association (AUA) symptom score (Fig. 12.2) is also a
of life.12 useful screening and baseline tool for LUTS/benign pros-
ESRD often causes reduced urine production and loss of tatic hyperplasia (BPH).
normal physiologic distention of the bladder, which may Noninvasive urodynamics (UDs) such as uroflow (Fig.
lead to a temporary reduction in bladder capacity and loss 12.3), and postvoid residual (PVR) are used routinely in
of compliance. In neurologically and structurally normal our clinic. The uroflow by itself, although it cannot actually
patients, this reduction in capacity has been shown to be determine the etiology of abnormal voiding (some classic
directly related to the duration of time on dialysis with patterns are seen however; Figs. 12.4 and 12.5), serves as
capacity of less than 150 mL seen after an average of 76 an excellent screening tool to determine whether further,
months on dialysis.13 However, it has been shown that more invasive testing is needed.19 In most patients with-
bladder capacity improved once cycling of the bladder has out symptoms, a normal uroflow study and the absence
been established after transplantation even in those with of residual urine on ultrasonography are sufficient to rule
bladder capacity of less than 100 mL at the time of trans- out significant bladder dysfunction. Abnormal uroflow pat-
plantation.14 A functional bladder may need to be reevalu- terns or incomplete bladder emptying may be situational
ated over time if the waiting time for renal transplantation and should be repeated.20,21
is prolonged or if new lower urinary tract symptoms occur. Routine laboratory investigations (in addition to trans-
It also is known that LUTD in children and adolescents plant workup) include urine analysis (UA) and culture.
occurs after transplantation, even when the bladder was If hematuria or sterile pyuria is detected, this will lead to
normal before renal transplantation, warranting careful more invasive investigations with cystoscopy (discussed
posttransplant attention.15,16 next).
The pretransplant urologic evaluation aims to diag-
nose, treat, and optimize any preexisting LUTD.2,17,18 All INDICATIONS FOR INVASIVE INVESTIGATIONS
patients in ESRD with known or suspected lower genito-
urinary abnormalities require evaluation of bladder func- Invasive UDs with fluoroscopy (videourodynamics, VUDs)
tion, and adults with lower urinary tract symptoms (LUTS) are a useful test for evaluating patients with LUTD. Some
should be evaluated by a urologist before transplant. Addi- uncomplicated patients may not need these after the non-
tionally, we feel that all pediatric patients require complete invasive investigations have been completed. The AUA has
evaluation of the lower urinary tract before renal trans- published guidelines on UDs and states that VUDs should
plantation and recommend referral to a pediatric urolo- be strongly considered in all NGB patients in the face of
gist with specialization in LUTD. We perform preoperative pending invasive surgical treatment.22 In nonneurogenic
evaluation with a detailed history and physical examina- patients with suspected detrusor overactivity (DO), abnor-
tion that includes a bladder diary kept over several days malities of filling and storage, and bladder outlet obstruc-
to record voided volumes and frequency, incontinent epi- tion, UDs may be performed. This can be helpful when there
sodes, and presence of nocturia or nocturnal enuresis. In is a diagnostic dilemma, symptoms do not correlate with
anuric patients, the history before the onset of anuria is objective findings, and when considering irreversible inva-
very valuable. sive therapy.
Our VUDs protocol includes cystometry and electro-
PHYSICAL EXAMINATION AND NONINVASIVE myography of the pelvic floor when the bladder capacity
and compliance are questionable, the uroflow pattern is
TESTS abnormal, or there is a history of bladder neck or out-
On examination we look for scars, stomas, and skeletal let dysfunction. The use of fluoroscopy can also be valu-
deformities, and perform a pertinent neurologic exami- able in helping determine the etiology of the voiding
nation. This includes mental status, sensory and motor dysfunction.
function, and reflex integrity. Evaluation of the S2-4 Cystoscopy is indicated in patients who demonstrate
reflexes using bulbocavernosus reflex and anal wink hematuria, sterile pyuria, and difficulty catheterizing
can help determine whether this pathway is intact, the or other indications of anatomic urethral or bladder
absence may indicate sacral nerve disease. In females, abnormality. Patients who have had a prior augmen-
determination of the pelvic floor strength and biman- tation cystoplasty or any other questionable surgeries
ual examination is performed to assess the neurologic should also undergo pretransplant cystoscopy to rule
system. out stones, foreign bodies, lesions, and possible tumors
12 • Transplantation and the Abnormal Bladder 175

Time Amount Leak Volume Activity During Urge? Fluid Intake


Voided (in cc’s 1 = Drop/Damp Leak (Yes/No) (Amount in
or Ounces) 2 = Wet Ounces/Type)
3 = Bladder
Emptied

Fig. 12.1 An example of a 24-hour voiding diary; patients are asked to complete this over 3 days. (Reproduced with permission from Springer, Practical
Urodynamics for the Clinician, page 24.)

that may arise in these situations. It is the gold standard After the evaluation is completed, one can make a judg-
test to rule out pathology of the bladder and urethra. ment as to the adequacy of the lower urinary tract. Criteria
It is also of some benefit in those with bladder outlet for a usable bladder relate to bladder capacity, bladder com-
obstruction and also provides information on the ure- pliance, the bladder’s ability to empty completely, and uri-
thral sphincter. nary continence. The presence of vesicoureteral reflux also
should be taken into consideration.
Errando et al. found it necessary to perform UDs in 6.9%
PRETRANSPLANT IMAGING
of 475 transplant recipients based on the following indica-
In addition to a screening ultrasound of the native kid- tions: (1) the presence of lower urinary tract symptoms as
neys performed in all transplant candidates, further outlined by history and patient reported outcome surveys,
imaging may be needed. In the case of hematuria and (2) the presence of a defunctionalized bladder in the anuric
sterile pyuria, routine imaging of the upper urinary tract patient, and (3) the presence of a complex urologic history
is indicated with contrast computed tomography (CT) where the diagnosis cannot be made on history alone. These
with a delayed protocol to opacify the native collecting investigators found that 45% of the evaluated patients had
system. In cases where there is not enough renal func- abnormal urodynamic studies.23 However, defunctional-
tion to opacify the collecting system, patients may need ized bladders that were normal before the onset of anuria,
to undergo cystoscopy with evaluation of the collecting as a rule, recover function after diuresis is restored without
system by the use of retrograde pyelograms. If a previ- specific management.20
ously unknown neurologic condition is suspected, inves- In some cases of LUTD, a voiding cystourethrogram
tigation with magnetic resonance imaging (MRI) of the (VCUG) is useful to estimate bladder capacity and to outline
brain and spinal cord may be necessary to evaluate the its contour, and it may be performed at the same time as
central nervous system for possible etiology. UDs when fluoroscopy is used. It is also essential to evaluate
176 Kidney Transplantation: Principles and Practice

AUA SYMPTOM SCORE (AUASS)

PATIENT NAME: ___________________________ TODAY’S DATE: _____________

(Circle One Number on Not at Less Less Than About More Almost
Each Line) All Than 1 Half the Half the Than Half Always
Time in 5 Time Time the Time

Over the past month or so,


how often have you had a 0 1 2 3 4 5
sensation of not emptying
your bladder completely after
you finished urinating?
During the past month or so,
how often have you had to
urinate again less than two 0 1 2 3 4 5
hours after you finished
urinating?
During the past month or so,
how often have you found
you stopped and started 0 1 2 3 4 5
again several times when
you urinated?
During the past month or so,
how often have you found it 0 1 2 3 4 5
difficult to postpone urination?
During the past month or so,
how often have you had a 0 1 2 3 4 5
weak urinary stream?
During the past month or
so, how often have you 0 1 2 3 4 5
had to push or strain to
begin urination?
None 1 Time 2 Times 3 Times 4 Times 5 or
More
Times
Over the past month, how
many times per night did
you most typically get up
to urinate from the time 0 1 2 3 4 5
you went to bed at night
until the time you got up in
the morning?

Add the score for each number above and TOTAL: _____________
write the total in the space to the right.
SYMPTOM SCORE: 1–7 (Mild) 8–19 (Moderate) 20–35 (Severe)

QUALITY OF LIFE (QOL)

Delighted Pleased Mostly Mixed Mostly Unhappy Terrible


Satisfied Dissatisfied

How would you


feel if you had to
live with your
urinary condition 0 1 2 3 4 5 6
the way it is
now, no better,
no worse, for the
rest of your life?

Fig. 12.2 The American Urology Association symptom score, also known as the International Prostate Symptom Score (IPSS).
12 • Transplantation and the Abnormal Bladder 177

urethral anatomy and to determine the presence of vesico- satisfactory for a given patient. With the capacity of the
ureteral reflux when that information is necessary. Ramirez newborn bladder at about 30 mL, and bladder capac-
et al. retrospectively looked at 271 pediatric renal trans- ity increasing by about 30 mL each year almost until
plantations and found VCUG useful in children less than 8 puberty,21 the formula (age in years + 1) × 30 = bladder
years of age and those with a history of urologic disease,18 capacity in mL is useful. For infants we prefer the estima-
whereas in adults without a history of urologic disease or tion of 7 mL/kg.25 Although most calculations use the
symptoms VCUG is not necessary.24 patient’s age, assuming that the body habitus is within
normal limits, this is often not the case in patients with
spina bifida and ESRD. In this population we prefer the for-
NORMAL CYSTOMETRIC VALUES
mula mentioned earlier, which assumes 7 mL of capacity
Bladder capacity varies with age. Known formulae exist for every kg of body weight. The presence of a lower than
to determine whether the bladder capacity for age is expected bladder capacity on a preoperative evaluation
should serve as an indication for more involved testing
Normal Curve
and referral to a urologist.
Bladder compliance is defined as the change in blad-
der pressure for a given change in volume. It is calcu-
Maximum
flow rate lated by dividing the volume change (ΔV) by the change
in detrusor pressure (ΔPdet)—compliance ΔV/ΔP detru-
Volume (mL)

sor—and is expressed in mL/cmH2O. Decreased bladder


Average compliance implies a poorly distensible bladder in which
flow rate the pressure/volume curve is steep, and the pressure rise
Voided
volume is rapid for low-volume increases. Although numerical
values are often used to express compliance, interpreta-
Time (s)
tion of these numbers must take into consideration age
Time to maximum flow and expected bladder capacity. The lowest full resting
Flow time pressure is preferable regardless of the maximal bladder
capacity. Normal compliance is thought by most experts
Fig. 12.3 Illustration of a normal uro-flow curve, continuous and bell
shaped with a tail; Qmax is reached in the first third and final phase and to be greater than 12 mL/cmH2O. Transplantation into
shows a rapid decrease and a sharp cutoff at the end. (Reproduced with a small, low compliant bladder is possible, as demon-
permission from Springer, Practical Urodynamics for the Clinician, page 36.) strated by a report on 13 patients transplanted into small

Continuous Flow Curves

A B

C D
Fig. 12.4 Abnormal continuous flow curves; (A) compressive (benign prostatic obstruction); (B) constrictive flow curve (urethral stricture); (C) idiopathic
detrusor overactivity; (D) detrusor underactivity. (Reproduced with permission from Springer, Practical Urodynamics for the Clinician, page 36.)

Intermittent Flow Curves

A B
Fig. 12.5 Abnormal intermittent flow curves; (A) detrusor sphincter dyssynergia; (B) Valsalva voiding curve: patients with areflexic or hypocontractile
bladder void with valsalva maneuver that is represented as an intermittent and irregular curve. (Reproduced with permission from Springer, Practical
Urodynamics for the Clinician, page 37.)
178 Kidney Transplantation: Principles and Practice

bladders that had been defunctionalized for 3 to 20 years compliance after refunctionalization by undiversion or
but not augmented (three PUVs). A graft survival of 62% transplantation. Cycling such bladders before transplan-
at 4 years was seen.26 tation is uncomfortable for the patient and unnecessary,
and we no longer recommend this practice.38 A bladder
may be anatomically small, as is often the case in patients
with a history of bladder exstrophy or epispadias. A small
Special Pretransplant Issues bladder per se is not a threat to the upper tracts as long as
the patient is incontinent and compliance is safe. However,
METHODS TO ENHANCE BLADDER EMPTYING when bladder outlet resistance has previously been surgi-
Periodic, complete emptying of the bladder is important to pre- cally increased in an attempt to achieve continence, high
vent infections, keep the intravesical pressure low, and allow intravesical pressure and hydronephrosis may ensue. Such
for urinary continence. Chronic urinary retention may lead bladders may require reconstruction with bladder augmen-
to overflow incontinence and urinary tract infection (UTI). tation or other aggressive therapies to improve their storage
When intravesical pressures are high, hydronephrosis with or ability to safely accept a transplant kidney.
without vesicoureteral reflux and renal damage may develop. Patients with neurogenic LUTD may have anatomically
McGuire and associates27 stated that sustained detrusor pres- normal but functionally small bladder capacity at safe pres-
sures greater than 40 cmH2O can cause upper tract damage. sures. Some cases of low-volume and poor compliance may
Medications that relax the bladder neck musculature, such as respond well to oral antimuscarinic medications or botuli-
alpha blockers, may be used but are of unproven effectiveness num A toxin injected in the detrusor. Antimuscarinic drugs,
in patients with neuropathic voiding dysfunction (NVD) or although not without potential side effects, can be effective
PUV.28,29 Symptomatic improvement of lower urinary tract and well tolerated for long periods of time and thus should
symptoms in adults has been documented.30,31 Botulinum A be tried before surgical augmentation. Botulinum toxin
toxin injection to the sphincter mechanisms may be effective injections, on the other hand, have a limited and temporary
to improve bladder emptying in high spinal cord lesions but effect and are impractical for long-term management.39
the effect is short-lived.32 Therefore in most cases of incomplete Both of these methods can only be effective when the cause
bladder emptying, clean intermittent catheterization may be of the decreased compliance lies in the detrusor.
necessary and provides a safe way to manage bladders that Surgical augmentation of bladder capacity with its con-
otherwise cannot function on their own.33 This is also true for sequent improvement in compliance is normally accom-
renal transplant patients.34 plished by adding a reconfigured intestinal segment to
the existing bladder. The majority of patients, particularly
VESICOURETERAL REFLUX AND ABNORMAL those with NGB, will require intermittent catheterization
to empty the reconstructed bladder. For patients without a
BLADDER COMPLIANCE bladder, an incontinent diversion such as an ileal or colonic
In nonneurogenic patients, the presence of vesicoureteral conduit can be constructed either before the transplant or
reflux (VUR) without LUTD may not necessarily put the at the time of transplant. With current improved immuno-
graft at risk. However, when voiding dysfunction exists, suppressive drugs, it is feasible to perform an ileal conduit
VUR in an immunocompromised patient may lead to renal in patients who are already established on immunosuppres-
infections and other complications. Hence when preop- sion therapy or at the same time as graft implantation.40–42
erative VUR exists, a complete evaluation is warranted. Success of transplantation into such conduits is good and
This includes VUDs to rule out bladder outlet issues and well documented.43,44 A continent urinary reservoir can
loss of compliance. If severe reflux is identified in a neuro- also be used to avoid an incontinent stoma.45
pathic setting and is thought to be from poor compliance
or high-pressure voiding, then augmentation may need to TIMING OF BLADDER RECONSTRUCTION AND
be performed preoperatively to produce a low capacity safe “DRY-DIVERSION”
bladder reservoir. High-grade vesicoureteral reflux that is
left untreated after transplantation is often accompanied The timing and type of bladder augmentation relative to
by a higher risk of UTIs, even if it was not a problem before the transplantation warrant comment. This can be a topic
transplantation.35 of contention; one argument is that healing of bowel anas-
Surgical options for treatment of VUR—with ureteral tomoses and incisions may be adversely affected by the
reimplantation or nephrectomy—have been associated immunosuppressive status. Historically most authors have
with a reduced risk of infection after transplantation.36 performed the augmentation or urinary diversion with
A typical case is that of a boy with PUV with renal insuf- ileal or ileocecal segment before transplantation.46–48 This
ficiency and bilateral high-grade reflux. If there are no provides an optimal environment for transplantation and
febrile UTIs, the best course of action is to wait until the familiarity with the diversion. On the other hand, some have
time for transplantation is near. Nephrectomy either before argued for a delay in reconstruction where it is possible for
transplant or at the time of transplant may be necessary to stabilization of renal function after successful transplanta-
prevent recurrent infections and is at the discretion of the tion.49,50 There is no randomized evidence, and reconstruc-
transplant surgeon.37 tion before transplantation seems to be a safe approach,
There are many possible causes that decrease compli- but it presents a management problem when the patient is
ance of the bladder. A previously normal defunctional- anuric. In those expecting a cadaver donor, the bladder or
ized bladder because of anuria or supravesical diversion neobladder must be kept sterile, so as not to miss possible
may be small but usually returns to normal capacity and opportunities to use a well-matched organ. We usually
12 • Transplantation and the Abnormal Bladder 179

recommend daily bladder irrigations and instillation of an commonly seen in transplantation into abnormal blad-
antibiotic solution. Instillation of aminoglycosides, which ders.54 The use of prophylactic single-dose antibiotic in
is usually safe in patients with normal renal function, may the first 6 months after transplantation does reduce the
lead to complications in patients with ESRD.51 The small incidence of UTI by half.55 In those with recurrent UTIs,
number of cases in which the bladder was augmented after imaging to evaluate for stones in the entire urinary tract,
transplantation attests to the feasibility of such an approach including native kidneys, in addition to cystoscopy of the
when needed.52 bladder or reservoir is indicated. Mucus production from
Recent case reports show that it is also feasible to per- ileum or colon bowel mucosa occurs equally and is normal.
form ileal conduit diversion simultaneously at transplanta- Mucus can be dealt with by daily irrigation with saline and
tion40,53 avoiding the dry-diversion scenario, reoperation, the use of alkalizers if needed.
and the complications associated with the staged approach.
Nevertheless, until there have been further studies on this Methods to Achieve Continence
issue, it is generally recommended that if a conduit or a Some patients with bladder abnormalities such as NGB and
bladder augmentation is needed, it should be done several PUV when associated with ESRD may also suffer from uri-
weeks before transplantation. nary incontinence. In general urinary incontinence can
be caused by: (1) failure of the bladder to empty (overflow
incontinence); (2) failure to store urine at low pressure; (3)
incompetence of the sphincter mechanisms (stress urinary
Posttransplant Issues incontinence); (4) urgency incontinence; and (5) rarely,
bypass of the sphincter complex (such as ectopic ureters,
MAINTENANCE OF LOWER URINARY FUNCTION fistulae). Frequently more than one factor is present. The
In all pediatric patients or adults who had prior LUTD, reg- history, imaging studies, and urodynamic evaluation are
ular evaluation by a urologist is recommended to ensure essential to establish the pathophysiology of incontinence
ongoing safe lower tract function and prolonged graft in a given patient. Overflow incontinence is managed by
survival. In addition, any new issues such as UTI in the removal of the obstruction or, when the cause is failure of
immunocompromised setting can be addressed. Evalua- detrusor contractility, intermittent catheterization. The
tion includes an interval history, patient reported outcome management of poor capacity and compliance was dis-
measures such as the AUA symptom score, bladder diary, cussed in the previous section.
examination and uroflowmetry (see Figs. 12.3–12.5), and The management of sphincter failure requires simulta-
residual measures when indicated. Care is usually coordi- neous attention to the storage capacity of the bladder. In
nated with the nephrologist so that any new issues can be the authors’ experience, injection of bulking agents in the
addressed in a multidisciplinary setting. bladder neck or proximal urethra has only a secondary role
in enhancing continence and seldom achieves satisfactory
Urinary Tract Infections results in cases of neuropathic sphincter failure.
In patients with bowel interposition into the urinary tract, Numerous procedures have been described in an attempt
the urinary tract will be colonized by bacteria. In the immu- to “reconstruct” the bladder neck. With the possible excep-
nocompromised setting, UTIs are one of the most common tion of occasional success in cases of bladder exstrophy,
and bothersome long-term issues. Asymptomatic bacteri- we have been disappointed with the results of such proce-
uria does not need treatment, although formation of stones dures for neuropathic incontinence in the adult and have
needs to be carefully monitored because urease splitting abandoned their use. Implantation of an artificial urinary
organisms may reside. We recommend annual imaging of sphincter (AUS) is effective as an initial option to manage
the urinary tract to look for bladder or upper-tract stones sphincter incompetence (Fig. 12.6) and can be implanted at
if there has been a history of stones. Symptomatic UTIs are the bladder neck or the bulbar urethra. The AUS has become

A B
Fig. 12.6 (A) The AMS-800 artificial urinary sphincter: It consists of a cuff implanted around the bulbar urethra, a pressure regulating balloon, and a
pump that is positioned in the scrotum or labium major. (B) The plain radiograph shows components of the AMS-800—that contains contrast media
within the system allowing troubleshooting of the device.
180 Kidney Transplantation: Principles and Practice

the gold standard for treatment of incontinence associated augmentation with patients with diversions; this is prob-
with sphincter function loss.56 The current model (AMS lematic because it is well recognized that nonrefluxing
800, Boston Scientific, Boston, MA) was introduced in ureteroenterostomies, in contrast to ureteroneocystosto-
1983 and has by and large remained unchanged to this mies, carry a risk of stenosis of greater than 10%.60 Nev-
day. Experience associated with the AMS-800 is therefore ertheless, one retrospective controlled study that included
very high and 80% continence rates have been reported mostly adult patients with urinary diversion failed to show
from numerous centers.57 In addition, the compatibility any differences with control patients with normal blad-
of the AUS with renal transplantation is well established. ders.61 Most authors agree that, although more compli-
Revisions of the device may be needed for device malfunc- cated, it is feasible to proceed with renal transplantation
tion, waning incontinence, infection, and urethral cuff ero- in patients who are known to have an abnormal bladder
sion.56 These secondary procedures and salvage operations with good results in both adult62,63 and pediatric and tran-
are associated with comparable outcomes compared with sition patients.33,44,47,64–69
primary surgery.58 In carefully selected patients, the use
of the male transobturator sling has comparable outcomes
compared with the AUS.59 Special Considerations for
In females, aponeurotic slings created with the patient’s
rectus fascia or other off-the-shelf biological materials
the Transplant Surgeon
(cadaveric fascia lata, porcine intestinal submucosa) are
POSTERIOR URETHRAL VALVES
effective for those dependent on intermittent catheteriza-
tion who have excellent bladder capacity and compliance Renal transplantation in patients with a history of PUV
(Fig. 12.7). presents unique challenges. Some of these patients have
bladder dysfunction (see Fig. 12.5) with poor compli-
ance,70,71 and the proportion may be higher in those who
Results of Renal Transplantation have renal failure. Although many uncontrolled studies
suggest that renal transplantation into the valve blad-
into Reconstructed and Abnormal der is associated with good results,72,73 close examina-
Bladders tion of every controlled study reported to date indicates
that patients with renal transplantation into nonrecon-
Transplantation can be performed safely in patients structed valve bladders exhibit higher creatinine lev-
with reconstructed bladders and urinary diversions with els at the end of 5 years compared with controls. This
acceptable graft survival and function. Whereas some higher creatinine level has been observed in virtually
authors report an increased incidence of urologic com- all studies reported and has been attributed to bladder
plications, such as urinary leak, ureteral stenosis, symp- dysfunction.6,7,74,75 Salomon and colleagues76 reported
tomatic UTIs, metabolic acidosis, and calculi, there are worse results of transplantation in children with PUVs
few controlled studies that permit meaningful compari- and symptomatic bladder dysfunction73 than in children
sons between results of transplantation in native versus without such symptoms. The graft survival may be nor-
reconstructed bladders. Comparison among reported mal or marginally decreased in these cases.77 Therefore
series is difficult because some fail to define the source of it has been tempting to pursue an aggressive approach
the graft, which is one of the best-known determinants of to the valve bladder in hopes of improving the lifespan of
graft survival. Some series combine patients with bladder the native kidneys and the results of renal transplanta-
tion. However, others78 have shown that patients with
PUVs managed by a limited intervention approach had
better outcomes than patients who underwent extensive
urologic procedures. Nonetheless, transplantation into a
nonreconstructed valve bladder and into an augmented
bladder can yield acceptable graft survival rates.28 With
the lack of controlled studies of patients with PUVs to
define the possible advantages and risks of lower urinary
tract reconstruction, no recommendations can be made
based on the available evidence as to the indications of
bladder augmentation in this condition.
In addition, one study indicates that the rate of post-
transplantation UTIs is greater in patients with a history
of PUV, regardless of the presence of reflux.6 This infor-
mation is important, not to discourage renal transplanta-
tion in young patients with a history of PUV, but rather
to pay particular attention to bladder care in these cases.
It would seem rational to do everything feasible to opti-
mize bladder function before transplantation by improv-
Fig. 12.7 Fluoroscopic image of voiding cystometrogram during uro- ing emptying, decreasing storage pressures, and providing
dynamics in an adult patient with a valve bladder, dilated posterior ure- adequate capacity. When evaluating these bladders, it must
thra, and cone-shaped bladder. be remembered that what is considered adequate bladder
12 • Transplantation and the Abnormal Bladder 181

capacity and compliance varies with the obligatory diuresis patient has progressed to 6 months posttransplant, allow-
of a given patient. Inadequate capacity in a polyuric child ing improvements in functional bladder capacity and meta-
with ESRD may become acceptable after the transplant bolic anomalies.
when the urine output normalizes.
FEMALE VOIDING DYSFUNCTION
PRUNE-BELLY SYNDROME
Common causes of bladder outlet obstruction and voiding
Renal failure develops in a significant number of patients dysfunction in adult females include severe organ prolapse
born with PBS. The causes are renal dysplasia, obstruc- and prior incontinence surgery. Functional bladder outlet
tion, and pyelonephritis.4 The first renal transplant in a obstruction and detrusor underactivity are also prevalent
patient with PBS was reported in 1976 by Shenasky and in women. The AUA symptom score has been shown to be
Whelchel,79 followed by other single-case reports. In 1989 a poor screening tool for defining severity of bladder func-
Reinberg and colleagues reported on a series of children tion in women with LUTS.87 Given the fact that no standard
with PBS that were transplanted, with results similar to definition for BOO exists in females and screening uroflow-
those of a control group.80 These results were confirmed by metry has limitations in recipient females, VUDs is the gold
Fontaine and colleagues68 in 1997. This is not surprising standard in diagnosis of female voiding dysfunction when
because bladder storage pressures are low in most cases of associated with pelvic floor abnormalities. Pelvic organ pro-
this syndrome. Later, an Italian group published their expe- lapse management and incontinence surgery with synthetic
rience with a series of five boys and reported good results mesh in transplant recipients improves quality of life and is
as well, but they stressed the need to address the lack of feasible.88,89 Our approach is the same as in nontransplant
abdominal wall musculature by performing abdominal wall patients, although we reserve all operative procedures for at
reconstruction in selected patients.81 A unique complica- least 6 months after renal transplantation.
tion specific to renal transplantation performed in patients
with PBS is torsion of the graft. Whether this complication
occurred because of the lax abdominal musculature or the Conclusion
fact that the kidney was grafted intraperitoneally is not
clear.82,83 ESRD caused by congenital genitourinary anomalies is
common, especially in pediatric patients. Furthermore,
lower urinary tract dysfunction is seen frequently in adult
MALE VOIDING DYSFUNCTION, BPH/LUTS
transplant candidates and recipients. Integrity of the lower
There are some special scenarios that warrant atten- urinary tract is mandatory before transplantation, and
tion. The mean age of transplant recipients is increasing proper investigation should be done in selected patients,
in the US as a result of improvement in care.84 In addi- ideally by a urologic specialist. Graft implantation into the
tion, because a greater number of older men are undergo- native bladder is always preferred. If the bladder is unsuit-
ing renal transplantation there are more issues with BPH. able, planning a multidisciplinary approach is advocated,
BPH is a chronic age-related condition and may lead to and staged reconstitution of the lower urinary tract is fea-
bladder outlet obstruction (BOO) and associated LUTS. sible. Bladder reconstruction may be done at the time of
The incidence of BPH/LUTS in this group may be masked transplant or prior when clinically feasible. Although not
in the pretransplant population because of decreased or exempt from complications, reconstruction is an acceptable
absence of diuresis. Adult male transplant recipients with method for patients with an abnormal lower urinary tract
BPH and LUTS should be treated medically similarly to who are candidates for renal transplantation. Posttrans-
their healthier counterparts. With regard to invasive surgi- plant monitoring of LUTS by a urologic specialist is man-
cal treatment, special considerations need to be taken into datory because procedures to correct incontinence, voiding
account. The gold standard outlet procedure is a transure- dysfunction, and bladder outlet obstruction have excellent
thral resection of the prostate (TURP), and the periopera- results in transplant recipients. Finally, even if the reported
tive surgical risks need to be carefully balanced. Those who series of renal transplantation into abnormal bladders are
are in active renal replacement therapy have chronic ure- small, the graft and patient survival rates in most series
mia-associated platelet dysfunction, higher cardiovascular seem to be comparable to the rates for transplants into non-
risk, and electrolyte and fluid imbalances, which increase reconstructed bladders.
the risk of the procedure. In addition, those who produce
minimal or low volumes (<500 mL/day) of urine may be References
at additional risk from low volume flow through the ure- 1. Burns MW, Watkins SL, Mitchell ME, Tapper D. Treatment of bladder
thra in the postoperative phase. These include poor healing dysfunction in children with end-stage renal disease. J Pediatr Surg
1992;27:170–4.
and stricture and scar formation from a “dry” TURP cav- 2. Ewalt DH, Allen TD. Urinary tract reconstruction in children undergo-
ity. Despite the immunocompromised state, posttransplant ing renal transplantation. Adv Ren Replace Ther 1996;3:69–76.
TURP is safe when indicated as the patient is metabolically 3. Lopez Pereira P, Espinosa L, Martinez Urrutina MJ, Lobato R, Navarro
optimized and overall cardiovascular risks improved. When M, Jaureguizar E. Posterior urethral valves: prognostic factors. BJU Int
performed at 6 months after transplantation, TURP has 2003;91:687–90.
4. Reinberg Y, Manivel JC, Pettinato G, Gonzalez R. Development
excellent safety and outcome profile and is durable.85 When of renal failure in children with the prune belly syndrome. J Urol
TURP is performed less than 60 days after transplantation, 1991;145:1017–9.
the incidence of UTI and loss of renal function is increased.86 5. Warne SA, Hiorns MP, Curry J, Mushtaq I. Understanding cloacal
Hence our approach is to perform a staged TURP once the anomalies. Arch Dis Child 2011;96:1072–6.
182 Kidney Transplantation: Principles and Practice

6. Reinberg Y, Gonzalez R, Fryd D, Mauer SM, Najarian JS. The outcome 32. Dykstra DD, Sidi AA. Treatment of detrusor-sphincter dyssynergia
of renal transplantation in children with posterior urethral valves. with botulinum A toxin: a double-blind study. Arch Phys Med Rehabil
J Urol 1988;140:1491–3. 1990;71:24–6.
7. Bryant JE, Joseph DB, Kohaut EC, Diethelm AG. Renal transplantation 33. Lapides J, Diokno AC, Silber SJ, Lowe BS. Clean, intermittent self-
in children with posterior urethral valves. J Urol 1991;146:1585–7. catheterization in the treatment of urinary tract disease. J Urol
8. Lopez Pereira P, Martinez Urrutia MJ, Espinosa L, Lobato R, Navarro 1972;107:458–61.
M, Jaureguizar E. Bladder dysfunction as a prognostic factor in 34. Flechner SM, Conley SB, Brewer ED, Benson GS, Corriere Jr JN. Inter-
patients with posterior urethral valves. BJU Int 2002;90:308–11. mittent clean catheterization: an alternative to diversion in continent
9. Kamal MM, El-Hefnawy AS, Soliman S, Shokeir AA, Ghoneim MA. transplant recipients with lower urinary tract dysfunction. J Urol
Impact of posterior urethral valves on pediatric renal transplantation: 1983;130:878–81.
a single-center comparative study of 297 cases. Pediatr Transplant 35. Bouchot O, Guillonneau B, Cantarovich D, et al. Vesicoureteral reflux
2011;15:482–7. in the renal transplantation candidate. Eur Urol 1991;20:26–8.
10. Montane B, Abitbol C, Seeherunvong W, et al. Beneficial effects of con- 36. Erturk E, Burzon DT, Orloff M, Rabinowitz R. Outcome of patients with
tinuous overnight catheter drainage in children with polyuric renal vesicoureteral reflux after renal transplantation: the effect of pretrans-
failure. BJU Int 2003;92:447–51. plantation surgery on posttransplant urinary tract infections. Urology
11. Lopez Pereira P, Jaureguizar E, Martinez Urrutia MJ, Meseguer C, 1998;51:27–30.
Navarro M. Does treatment of bladder dysfunction prior to renal 37. Mildenberger H, Habenicht R, Zimmermann H. Infants with posterior
transplant improve outcome in patients with posterior urethral urethral valves: a retrospective study and consequences for therapy.
valves? Pediatr Transplant 2000;4:118–22. Prog Pediatr Surg 1989;23:104–12.
12. Thibodeau BA, Metcalfe P, Koop P, Moore K. Urinary incontinence 38. Errando C, Batista JE, Caparros J, Arano P, Villavicencio H. Is bladder
and quality of life in children. J Pediatr Urol 2013;9:78–83. cycling useful in the urodynamic evaluation previous to renal trans-
13. Chen JL, Lee MC, Kuo HC. Reduction of cystometric bladder capac- plantation? Urol Int 2005;74:341–5.
ity and bladder compliance with time in patients with end-stage renal 39. Horst M, Weber DM, Bodmer C, Gobet R. Repeated botulinum-A
disease. J Formos Med Assoc 2012;111:209–13. toxin injection in the treatment of neuropathic bladder dysfunction
14. Chun JM, Jung GO, Park JB, et al. Renal transplantation in patients and poor bladder compliance in children with myelomeningocele.
with a small bladder. Transplant Proc 2008;40:2333–5. Neurourol Urodyn 2011;30:1546–9.
15. Van der Weide MJ, Cornelissen EA, Van Achterberg T, de Gier RP, 40. Teng L, Wang C, Li J. Long-term outcome of simultaneous or staged
Feitz WF. Lower urinary tract symptoms after renal transplantation urinary diversion and kidney transplantation. Urol Int 2013;91:
in children. J Urol 2006;175:297–302; discussion. 310–4.
16. Herthelius M, Oborn H. Bladder dysfunction in children and adoles- 41. Prabharasuth D, Moses KA, Bernstein M, Dalbagni G, Herr HW.
cents after renal transplantation. Pediatr Nephrol 2006;21:725–8. Management of bladder cancer after renal transplantation. Urology
17. Kashi SH, Wynne KS, Sadek SA, Lodge JP. An evaluation of vesi- 2013;81:813–9.
cal urodynamics before renal transplantation and its effect on renal 42. Demirdag C, Citgez S, Talat Z, Onal B. Management of bladder cancer
allograft function and survival. Transplantation 1994;57:1455–7. after renal transplantation. Transplant Proc 2017;49:293–6.
18. Ramirez SP, Lebowitz RL, Harmon WE, Somers MJ. Predictors for 43. Chaykovska L, Deger S, Wille A, et al. Kidney transplantation into
abnormal voiding cystourethrography in pediatric patients undergo- urinary conduits with ureteroureterostomy between transplant and
ing renal transplant evaluation. Pediatr Transplant 2001;5:99–104. native ureter: single-center experience. Urology 2009;73:380–5.
19. Reynard JM, Peters TJ, Lim C, Abrams P. The value of multiple free- 44. Nguyen DH, Reinberg Y, Gonzalez R, Fryd D, Najarian JS. Outcome of
flow studies in men with lower urinary tract symptoms. Br J Urol renal transplantation after urinary diversion and enterocystoplasty: a
1996;77:813–8. retrospective, controlled study. J Urol 1990;144:1349–51.
20. Hjalmas K. Urodynamics in normal infants and children. Scand J Urol 45. Kocot A, Spahn M, Loeser A, Lopau K, Gerharz EW, Riedmiller H.
Nephrol Suppl 1988;114:20–7. Long-term results of a staged approach: continent urinary diversion
21. Neveus T, von Gontard A, Hoebeke P, et al. The standardization of ter- in preparation for renal transplantation. J Urol 2010;184:2038–42.
minology of lower urinary tract function in children and adolescents: 46. Hamdi M, Mohan P, Little DM, Hickey DP. Successful renal transplan-
report from the Standardisation Committee of the International Chil- tation in children with spina bifida: long term single center experi-
dren’s Continence Society. J Urol 2006;176:314–24. ence. Pediatr Transplant 2004;8:167–70.
22. Winters JC, Dmochowski RR, Goldman HB, et al. Urodynamic studies 47. Power RE, O’Malley KJ, Little DM, et al. Long-term followup of
in adults: AUA/SUFU guideline. J Urol 2012;188:2464–72. cadaveric renal transplantation in patients with spina bifida. J Urol
23. Errando C, Batista JE, Caparros J, Vicente J, Arano P. Urodynamic 2002;167:477–9.
evaluation and management prior to renal transplantation. Eur Urol 48. Djakovic N, Wagener N, Adams J, et al. Intestinal reconstruction of
2000;38:415–8. the lower urinary tract as a prerequisite for renal transplantation. BJU
24. Glazier DB, Whang MI, Geffner SR, et al. Evaluation of voiding cysto- Int 2009;103:1555–60.
urethrography prior to renal transplantation. Transplantation 1996 49. Sullivan ME, Reynard JM, Cranston DW. Renal transplantation into
27;62:1762–5. the abnormal lower urinary tract. BJU Int 2003;92:510–5.
25. Fairhurst JJ, Rubin CM, Hyde I, Freeman NV, Williams JD. Bladder 50. Thomalla JV, Mitchell ME, Leapman SB, Filo RS. Renal transplanta-
capacity in infants. J Pediatr Surg 1991;26:55–7. tion into the reconstructed bladder. J Urol 1989;141:265–8.
26. MacGregor P, Novick AC, Cunningham R, et al. Renal transplantation 51. de Jong TP, Donckerwolcke RA, Boemers TM. Neomycin toxicity in
in end stage renal disease patients with existing urinary diversion. bladder irrigation. J Urol 1993;150:1199.
J Urol 1986;135:686–8. 52. Basiri A, Otookesh H, Hosseini R, Simforoosh N, Moghaddam SM. Kid-
27. McGuire EJ, Woodside JR, Borden TA, Weiss RM. Prognostic value ney transplantation before or after augmentation cystoplasty in chil-
of urodynamic testing in myelodysplastic patients. J Urol 1981; dren with high-pressure neurogenic bladder. BJU Int 2009;103:86–8;
126:205–9. discussion 88.
28. Abraham MK, Nasir AR, Sudarsanan B, et al. Role of alpha adrenergic 53. Broniszczak D, Ismail H, Nachulewicz P, et al. Kidney transplantation
blocker in the management of posterior urethral valves. Pediatr Surg in children with bladder augmentation or ileal conduit diversion. Eur
Int 2009;25:1113–5. J Pediatr Surg 2010;20:5–10.
29. Austin P. The role of alpha blockers in children with dysfunctional 54. Barry JM. Kidney transplantation into patients with abnormal blad-
voiding. Sci World J 2009 1;9:880–3. ders. Transplantation 2004 15;77:1120–3.
30. Costantini E, Lazzeri M, Bini V, et al. Open-label, longitudinal study of 55. Neild GH, Dakmish A, Wood S, Nauth-Misir R, Woodhouse CR. Renal
tamsulosin for functional bladder outlet obstruction in women. Urol transplantation in adults with abnormal bladders. Transplantation
Int 2009;83:311–5. 2004 15;77:1123–7.
31. Kaplan SA, Roehrborn CG, Chancellor M, Carlsson M, Bavendam 56. Peterson AC, Webster GD. Artificial urinary sphincter: lessons
T, Guan Z. Extended-release tolterodine with or without tamsulosin learned. Urol Clin North Am 2011;38:83–8. vii.
in men with lower urinary tract symptoms and overactive bladder: 57. Kryger JV, Gonzalez R, Barthold JS. Surgical management of urinary
effects on urinary symptoms assessed by the International Prostate incontinence in children with neurogenic sphincteric incompetence.
Symptom Score. BJU Int 2008;102:1133–9. J Urol 2000;163:256–63.
12 • Transplantation and the Abnormal Bladder 183

58. Raj GV, Peterson AC, Webster GD. Outcomes following erosions of the 74. Adams J, Mehls O, Wiesel M. Pediatric renal transplantation and the
artificial urinary sphincter. J Urol 2006;175:2186–90; discussion 2190. dysfunctional bladder. Transpl Int 2004;17:596–602.
59. Vainrib M, Reyblat P, Ginsberg D. Outcomes of male sling mesh kit 75. Groenewegen AA, Sukhai RN, Nauta J, Scholtmeyer RJ, Nijman RJ.
placement in patients with neuropathic stress urinary incontinence: a Results of renal transplantation in boys treated for posterior urethral
single institution experience. Urol Int 2015;95:406–10. valves. J Urol 1993;149:1517–20.
60. Stein R, Fisch M, Ermert A, et al. Urinary diversion and orthotopic 76. Salomon L, Fontaine E, Guest G, Gagnadoux MF, Broyer M, Beurton
bladder substitution in children and young adults with neurogenic D. Role of the bladder in delayed failure of kidney transplants in boys
bladder: a safe option for treatment? J Urol 2000;163:568–73. with posterior urethral valves. J Urol 2000;163:1282–5.
61. Warholm C, Berglund J, Andersson J, Tyden G. Renal transplantation 77. Dewan PA, McMullin ND, Barker AP. Renal allograft survival in
in patients with urinary diversion: a case-control study. Nephrol Dial patients with congenital obstruction of the posterior urethra. Aust NZ
Transplant 1999;14:2937–40. J Surg 1995;65:27–30.
62. Nahas WC, Mazzucchi E, Arap MA, et al. Augmentation cystoplasty 78. Bartsch L, Sarwal M, Orlandi P, Yorgin PD, Salvatierra Jr O. Lim-
in renal transplantation: a good and safe option—experience with 25 ited surgical interventions in children with posterior urethral valves
cases. Urology 2002;60:770–4. can lead to better outcomes following renal transplantation. Pediatr
63. Riedmiller H, Gerharz EW, Kohl U, Weingartner K. Continent urinary Transplant 2002;6:400–5.
diversion in preparation for renal transplantation: a staged approach. 79. Shenasky II JH, Whelchel JD. Renal transplantation in prune belly
Transplantation 2000;70:1713–7. syndrome. J Urol 1976;115:112–3.
64. Hatch DA, Koyle MA, Baskin LS, et al. Kidney transplantation in 80. Reinberg Y, Manivel JC, Fryd D, Najarian JS, Gonzalez R. The outcome
children with urinary diversion or bladder augmentation. J Urol of renal transplantation in children with the prune belly syndrome.
2001;165:2265–8. J Urol 1989;142:1541–2.
65. Martin MG, Castro SN, Castelo LA, Abal VC, Rodriguez JS, Novo 81. Fusaro F, Zanon GF, Ferreli AM, et al. Renal transplantation in prune-
JD. Enterocystoplasty and renal transplantation. J Urol 2001;165: belly syndrome. Transpl Int 2004;17:549–52.
393–6. 82. Marvin RG, Halff GA, Elshihabi I. Renal allograft torsion associated
66. DeFoor W, Minevich E, McEnery P, Tackett L, Reeves D, Sheldon C. with prune-belly syndrome. Pediatr Nephrol 1995;9:81–2.
Lower urinary tract reconstruction is safe and effective in children with 83. Abbitt PL, Chevalier RL, Rodgers BM, Howards SS. Acute torsion of a
end stage renal disease. J Urol 2003;170:1497–500; discussion 1500. renal transplant: cause of organ loss. Pediatr Nephrol 1990;4:174–5.
67. Rischmann P, Malavaud B, Bitker MO, et al. Results of 51 renal 84. United States Renal Data System. 2014 USRDS annual data report: Epi-
transplants with the use of bowel conduits in patients with impaired demiology of kidney disease in the United States. Bethesda, MD: National
bladder function: a retrospective multicenter study. Transplant Proc Institutes of Health, National Institute of Diabetes and Digestive and
1995;27:2427–9. Kidney Diseases; 2014.
68. Fontaine E, Salomon L, Gagnadoux MF, Niaudet P, Broyer M, Beur- 85. Volpe A, Billia M, Quaglia M, et al. Transurethral resection of the pros-
ton D. Long-term results of renal transplantation in children with the tate in kidney transplant recipients: urological and renal functional
prune-belly syndrome. J Urol 1997;158:892–4. outcomes at long-term follow-up. BJU Int 2013;112:386–93.
69. Sheldon CA, Gonzalez R, Burns MW, Gilbert A, Buson H, Mitchell 86. Piovesan AC, Andrade H, Messi GB, et al. Transurethral resection or
ME. Renal transplantation into the dysfunctional bladder: the role of incision of the prostate after renal transplantation: is there a safe time
adjunctive bladder reconstruction. J Urol 1994;152:972–5. for the procedure? J Urol 2014;191:E777.
70. Campaiola JM, Perlmutter AD, Steinhardt GF. Noncompliant bladder 87. Min-Hsin Y, Yu-Lin K, Sun-Lang C, Shao-Chuan W, Wen-Jung C,
resulting from posterior urethral valves. J Urol 1985;134:708–10. Zuou-Yi S. Characteristics of voiding dysfunction in renal transplant
71. Peters CA, Bauer SB. Evaluation and management of urinary inconti- recipients. ICS Tokyo. 2016: ePoster Station 7, Abstract 142. Avail-
nence after surgery for posterior urethral valves. Urol Clin North Am able online at: https://www.ics.org/2016/abstract/142. Accessed
1990;17:379–87. January 24, 2018.
72. Connolly JA, Miller B, Bretan PN. Renal transplantation in patients 88. Hoda MR, Wagner S, Greco F, Heynemann H, Fornara P. Pelvic organ
with posterior urethral valves: favorable long-term outcome. J Urol prolapse management in female kidney transplant recipients. J Urol
1995;154:1153–5. 2010;184:1064–8.
73. Ross JH, Kay R, Novick AC, Hayes JM, Hodge EE, Streem SB. Long- 89. Shveiky D, Blatt A, Sokol AI, Nghiem HG, Iglesia CB. Pelvic recon-
term results of renal transplantation into the valve bladder. J Urol structive surgery in renal transplant recipients. Int Urogynecol J Pel-
1994;151:1500–4. vic Floor Dysfunct 2009;20:551–5.
13 Perioperative Care of
Patients Undergoing
Kidney Transplantation
KATE KRONISH, ANDREA OLMOS, and CLAUS U. NIEMANN

CHAPTER OUTLINE Comorbidities in End-Stage Renal Disease Anesthesia for Patients After Kidney
Cardiovascular Complications Transplantation
Hematologic Abnormalities Kidney–Pancreas Transplantation
Uremia Preoperative Considerations
Preoperative Considerations Intraoperative Considerations
Intraoperative Considerations Postoperative Care
Postoperative Care

The first description of anesthesia for kidney transplanta- Comorbidities in End-Stage Renal
tion (KTx) appeared in the early 1960s. It details the pio-
neering efforts in Boston with living related KTx between
Disease
identical twins.1 The only monitors used in the 17 initial
CARDIOVASCULAR COMPLICATIONS
recipient cases described were a blood pressure cuff and an
electrocardiogram (ECG). All recipients received neuraxial The two main cardiovascular complications of chronic
anesthesia. Within a few years, general anesthesia had renal failure are arterial hypertension and atherosclero-
become the norm and the first generation of immunosup- sis, both of which predispose the patient to ischemic heart
pressants emerged, providing better deceased donor graft disease. The prevalence of preoperative hypertension in
survival. As a result, the number of kidney transplants per- patients undergoing renal transplantation ranges from
formed increased significantly.2 50% to 80%.7 Hypertension in chronic renal disease devel-
Today, KTx is performed in most countries in the world, ops as a consequence of volume expansion secondary to
with the most active ones being the US, Canada, Austra- salt and water retention.8 Other factors that contribute
lia, and most European countries, where the rate is greater to the pathophysiology of hypertension in chronic kidney
than 35 per million citizens.3 Worldwide more than disease include dysregulation of the renin-angiotensin-
79,000 KTxs were performed in 2014. The kidney remains aldosterone system, sympathetic overactivity, imbalance of
the most commonly transplanted solid organ. Neverthe- endothelium-derived vasoactive substances (endothelin-1
less, the need for KTx far outnumbers the transplantation and nitric oxide), erythropoietin replacement therapy, and
rate. In the US in 2016, there were more than 19,000 secondary hyperparathyroidism.9 If untreated, elevated
KTxs performed but more than 96,000 patients remained systemic pressure within the kidney can cause sclerotic
on the waitlist.4 In fact, the active waitlist grew 27% from changes in the renal vasculature that further contribute to
10 years prior.5 a vicious cycle of increasing hypertension and accelerated
Despite much progress and advancement, kidney trans- kidney injury.10
plant patients continue to be a challenge during the peri- The combination of fluid overload and increased systemic
operative period. End-stage renal disease (ESRD) itself vascular resistance leads to increased myocardial afterload
frequently results in the dysfunction of other major organ and wall stress. The heart is able to partially compensate
systems. In addition, the underlying cause of renal dis- with left ventricular (LV) hypertrophy. However, LV hyper-
ease may be associated with other important comorbidi- trophy and elevated wall stress result in increased myocar-
ties. These patients are often at high risk for cardiac and dial oxygen requirements. At the same time, a rise in LV
other perioperative complications. Kidney transplanta- end-diastolic pressure reduces subendocardial coronary
tion remains the standard of care in patients with ESRD, perfusion, reducing myocardial oxygen supply. Together,
imparting a significant survival advantage. After under- these forces increase the risk of myocardial ischemia.11
going renal transplantation, patients are 10 times less Cardiac dysfunction may improve with the initiation of
likely to die of cardiovascular disease than patients on dialysis and the appropriate antihypertensive therapy. In
hemodialysis.6 patients in whom the hypertension cannot be controlled
184
13 • Perioperative Care of Patients Undergoing Kidney Transplantation 185

by dialysis alone, an abnormal relationship may exist concentric hypertrophy are seen in dialysis patients. The
among plasma renin activity, intravascular fluid volume, accumulation of uremic toxins and metabolic acids also
blood pressure, and inappropriate levels of sympathetic contributes to poor myocardial performance. Fluid overload
activity.9,12 Patients needing antihypertensive therapy and congestive heart failure occur when the kidneys can-
in addition to dialysis are often refractory to single anti- not excrete the daily fluid. Other cardiac conditions, such
hypertensive drug regimens and require a combination as pericardial disease and arrhythmia, may be encountered
of antihypertensive drugs, which can have significant in patients with ESRD. Pericarditis, which may coexist with
implications for the perioperative period. Angiotensin-con- hemorrhagic pericardial effusion, may reverse with dialy-
verting enzyme (ACE) inhibitors and angiotensin receptor sis.29 Arrhythmias may result from electrolyte abnormali-
blockers (ARB) are two of the most commonly used anti- ties or MI.
hypertensive agents in renal disease.13 Other commonly
used antihypertensives include loop and thiazide diuretics, HEMATOLOGIC ABNORMALITIES
dihydropyridine calcium channel blockers, beta blockers,
and hydralazine. A prospective study by Sear et al. demon­ Patients in renal failure most often have normochromic,
strated no difference in hemodynamic response to the normocytic anemia due to decreased erythropoietin syn-
induction of anesthesia and laryngoscopy between patients thesis and release. Other factors contributing to anemia
with mild to moderate hypertension on different single in renal failure include a decreased red blood cell lifespan,
antihypertensive drug regimens.14 However, more recent increased hemolysis and bleeding, repeated blood loss dur-
studies have shown an increase in the incidence of intra- ing hemodialysis, aluminum toxicity, uremia-induced bone
operative hypotension in patients taking ACE inhibitors or marrow suppression, and iron, folate, and vitamin B6 and
ARBs.15–17 Furthermore, patients with poorly controlled B12 deficiencies. Treatment with recombinant erythropoi-
hypertension or requiring several antihypertensive agents etin can frequently raise hemoglobin levels to 10 to 13 g/
often experience significant hemodynamic lability during dL,30 which reduces symptoms of fatigue and improves
general anesthesia. cerebral and cardiac function.31 Pretransplant treat-
Chronic kidney disease can accelerate the progression of ment with recombinant erythropoietin in anemic patients
atherosclerosis, including coronary artery disease (CAD), improves long-term graft survival compared with blood
likely via modulation of lipid metabolism, which leads transfusion, which causes allostimulation.32 However,
to dyslipidemia and accumulation of atherogenic parti- preexisting hypertension can worsen with erythropoietin
cles.18,19 Cardiac disease is particularly prominent in ESRD therapy in some patients.30
patients with diabetes mellitus (DM). DM type 2 is the cause An association between renal failure and bleeding ten-
of ESRD in nearly 40% of patients.20 Patients with ESRD dency has long been recognized. Uremia produces a quali-
and DM have higher cardiovascular risk than do patients tative defect in platelet function wherein decreased levels
with uremia alone because of the acceleration of small-ves- of platelet factor III impair platelet adhesion. A prolonged
sel atherosclerosis associated with DM and the frequently bleeding time is seen (although with poor clinical utility),
concomitant metabolic syndrome.21 although prothrombin and partial thromboplastin time
The prevalence of CAD in patients with ESRD has been are usually normal. Treatments for uremic coagulopa-
reported in the range of 42% to 80%.22 Acute myocardial thy include dialysis, platelet transfusion, cryoprecipitate,
infarction (MI), cardiac arrest of unknown etiology, car- and desmopressin (0.3 mcg/kg). Recent studies suggest
diac arrhythmia, and cardiomyopathy account for over a prothrombotic state may in fact exist with uremia.
50% of deaths in patients maintained on dialysis.23 Car- A thromboelastographic study of whole blood clotting
diac mortality in dialysis patients increases with age. It is found increased coagulability and decreased fibrinoly-
approximately two times higher for 45-to 64-year-olds and sis in uremic patients versus controls.33 Platelet-derived
four times higher in patients older than 65 compared with procoagulant microparticles may be involved in clinical
younger patients in the 20-to 44-year-old range.23 thrombogenesis.34
Cardiovascular disease remains one of the largest
causes of death even after successful KTx, accounting UREMIA
for 17% to 50% of deaths in KTx recipients.23,24 Humar
and colleagues reported a 6.1% overall perioperative Uremia can cause central nervous system disturbances
cardiac complication rate among 2694 KTx recipients.25 ranging from drowsiness, memory loss, and decreased con-
Another large study by Gill and Pereira reported a 4.6% centration to myoclonus, seizures, stupor, and coma. How-
first-year all-cause mortality rate in 23,546 adult KTx ever, severe uremic central nervous system disturbances
patients, with greater than 25% of these being second- are rarely seen in patients when appropriately dialyzed.
ary to cardiac causes.26 The main predictors of adverse Chronic uremia may also cause delayed gastric emptying.
outcome were a history of pretransplant cardiac disease Although the mechanism is not understood, gastric dys-
or MI within the previous 6 months and age older than rhythmia with discoordinated myoelectrical activity has
40 years. been found in uremic patients on maintenance hemodialy-
When echocardiography is performed on dialysis patients sis.2 In addition, renal failure patients have an increase in
as a screening tool, a high incidence of abnormalities is acidity and gastric volume unrelated to Helicobacter pylori
found.27 In one study, left or right ventricular hypertrophy infection.35,36 There appears to be no difference in gastric
or pericarditis was detected in 60% of autopsies performed emptying time between patients undergoing peritoneal
on dialysis patients.28 Both dilated cardiomyopathy and dialysis and hemodialysis.
186 Kidney Transplantation: Principles and Practice

Preoperative Considerations discrepancies exist among the guidelines and most do not
consider the unique clinical characteristics of patients with
Preoperative evaluation begins with assessing medical ESRD. Not surprisingly, there is no absolute consensus on the
suitability before listing for transplantation. Careful assess- optimal cardiac workup for ESRD patients being evaluated for
ment of cardiorespiratory function is critical. In particular, KTx, and cardiac evaluation practices vary across the US.38
patients must be screened for CAD, congestive heart failure, Several noninvasive screening tests for CAD diagnosis
and autonomic dysfunction before listing. Once the trans- have been studied in this patient population. However, the
plant is scheduled, history and physical evaluation should optimal noninvasive test for kidney transplant candidates
focus on cardiorespiratory complications and comorbidities, has yet to be determined. In a prospective study, Herzog and
volume status, electrolyte derangements, and any recent colleagues performed dobutamine stress echocardiography
infections. With the benefit of advanced planning, living (DSE) before quantitative coronary angiography in a mixed
donor kidney transplant recipients have enough time to population of ESRD patients who were candidates for trans-
be medically optimized before surgery. Although deceased plantation.39 More than 50% of patients had some degree
donor kidney transplants are often scheduled as urgent or of CAD (defined as coronary stenosis >50%). However,
emergency cases, prolonged cold preservation of the kidney the DSE displayed a sensitivity of only 52% to 75% and a
is well tolerated, and recipients should have sufficient time specificity of 74% to 76% for identifying patients with CAD.
to be dialyzed and to address other preoperative issues. These patients were monitored for up to 2 years. During
Almost all patients receiving an organ from a deceased this time, 20% of those with a negative DSE suffered cardiac
organ donor receive some form of dialysis before transplan- death or MI or underwent coronary revascularization. The
tation. When possible, living donation is planned before authors concluded that DSE was a useful but imperfect tool
initiation of dialysis. History of long-term chronic hemo- for identifying patients needing further cardiac workup. In
dialysis decreases graft function, but less than 6 months a subsequent study, Sharma and colleagues performed DSE
of dialysis was not found to decrease patient or graft sur- and baseline troponin measurements in 118 ESRD patients
vival.37 Patients maintained on hemodialysis usually referred for KTx and found significant CAD (>70% steno-
undergo a dialysis session during the 24- to 36-hour period sis on angiography) in 30% of patients, abnormal DSE in
before transplantation to correct electrolyte imbalances 35%, and elevated troponin in 26%. DSE had a sensitivity
and optimize volume status before surgery. Patients main- and specificity of 88% and 94%, respectively, for detecting
tained on continuous ambulatory peritoneal dialysis often significant CAD, whereas an elevated troponin had a sensi-
have more stable volume status and electrolytes. tivity and specificity of 54% and 62%, respectively.40
Determination of volume status should include an assess- A Cochrane Review meta-analysis investigated the accu-
ment for dyspnea, orthopnea, and lower extremity edema. racy of DSE and myocardial perfusion scintigraphy (MPS)
Patients should be questioned about how much urine vol- compared with coronary angiography to detect CAD in
ume they produce, and dry weight should be compared with kidney transplant candidates.41 These authors identified
current weight. Removing fluid with hemodialysis before 13 studies in which DSE was the screening test and nine
surgery facilitates perioperative fluid management, as 40 studies in which MPS was the screening test for CAD. They
to 50 mL/kg of crystalloid is often given intraoperatively. reported that DSE had an aggregate sensitivity and specific-
Nevertheless, overly aggressive removal of fluid during ity for detecting CAD of 79% and 89% whereas MPS had a
preoperative dialysis may make patients hypovolemic and sensitivity and specificity of 74% and 70%. Pooled sensitiv-
put them at risk for significant intraoperative hypotension, ity and specificity of each screening test changed only mar-
especially during induction of anesthesia. The production of ginally when the authors limited their analysis to studies
urine may be delayed if the new graft does not begin func- that defined CAD as >70% stenosis on angiography. DSE
tioning immediately. These patients may require hemodial- showed improved accuracy over MPS for detecting CAD
ysis postoperatively until the new graft begins functioning. (P = 0.02) when all 22 studies were included in the anal-
When patients without hemodialysis access are to receive ysis; however, this difference became insignificant when
potentially marginal grafts, plans can be made for the place- the authors excluded studies that did not use a reference
ment of hemodialysis lines intraoperatively. threshold of ≥70% stenosis on coronary angiography.
Recent electrolyte concentrations, particularly K+ and Although noninvasive testing for CAD in ESRD is cer-
HCO3−, should be reviewed preoperatively. Patients receiv- tainly imperfect, abnormal MPS and DSE test results have
ing hemodialysis may experience significant swings in K+ been associated with prognostic value for major adverse
and HCO3− levels. Therefore, patients who are hemodialy- cardiac events (MACE) in this patient population. In a study
sis-dependent may require serum potassium levels checked of 126 patients with ESRD who underwent technetium-
on the day of surgery. Potassium concentrations greater 99m MPS as part of their pretransplantation assessment,
than 6.0 mEq/L may require a delay in surgery to correct a reversible defect was associated with three times the risk
potassium levels. Chronic hyperkalemia is less likely to of posttransplantation cardiac events and nearly twice the
cause arrhythmia than acute hyperkalemia, thus evaluat- risk of death compared with normal test results.42 A meta-
ing trends in potassium levels is useful. analysis of 12 studies involving either thallium-201 scin-
Cardiac risk assessment is an important component of tigraphy or DSE demonstrated that ESRD patients with
the preoperative evaluation and is dictated by the underly- inducible ischemia had six times the risk of MI and four
ing renal disease, its duration, and attendant comorbidities. times the risk of cardiac death as patients without inducible
National organizations have published guidelines and posi- defects, whereas patients with fixed defects had nearly five
tion papers to inform cardiac evaluation practices; however, times the risk of cardiac death.43
13 • Perioperative Care of Patients Undergoing Kidney Transplantation 187

Other studies suggest that cardiac risk assessment should management. In the COURAGE trial, an examination of the
begin with the analysis of easily obtainable clinical variables subgroup of patients with chronic kidney disease found no
rather than with the wide pursuit of expensive tests with benefit of percutaneous coronary intervention versus medi-
limited sensitivity and specificity.44 For example, a history cal management.49–51
of chest pain is a helpful starting point in detecting CAD in Medical management for these patients at high periop-
these patients because it has a sensitivity and specificity of erative cardiac risk often includes beta-blockade. Several
65% for CAD.45 A more comprehensive system, the revised studies throughout the late 1990s established that periop-
cardiac risk index (RCRI), was originally derived from ret- erative beta-blockade provides significant protection from
rospective data and shown in a prospective population to major cardiac events in high-risk, nontransplant patients,
predict cardiac risk for nonrenal failure patients undergo- although the efficacy of perioperative beta-blockade in
ing noncardiac surgery.46 It focuses on the presence or patients undergoing noncardiac surgery remains contro-
absence of six variables: (1) high-risk surgical procedure; versial. In 2009 the DECREASE-IV trial demonstrated a
(2) history of ischemic heart disease (excluding previous safe and effective way to provide perioperative beta-block-
coronary revascularization); (3) history of heart failure; (4) ade to patients with an estimated risk of MI or cardiovas-
history of stroke or transient ischemic attacks; (5) preopera- cular death greater than 1%.52 However, no prospective
tive insulin therapy; and (6) preoperative creatinine levels randomized trials have been conducted with perioperative
higher than 2 mg/dL (152.5 μmol/L). With zero or one beta-blockade in the ESRD or kidney transplant population.
risk factor, the rate of a major perioperative cardiac event Currently it is unknown whether such treatment can be
is quite low; however, the rates rise rapidly to 6.6% and applied safely to these patients, especially those with DM.
11.0% when two or at least three of these risk factors are No existing guidelines specifically address the evaluation
present. Although the RCRI was not designed specifically and management of pulmonary function in KTx candidates.
for patients with ESRD, a study by Hoftman et al. found this However, a thorough pulmonary history and physical
risk index to be an effective tool for predicting perioperative examination should be performed before transplantation,
cardiovascular complications in patients undergoing KTx. with additional studies pursued when deemed appropriate.
Of the 325 patients included in the study, 7.1% suffered Patients with severe pulmonary limitations, such as a base-
cardiac complications. An increasing number of RCRI risk line oxygen requirement, uncontrolled asthma, cor pulmo-
factors was significantly associated with a higher rate of nale, or severe obstructive or restrictive disease may be too
perioperative cardiac morbidity (receiver operating charac- high risk to undergo KTx surgery. Active smokers should be
teristic area, 0.77; P < 0.0001).47 encouraged to quit preoperatively.53
The guidelines for perioperative cardiovascular evalua- Coagulation status, as reflected by the prothrombin time,
tion and management for noncardiac surgery published by international normalized ratio, partial thromboplastin
the American College of Cardiology (ACC) and the Ameri- time, fibrinogen, and platelet count, is routinely assessed
can Heart Association (AHA) do not take into consideration before surgery. The bleeding time is not a useful screening
the unique clinical characteristics of patients with ESRD. test to predict intraoperative bleeding.54 Patients who are
Additionally, cardiovascular screening and treatment prac- on anticoagulants or antiplatelet agents as maintenance
tices vary widely across transplant programs and are often medications to prevent thrombosis of their dialysis access
inconsistent with published guidelines. In response, the ACC or because of cardiovascular pathology should discon-
and AHA worked with the American Society of Transplant tinue these medications and potentially receive a reversal
Surgeons, the American Society of Transplantation, and agent as soon as they are notified of a transplant. However,
the National Kidney Foundation to develop the expert con- because deceased organ transplants are scheduled for sur-
sensus document entitled “Cardiac Disease Evaluation and gery urgently with minimal advanced notice, anticoagula-
Management Among Kidney and Liver Transplantation tion and antiplatelet therapy often cannot be discontinued
Candidates.”48 This document provides recommendations sufficiently early. One center’s review of 105 patients on
for appropriate cardiovascular screening and management a variety of anticoagulants and antiplatelet medications
of kidney transplantation candidates, as well as ongoing before KTx found no difference in reoperation rates and
surveillance. A summary of these recommendations can be transfusion utilization compared with a control group of
found in Table 13.1. Overall, these guidelines have a lower transplant patients not on these medications.55 Despite
threshold for formal cardiac testing. For example, the guide- these potential coagulation problems, blood loss during
lines recommend that providers consider noninvasive stress renal transplantation is normally less than 250 mL in expe-
testing in kidney transplantation candidates with no active rienced centers. Patients should be typed and crossed for
cardiac conditions based on the presence of multiple CAD blood. It is usually appropriate to have at least two units
risk factors regardless of functional status. Relevant risk of packed red blood cells available given the risk of major
factors in this population include DM, prior cardiovascu- vascular bleeding. More blood products may be ordered in
lar disease, more than 1 year on dialysis, LV hypertrophy, anemic patients and those taking antiplatelet and antico-
age greater than 60 years, smoking, hypertension, and agulant agents.
dyslipidemia. Another preoperative consideration is pregnancy testing
Depending on risk factors and the results of screening in female patients. The American Society of Anesthesiolo-
tests, patients may require medical management or a revas- gists recommends offering pregnancy testing with informed
cularization procedure. The benefit of revascularization is consent to female patients of childbearing age for whom the
controversial. Data from the CARP trial, where revascu- result would alter management. In some institutions, preop-
larization was performed before vascular surgery, showed erative pregnancy testing is performed routinely. Although
no benefit of revascularization compared with medical fertility is decreased in patients with ESRD, irregular menses
188 Kidney Transplantation: Principles and Practice

TABLE 13.1 ACA/AHA Recommendation for Perioperative Cardiac Evaluation of Kidney Transplant Recipients
Evaluation and Management Categories ACA/AHA Recommendations
Determining active cardiac conditions Perform a thorough history and physical examination to identify active cardiac
conditions before solid-organ transplantation.
Noninvasive stress testing in KTx candidates without active Consider noninvasive stress testing in KTx candidates with no active cardiac condi-
cardiac conditions tions based on the presence of multiple CAD risk factors regardless of functional
status. Relevant risk factors among transplantation candidates include DM, prior
cardiovascular disease, more than 1 year on dialysis, left ventricular hypertrophy,
age greater than 60 years, smoking, hypertension, and dyslipidemia.
Cardiac surveillance after listing for transplantation The usefulness of periodically screening asymptomatic KTx candidates for myocar-
dial ischemia while on the transplant waiting list to reduce the risk of MACE is
uncertain.
Resting echocardiography in KTx candidates It is reasonable to perform preoperative assessment of left ventricular function
by echocardiography in potential KTx candidates. There is no evidence for or
against surveillance by repeated left ventricular function tests after listing for
kidney transplantation.
Echocardiographic surveillance for ESRD patients with It may be reasonable to consider ESRD patients with moderate AS to be equivalent
moderate aortic stenosis (AS) to “rapid progressors” who warrant a yearly echocardiogram.
Evaluation and management of KTx candidates with It is reasonable to evaluate KTx candidates with echocardiographic evidence of
echocardiographic evidence of significant pulmonary significant pHTN.
hypertension (pHTN) It may be reasonable to confirm echocardiographic evidence of elevated pulmo-
nary arterial pressures in KTx candidates by right heart catheterization.
If right heart catheterization confirms the presence of significant pulmonary arterial
hypertension, referral to an expert consultant and advanced vasodilator thera-
pies is reasonable.
Preoperative 12-lead ECG in KTx candidates A preoperative resting 12-lead ECG is recommended for potential KTx candidates
with CAD, peripheral arterial disease, or any cardiovascular symptoms.
A preoperative resting 12-lead ECG is reasonable in potential KTx candidates with-
out known cardiovascular disease.
Annual 12-lead ECG after listing for KTx may be reasonable.
Biomarkers for cardiac evaluation in KTx candidates Cardiac troponin level at the time of evaluation for KTx may be considered as an
additional prognostic marker.
Cardiac computed tomography (CT) in KTx candidates The usefulness of noncontrast CT calcium scoring and cardiac CT angiography is
uncertain for the assessment of pretransplantation cardiovascular risk.
Referral to a cardiologist KTx candidates who have an LVEF <50%, evidence of ischemic left ventricular
dilation, exercise-induced hypotension, angina, or ischemia in the distribution of
multiple coronary arteries should be referred to a cardiologist for evaluation and
management according to ACC/AHA guidelines for the general population.
Coronary revascularization and related care before KTx Coronary revascularization before KTx should be considered in patients who meet
the criteria outlined in the “2011 ACCF/AHA Guidelines for Coronary Artery
Bypass Graft Surgery.” In some asymptomatic KTx candidates, the risk of coronary
revascularization may outweigh the risk of transplantation and these risks must
be weighed by the transplantation team on a case-by-case basis until further
studies are performed in this population.
CABG is probably recommended over PCI to improve survival in patients with
multivessel CAD and DM.
CABG may be reasonable for patients with ESRD with significant (>50%) left main
stenosis or significant (>70%) stenoses in three major vessels or in the proximal
LAD artery plus one other major vessel, regardless of left ventricular systolic
function.
It is not recommended that routine prophylactic coronary revascularization be per-
formed in patients with stable CAD, absent symptomatic or survival indications,
before transplantation surgery.
Perioperative medical management of cardiovascular Among patients already taking beta-blockers before KTx, continuing the medica-
risk before KTx tion perioperatively and postoperatively is recommended to prevent rebound
hypertension and tachycardia.
Among potential KTx candidates with clinical markers of cardiac risk (DM, prior
known CAD, prior heart failure, extracardiac atherosclerosis) and those with
unequivocal myocardial ischemia on preoperative stress testing, it is reasonable
to initiate beta blockers preoperatively and to continue them postoperatively pro-
vided that dose titration is done carefully to avoid bradycardia and hypotension.
Perioperative initiation of beta blockers in beta blocker–naïve patients may be con-
sidered in KTx candidates with established CAD or two or more cardiovascular risk
markers to protect against perioperative cardiovascular events if dosing is titrated
and monitored.
Initiating beta blocker therapy in beta blocker–naïve patients the night before and/
or the morning of noncardiac surgery is not recommended.

Modified based on JACC 2012;60(5):152–67.


13 • Perioperative Care of Patients Undergoing Kidney Transplantation 189

can make pregnancy testing especially important. Serum limb with an AV graft or fistula. Occasionally, anesthesiolo-
beta-HCG can be falsely elevated due to ESRD. Urine HCG gists will elect to place the NIBP cuff on a lower extremity
is the recommended confirmatory test in such cases, but to avoid interference with the flow of an intravenous line.
it is often not possible to obtain due to anuria. For patients Before placing a lower extremity NIBP cuff, the anesthesi-
on the transplant list, serial serum beta-HCG testing can be ologist must confirm with the surgeon that the kidney graft
helpful to establish an elevated baseline value that can be will not be transplanted on the same side as the NIBP cuff,
distinguished from the rapid rise in HCG characteristic of as this could compromise blood flow to the new graft.
pregnancy.56,57 Significant acute changes in blood pressure may occur
Patients with DM presenting for KTx can be considered to throughout the surgical procedure, with hypotension (50%)
have full stomachs despite preoperative fasting. Gastric vol- being more likely than hypertension (27%) in one series.59
umes greater than 0.4 mL/kg were seen in 50% of diabetic Patients on multiple antihypertensive medications can have
uremic patients but in only 17% of nondiabetic uremics.58 severe hypotension when volatile and intravenous anes-
The routine prophylactic administration of antacids may be thetic agents are administered, especially during induction
advocated for patients with symptoms of esophageal reflux; of general anesthesia.60,14 Hypotension during KTx has been
a single dose of sodium citrate (30 mL) immediately before associated with delayed graft function, which, in turn, is asso-
surgery is appropriate. Histamine H2-receptor antagonists ciated with short- and long-term complications.61 Hyperten-
can be given to reduce gastric hyper-acidity (e.g., ranitidine sion is commonly seen just before induction of anesthesia
150 mg orally 2 hours before the procedure or ranitidine 50 and during endotracheal intubation, during emergence from
mg IV 30 minutes before surgery). Phenothiazine antiemet- anesthesia, and in the postanesthesia care unit. Several
ics and metoclopramide should be administered with care methods have been used to achieve adequate heart rate and
because they may cause prolonged sedation and extrapyra- blood pressure control during the critical periods of induc-
midal side effects in patients with renal failure. tion and endotracheal intubation, including high potency
opioids, beta blockers, and intravenous lidocaine. Fentanyl,
with a time to peak effect of 3.6 minutes, can blunt the sym-
Intraoperative Considerations pathetic response to laryngoscopy and tracheal intubation
at doses of 2 to 6 mcg/kg.62 However, patients frequently
Spinal anesthesia was used in the first reports of anesthe- experience hypotension immediately and well after induc-
sia for KTx performed in Boston.1 Today, the vast majority tion with these moderate to large doses of fentanyl. Subse-
of transplant centers use general endotracheal anesthe- quently, they will often require vasoconstrictors to maintain
sia; however, neuraxial anesthesia can provide adequate adequate blood pressure, especially because there is little sur-
surgical conditions and excellent postoperative analgesia. gical stimulation once the fascia is dissected. The short-act-
Intraoperative management should focus on tailoring the ing, potent opioid remifentanil, which is rapidly metabolized
anesthetic to the patient’s medical status rather than on in the plasma, is an effective drug for heart rate control both
the type of anesthesia used. Patients presenting for kidney during induction and maintenance of anesthesia. Remifent-
transplant range from the young and otherwise healthy anil dosing can be titrated to rapidly adjust anesthetic depth.
with IgA nephropathy to the elderly with severe hyperten- The short-acting β-adrenergic blocker, esmolol, is an excel-
sion, DM, and CAD. Anesthetic depth and pharmacologic lent choice for blunting the hemodynamic response to endo-
interventions need to be tailored to two different biological tracheal intubation and is well suited for kidney transplant
systems—the transplant recipient and the allograft—with patients with preserved ventricular function. Patients with
sometimes conflicting management needs. For example, long-standing severe hypertension often require high doses
maintenance of adequate anesthetic depth to avoid intra- of esmolol, best given in increments. In a comparison study
operative awareness may also reduce blood pressure and of hemodynamics with fentanyl, lidocaine and esmolol for
perfusion pressure to the newly reperfused graft. Aggressive intubation, esmolol at 2 mg/kg prevented tachycardia and
fluid loading to optimize graft perfusion may be dangerous hypertension with intubation, fentanyl (3 mcg/kg) blocked
in patients with a low ejection fraction and a history of con- hypertension but not tachycardia, and lidocaine (2 mg/kg)
gestive heart failure. was ineffective.63
Intraoperative monitoring includes standard monitors as The single most common agent used for the induction of
recommended by the American Society of Anesthesiologists general anesthesia during KTx is propofol. Other hypnotics
for all patients. Patients with advanced cardiac disease, such such as thiopental and etomidate have also been used suc-
as CAD or heart failure, may require additional invasive cessfully. Several studies have demonstrated that the induc-
monitoring, such as continuous arterial pressure or central tion dose of propofol needed to achieve clinical hypnosis and
venous pressure (CVP) monitoring, or both. Pulmonary appropriate reduction of the bispectral index (a quantitative
artery catheters and transesophageal echocardiography indicator of depth of anesthesia) was 40% to 60% higher in
are rarely indicated. Nevertheless, there is no consensus on patients with ESKD compared with normal patients.64,65 In
appropriate intraoperative monitoring, and most protocols the study by Goyal and colleagues, 0.2 mg/kg of propofol
are based on institutional preference and experience. was titrated every 15 seconds to predefined end points. The
Care must be taken when positioning these patients, with authors found a negative correlation between required pro-
special attention to arteriovenous grafts or fistulae. Grafts pofol dose and preoperative hemoglobin level.64 However,
and fistulae must be properly padded, and anesthesiologists caution is advised when considering these studies. Propofol
should confirm appropriate thrill intermittently through- is a vasodilator and larger induction doses can cause signifi-
out the procedure. Venous and arterial lines and noninva- cant hypotension particularly in volume-depleted patients
sive blood pressure (NIBP) cuffs should not be placed on a dialyzed immediately before surgery.
190 Kidney Transplantation: Principles and Practice

Neuromuscular blockade is useful for both endotracheal concerns or harm associated with sevoflurane administra-
intubation and surgical muscle relaxation. The most com- tion in the setting of a newly transplanted kidney. There are
mon nondepolarizing neuromuscular blocking agents used two elements of potential concern with regard to sevoflu-
today are rocuronium, vecuronium, atracurium, and cisa- rane and renal toxicity: (1) production of fluoride ion from
tracurium. Vecuronium is primarily metabolized and elim- the metabolism of sevoflurane; and (2) generation of “com-
inated by the liver, with up to 25% renally excreted. The pound A” from the breakdown of sevoflurane by sodium
principal metabolite is also a potent neuromuscular blocker or barium hydroxide lime. Sevoflurane appears to have a
and can cause prolonged effect in patients with renal failure. very good safety record; it has been administered to mil-
Rocuronium is also primarily eliminated in the liver, with lions of patients worldwide without conclusive evidence of
10% to 25% renal excretion, but with no active metabolites. renal toxicity. Two volunteer studies have found biochemi-
Rocuronium at a bolus dose of 0.6 mg/kg also has a pro- cal evidence of renal injury during sevoflurane anesthesia,
longed duration of action in patients with renal failure.66 whereas five other volunteer studies have not.71–77 Nev-
Although many anesthesiologists will avoid these drugs in ertheless, KTx may represent a period of increased risk for
patients with ESRD, rocuronium and vecuronium can be renal injury, as defined by Artru.78 One study found that
safely used with appropriate clinical monitoring and under- exposure to sevoflurane or isoflurane had no significant
standing of their prolonged duration. Atracurium and effect on renal function parameters such as serum creati-
cisatracurium are metabolized primarily by spontaneous nine or creatinine clearance in patients with baseline renal
Hofmann degradation, an organ-independent metabolism. insufficiency (Cr >1.5 mg/dL).79–81 Additionally, a study
The parent agent, atracurium, is associated with histamine of 200 patients who underwent KTx reported no differ-
release, but cisatracurium is not.62 ence in postoperative creatinine, postoperative dialysis, and
Succinylcholine is a very short acting depolarizing neuro- the incidence of rejection between patients who received
muscular blocker and is the drug of choice in rapid sequence sevoflurane and those who received isoflurane during
intubations. Succinylcholine is known to cause an increase transplantation.82
in extracellular potassium. The increase in serum potas- Intraoperative management during KTx should primar-
sium after an intubating dose of succinylcholine was found ily focus on hemodynamic stability with preservation of
to be the same, approximately 0.6 mEq/L, for patients with adequate perfusion pressure to the graft. KTx surgery often
and without ESRD.67 This increase can be tolerated with- has prolonged periods of minimal stimulation. Blood loss
out significant risk of arrhythmia even by patients with an rarely exceeds 300 mL and large fluid shifts are not com-
initial serum K+ concentration greater than 5 mEq/L. The mon. Hypotension is frequently encountered during peri-
use of succinylcholine therefore is not absolutely contrain- ods of minimal surgical stimulation and may be aggravated
dicated in patients with ESRD.68 with unclamping of the iliac vessels and reperfusion of the
As previously mentioned, rapid sequence intubation may graft. Individualized fluid management is the cornerstone of
be required to reduce the risk of aspiration during induc- intraoperative hemodynamic management, whereas usage
tion in ESRD patients with uremic or diabetic gastropare- of vasoconstrictors with strong α-adrenergic effects, such as
sis, or with other typical indications such as acid reflux or phenylephrine, is discouraged.
full stomach. If there is a contradiction to succinylcholine, Maintenance of intravascular fluid status can be accom-
high-dose rocuronium (1.2 mg/kg) can provide rapid intu- plished using natural colloids (albumin), synthetic colloids
bating conditions. However, high-dose rocuronium in a (hydroxyethyl starches, dextrans, gelatins), and crystal-
patient with ESRD will likely have a significantly prolonged loids (normal saline, Ringer’s lactate, plasmalyte). Given
effect. the minimal blood loss and fluid shifts during KTx, crystal-
Sugammadex is a reversal agent for rocuronium, act- loids are sufficient for volume replacement and should be
ing via selective binding to rocuronium. The rocuronium- the preferred choice of fluid. Data for natural and synthetic
sugammadex complex is then eliminated by the kidneys. colloids in KTx are sparse. Therefore the routine use of these
Sugammadex is an effective reversal agent in patients with fluids in KTx cannot be recommended.
ESRD, however excretion of rocuronium and sugammadex The contents of the crystalloid solution administered
is significantly slower in patients with ESRD. This complex during KTx should be considered. In large amounts, nor-
will remain in the plasma for a prolonged period. The effects mal saline can cause a hyperchloremic metabolic acido-
of prolonged exposure to the rocuronium-sugammadex sis, whereas Ringer’s lactate and plasmalyte both contain
complex is not clear.69 Therefore neostigmine is the pre- potassium, which raises concern for hyperkalemia in the
ferred neuromuscular blocker reversal agent for patients patient with ESRD. A prospective, randomized, double-
with ESRD. It can be used for reversal of all of the nonde- blinded study compared normal saline to Ringer’s lactate
polarizing neuromuscular blockers. Of note, neostigmine is for intraoperative intravenous fluid therapy during KTx.
also partially eliminated in the kidneys, with an elimination The study was terminated for safety reasons and interim
half-life of 3 hours versus 1.3 hours in patients with normal analysis showed no difference in postoperative creatinine
renal function.70 levels. However, it demonstrated higher rates of severe
Overall, there is no evidence that the type of anesthetic hyperkalemia and metabolic acidosis in the normal saline
used during KTx is associated with patient and graft out- group.83 Most recently, a 2016 Cochrane Review meta-
comes. Most commonly, an inhaled volatile is used, either analysis investigated the effect of lower-chloride solutions
desflurane, isoflurane, or sevoflurane. All three of these vol- versus normal saline on delayed graft function, hyperkale-
atile anesthetics have been reported to be safe during KTx. mia and acid–base status in kidney transplant recipients.
The metabolism of sevoflurane has been implicated in renal The authors found no difference in the rates of delayed graft
toxicity, but there are no controlled studies identifying safety function or hyperkalemia, but showed that intraoperative
13 • Perioperative Care of Patients Undergoing Kidney Transplantation 191

balanced electrolyte solutions were associated with higher output. Some small trials have shown improved urine out-
blood pH, higher serum bicarbonate and lower serum put91 and creatinine clearance92 with low-dose dopamine
chloride.84 during KTx, whereas other larger studies have shown no
The debate continues over whether CVP monitoring is significant improvement in either parameter.93,94 The util-
required to guide fluid administration during KTx. When ity of this approach has been questioned on the basis that a
CVP monitoring is in place, most centers recommend main- newly transplanted, denervated kidney may not respond to
taining the CVP at 10 to 15 mm Hg. The effect of timing and low-dose dopamine normally. Doppler ultrasound exami-
duration of fluid replacement during KTx were examined in nation of newly transplanted kidneys found no significant
a prospective randomized trial.85 Patients were randomized change in blood flow with dopamine infusion rates of 1 to
to a continuous crystalloid infusion or a CVP-targeted crys- 5 μg/kg/min.95
talloid infusion with a low CVP target of 5 mm Hg through- Pain associated with surgery is moderate and is typi-
out most of the case and a CVP target of 15 mm Hg at the cally managed in the immediate postoperative phase with
end of renal vascular anastomoses (achieved by rapid infu- intravenous opioids, often using patient-controlled analge-
sion). Primary endpoints were markers of allograft function sia. Patients are transitioned to oral opioids quickly. When
within 5 postoperative days. Although both groups received continuous epidural is placed for intraoperative anesthesia,
equal total volumes of crystalloid, the CVP-targeted group it can provide excellent postoperative analgesia as well,
had better early allograft function. However, several con- but it is typically not necessary. As shown in Table 13.2,
founding factors in the study design warrant larger studies. the opioids morphine, meperidine, and oxycodone should
A small prospective study by Hadimioglu et al. demon- be used cautiously in patients with ESRD because they (or
strated a highly significant relationship between peripheral their active metabolites) are renally excreted and thus may
venous pressure (PVP) and CVP in patients undergoing accumulate in such patients. This risk of opioid accumula-
kidney transplantation, suggesting that PVP may be a rea- tion persists in the period after transplantation when the
sonable surrogate for CVP in patients without significant allograft may suffer from delayed graft function. In contrast,
cardiac dysfunction.86 the opioids fentanyl, sufentanil, alfentanil, and remifentanil
Pharmacologic blood pressure support using α-agonist have been shown to be safe alternatives, with fentanyl being
vasoconstrictors is usually discouraged based on some the most commonly used.96 Ketamine is a nonopioid anal-
limited experimental animal data. Taken together, stud- gesic with the active metabolite norketamine, which is also
ies indicate that there is a substantial attenuation of renal metabolized by the kidney and can cause prolonged effect.
hemodynamic responsiveness in transplanted kidney grafts The large dose of steroid (usually methylprednisolone)
with α-agonist administration. Furthermore, the studies given intraoperatively for induction of immunosuppression
suggest that the transplanted, denervated kidney loses its contributes an important analgesic effect as well. Intrathe-
capacity for autoregulation and that the renal response cal opioid and transversus abdominis plane (TAP) blocks
to sympathomimetics is altered with a shift toward time- have been studied in kidney transplant patients as part of
dependent flow reduction to the kidney. In one rat study, the enhanced recovery pathways, with improved satisfaction
response to sympathomimetics in the transplanted kidney and decreased hospital length of stay.97 Some institutions
was shifted toward renal blood flow reduction. The authors instead utilize TAP blocks as a rescue method for patients
postulated enhanced vasoconstriction via stimulation of with severe pain postoperatively.98
α-adrenoceptors and blunted vasodilatation via stimula- Intraoperative and postoperative hyperglycemia are
tion of β-adrenoceptors as a possible mechanism.87,88 How- common, especially, but not exclusively, in diabetics, due to
ever, the clinical significance of these findings in human the stress response to surgery and the large dose of cortico-
kidney transplant recipients remains unclear. A retrospec- steroids administered. A Cochrane Review showed limited
tive study compared 75 renal transplant recipients who evidence that more intensive insulin therapy had any effect
required phenylephrine intraoperatively to 75 matched on graft survival, all-cause mortality and adverse effects.99
controls who did not. The authors found no difference in A larger Cochrane Review of more than 1400 surgical
rates of delayed graft function or in serum creatinine 30, patients recommended caution with intensive perioperative
90, and 365 days after transplant.89 glycemic control compared conventional glycemic control.
Intraoperative urine production, a surrogate for allograft Although mortality was unchanged, a post hoc analysis
function, is frequently augmented with mannitol and loop showed more hypoglycemic episodes in the intensive glyce-
diuretics. Mannitol is freely filtered and not reabsorbed by mic control group.100
the nephron, causing osmotic expansion of urine volume. It
may also have a protective effect on the cells lining the renal
tubule. It is usually administered during the warm ischemia Postoperative Care
phase; thus mannitol may protect against ischemic injury,
as well as induce osmotic diuresis in the newly transplanted All renal transplant patients should be considered for post-
kidney. In most centers, relatively low doses of mannitol are operative extubation using standard criteria. Postopera-
administered, ranging between 0.25 and 0.5 mg/kg. Some tively, most kidney transplant recipients are admitted to the
data have shown that delayed graft function of the deceased postanesthesia care unit (PACU), with only a small percent-
donor renal allograft can be prevented by intraoperative age requiring admission to the intensive care unit (ICU).101
administration of mannitol.90 However, some centers do elect to admit all kidney trans-
Low-dose dopamine (2–3 μg/kg/min) is commonly used plant patients to the ICU.
to stimulate DA1 dopaminergic receptors in the kidney vas- In the PACU, patients are managed according to institu-
culature and thereby induce vasodilation and increase urine tional protocols with pain management guidelines tailored
192 Kidney Transplantation: Principles and Practice

TABLE 13.2 Clearance of Commonly Used Opioids and Effect of Renal Failure
SAFE TO USE IN PATIENTS WITH RENAL FAILURE
Fentanyl Metabolized by liver, no active metabolites
Sufentanil Fentanyl congener, same as above
Alfentanil Fentanyl congener, same as above
Methadone Metabolized by liver, no active metabolites
Remifentanil Hydrolyzed rapidly by plasma esterases, no active metabolites
CONSIDER CAUTION IN PATIENTS WITH RENAL FAILURE
Oxycodone Metabolized by liver, with multiple metabolites, oxymorphone is active metabolite which can
accumulate in renal failure. Large interindividual variation. About 10% excreted unchanged by
kidney.
Hydromorphone Metabolized in liver, hydromorphone-3-glucuronide is metabolite excreted in kidney. Hydromor-
phone-3-glucuronide has no analgesic effect but may have neuroexcitatory effect.
AVOID IN PATIENTS WITH RENAL FAILURE
Morphine Metabolized in liver to normorphine, morphine-3-glucuronide, and morphine-6-glucuronide (10%),
also 40% metabolized by kidney. Metabolites excreted by kidney. Morphine-6-glucuronide is
active metabolite, potent agonist at mu-receptor, can accumulate in renal failure.
Hydrocodone Metabolized to hydromorphone and hydromorphone-3-glucuronide, both can accumulate in renal
failure.
Codeine Multiple active metabolites including codeine-6-glucuronide, morphine, and morphine-6-glucuro-
nide, which can accumulate in renal failure.
Meperidine Primarily metabolized by liver, normeperidine is the active metabolite with a potent analgesic
effect and neuroexcitation causing seizures.
Normeperidine is renally excreted, accumulates in renal failure.

Modified based on Miller RD, Eriksson LI, Fleisher LA, et al. Miller’s Anesthesia. 7th ed. Philadelphia, PA: Churchill Livingstone/Elsevier; 2010:2113; Dean M. Opioids
in renal failure and dialysis patients. J Pain Symptom Manage 2004;28(5):497–504.

to ESRD patients, as previously discussed. Particular atten- have a stable GFR. In a study that followed patients 10 or
tion must be paid to graft function, which is primarily evalu- more years post-KTx, GFR decline over the first 12 months
ated by urine output over time. The majority of living donor after transplantation was an independent predictor of long-
kidney transplant recipients have immediate graft function. term allograft failure.104
In a study by Yadav et al., 84% of living donor kidney trans- Despite the significant improvement in overall mortality,
plant recipients experienced immediate graft function.102 post-KTx patients remain at high risk of cardiovascular dis-
Delayed graft function is more likely with deceased donor ease. Cardiac disease is the primary cause of death after KTx,
grafts, especially from hypertensive donors, older donors, with cardiac death 10 times more likely than in the general
or other extended criteria donors. Poor graft function may population. Patients are 50 times more likely to have a fatal
be attributable to the graft itself, the vessels, the ureter, or or nonfatal cardiovascular event annually than the general
clotting of the Foley catheter, all of which should be consid- population.70 One study found considerable progression of
ered in the differential diagnosis. The Foley catheter should coronary artery calcification in renal transplant patients at
be irrigated to ensure that clot or tissue has not affected least 1 year after surgery.105
its patency. Flow in the arterial and venous anastomoses Nevertheless, post-KTx patients have an overall lower
can be examined with ultrasound. Lastly, surgical reex- risk of cardiovascular morbidity compared with pretrans-
ploration should not be delayed if kinking of the vascular plant ESRD patients.106 Cho and colleagues documented
attachments or obstruction of the ureter along its course or that some patients with low ejection fractions secondary to
at the site of bladder reimplantation is suspected. In cases of uremic cardiomyopathy normalized their cardiac function
delayed graft function, patients may require dialysis postop- after successful KTx.107 Fig. 13.1 shows that patients who
eratively. Patients without preexisting hemodialysis access remain on the transplant list have a persistent 3% per year
may require urgent placement of a hemodialysis line. This incidence of MI, whereas patients who are transplanted
includes those patients who used peritoneal dialysis pre- have an initial rise in MI rate—probably due to periopera-
operatively, if their peritoneal dialysis catheter has been tive stress—followed by a slower increase in the rate of MI
removed during the transplant surgery. compared with patients who do not receive a transplant.108
The lower MI risk in living donor transplant recipients com-
pared with deceased donor graft recipients is also notable.
Anesthesia for Patients After The Kidney Disease: Improving Global Outcomes (KDIGO)
Kidney Transplantation clinical practice guideline recommends aspirin therapy for
all posttransplant patients with atherosclerosis.109
Patients with functioning grafts as determined by labora- Congestive heart failure, LV hypertrophy, and ischemic
tory values (blood urea nitrogen, creatinine) and with suf- heart disease remain important complications in KTx
ficient urine volume should be considered to have normal recipients. This is largely due to pretransplant risk factors
renal function. The average glomerular filtration rate (GFR) that persist posttransplantation, such as hypertension, dia-
6 months after deceased renal transplantation is almost 50 betes, dyslipidemia, and metabolic syndrome. In addition,
mL/min.103 About 50% of patients will manifest a slow graft rejection, viral infection, anemia,23,110 and immuno-
decline in GFR over the course of several years, but 30% will suppressive medications cause significant cardiovascular
13 • Perioperative Care of Patients Undergoing Kidney Transplantation 193

10% manifestations of DM. More recently, however, increasing


9% numbers of older diabetic patients are being considered for
Cumulative incidence of AMI

8% ist pancreas transplantation. In 2015, 25% of SPK candidates


gl
itin in the US were older than 50 years of age, up from 18% in
7% Wa 2004.111 These older patients with long-term DM and ESRD
6% therefore face elevated cardiovascular risk for surgery. After
do nor
5% ased successful SPK, cardiac pathology such as diastolic dys-
Dece plant
4% trans function and LV hypertrophy can improve or stabilize.113
3% It is not clear whether other manifestations of DM, such as
donor accelerated atherosclerosis, neuropathy, or vascular insuf-
2% Living nt
p la
trans ficiency, improve or stabilize after SPK.
1%
0%
0 12 24 36 PREOPERATIVE CONSIDERATIONS
Months post WL or TX In addition to the preoperative assessment process required
Fig. 13.1 Cumulative (Kaplan–Meier) incidence of acute myocardial for KTx alone, diabetic patients should be queried about
infarction (AMI) on the waiting list (WL) and after kidney transplan- blood glucose control and antiglycemic medications. Orally
tation (TX). Most transplant recipients also spent time on the waiting
list, but time was reset to “0” at transplantation. Most of the differ-
administered antiglycemic agents should not be taken on
ence in AMI incidence in recipients of deceased versus living donor the day of surgery to avoid unrecognized hypoglycemia
kidney transplants occurred very early after transplantation. There- under anesthesia. Blood glucose level should be checked
after, the incidence of AMI was similar in deceased and living donor preoperatively and corrected as appropriate before surgery.
transplant recipients, with both eventually having a lower incidence Insulin-dependent patients who experience large swings in
than patients on the waiting list. (From Kasiske BL, Maclean JR, ­Snyder JJ.
Acute myocardial infarction and kidney transplantation. J Am Soc Nephrol serum glucose levels are at risk for ketosis and intraopera-
2006;17:900–7.) tive acidemia.
Given their significant risk factors for cardiac disease,
an extensive cardiac workup is indicated in diabetic ESRD
morbidity. Corticosteroids and calcineurin inhibitors con- patients before surgery to rule out severe CAD. Preopera-
tribute to hypertension and hyperlipidemia and can cause tive evaluation by history and physical examination, ECG,
new-onset diabetes. Azathioprine and mycophenolate can treadmill testing, echocardiography with or without dobu-
cause anemia. tamine stress, radionuclide scintigraphy, and cardiac angi-
ography represent the full spectrum of workup that may be
applicable.
Kidney–Pancreas Transplantation
INTRAOPERATIVE CONSIDERATIONS
Far less common than KTx, simultaneous pancreas–kidney
transplantation (SPK) is offered to patients with poorly con- SPKs are long and surgically demanding operations involv-
trolled DM and diabetic nephropathy causing ESRD. Over ing extensive abdominal exposure. Therefore general endo-
80% of SPK transplants are performed in type 1 diabetics. In tracheal anesthesia with neuromuscular blockade is the
2015, 947 pancreas transplants were performed in the US. best anesthetic approach for these cases. As mentioned pre-
Approximately 80% received a SPK, and the others received viously, patients with DM and uremia often have gastropa-
either pancreas transplantation alone or pancreas transplan- resis and large residual gastric volumes. These patients may
tation after kidney transplantation. However, pancreas trans- benefit from a nonparticulate antacid and a rapid sequence
plantation overall has decreased more than 30% since 2006, induction.
matching a decrease in demand and likely reflecting both an Patients may have more significant postoperative pain
improvement in medical management of DM as well as the given the larger incision and more extensive abdominal
complications associated with transplantation.111 Interest- dissection compared with KTx alone, so placement of an
ingly, despite the decline in pancreas transplantation in the epidural catheter for postoperative pain control may be
US, the number of pancreas transplants has been increasing warranted. However, epidural local anesthetic may cause
internationally, particularly in Europe and Asia.112 sympathetic blockade, which in theory could compromise
Patients with ESRD superimposed on DM are subject to graft perfusion. Therefore some centers prefer to defer epi-
the same hemodynamic, volume, and electrolyte problems dural placement.
as those with ESRD alone. Most are maintained on some Standard monitors (five-lead ECG, NIBP measurement,
form of dialysis to manage fluid overload and accumulation pulse oximetry, end-tidal gas concentrations, and tempera-
of electrolytes. These patients may also have the same prob- ture) are required as with any general anesthetic. Patients
lems of chronic anemia and uremic coagulopathy as those require frequent intraoperative and postoperative assess-
with renal failure alone. ment of serum glucose and electrolytes and an arterial line or
In addition to the hypertension often seen in ESRD, central venous catheter can facilitate frequent blood draws.
patients with diabetes often have accelerated atherosclero- Select patients may warrant both an arterial line and central
sis and autonomic nervous system dysfunction. venous catheter depending on their underlying comorbidities.
Historically, candidates for pancreas transplantation Patients with autonomic neuropathy are often consid-
have been younger than those listed for kidney trans- ered to be at greater risk for severe cardiovascular depres-
plantation, and therefore tended to have fewer long-term sion during the induction of anesthesia. However, no
194 Kidney Transplantation: Principles and Practice

studies have shown a difference in hemodynamic response POSTOPERATIVE CARE


to induction between diabetic patients with preexisting
autonomic neuropathy and nondiabetic uremic patients Successful pancreas transplantation usually results in rap-
undergoing transplantation. Hemodynamic stability over idly declining insulin requirements. Blood glucose levels
long periods may be best achieved using a balanced anes- should be monitored closely in the recovery room or the
thetic technique that involves both inhaled and intrave- ICU to avoid hypoglycemia. With SPK as with KTx alone,
nous anesthetic agents as well as intravenous opioids. urine output should also be closely monitored to detect
Blood pressure and fluid management can be challenging reversible kidney graft injury. Postoperative pain con-
during SPK. As with KTx, perfusion pressure is important trol can be managed with epidural or patient-controlled
for a newly anastomosed pancreas. A lengthy intraabdomi- analgesia.
nal procedure may necessitate significant amounts of fluid. Patient and graft survival in SPK have increased signifi-
However, patients with hypertension and diastolic dysfunc- cantly over the past several decades.118 During the period
tion may be at risk for heart failure from volume overload. of 2010 to 2014, SPK patients in the US had a 1-year sur-
Therefore fluid administration should be guided by an vival of 97.4%, pancreas graft survival was 91.3% and
assessment of volume status, as determined by heart rate, kidney graft survival was 95.5%.112 SPK does not appear
blood pressure, plethysmography variability index and to decrease the success of the kidney graft compared with
pulse pressure or systolic pressure variation, for example. KTx alone in diabetic patients. Like patients post KTx, car-
As opposed to in KTx alone, colloid administration is some- diovascular disease remains the primary cause of death
times advocated in SPK instead of large volumes of crys- post SPK.112
talloid due to theoretical concern for pancreatic swelling,
although no controlled studies have been conducted. Blood
products should be administered per standard criteria. References
Abdominal muscle relaxation is essential for SPKs with
1. Vandam LD, Harrison JH, Murray JE, et al. Anesthetic aspects of
their extensive intraabdominal dissection. As with KTx renal homotransplantation in man. With notes on the anesthetic
alone, attention to the metabolism of neuromuscular block- care of the uremic patient. Anesthesiology 1962;23:783–92.
ers is required. Cisatracurium remains a good choice as it 2. Ko CW, Chang CS, Wu MJ, et al. Gastric dysrhythmia in uremic
does not rely on renal metabolism. A continuous infusion patients on maintenance hemodialysis. Scand J Gastroenterol
1998;33(10):1047–51.
of cisatracurium can be used given the long case duration, 3. Global Observatory on Donation and Transplantation. http://www.
allowing for titration of the depth of the neuromuscular transplant-observatory.org. Accessed October 1, 2017.
block with reliable reversibility. Intermittent administra- 4. United Network of Organ Sharing, Transplant Trends. https://www.
tion of vecuronium, if titrated by train-of-four monitor- unos.org/data/transplant-trends/#transplants_by_organ_type+y
ing, can also provide excellent relaxation conditions. For ear+ 2016. Accessed October 1, 2017.
5. OPTN/SRTR 2015 Annual Data Report: Introduction. Am J Trans-
patients who are undergoing pancreas transplant alone plant 2017;17(1):11–20.
or pancreas transplant after KTx and have adequate renal 6. Liefeldt L, Budde K. Risk factors for cardiovascular disease in renal
function, any intermediate-acting nondepolarizing muscle transplant recipients and strategies to minimize risk. Transplant Int
relaxant may be used. 2010;23(12):1191–204.
7. Weir MR, Burgess ED, Cooper JE, et al. Assessment and manage-
Intraoperative blood glucose control is important to ment of hypertension in transplant recipients. J Am Soc Nephrol
prevent ketoacidosis in patients with unopposed counter- 2015;26:1248–60.
regulatory hormone secretion and to assess the function 8. Brown JJ, Dusredieck G, Fraser R, et al. Hypertension and chronic
of the transplanted pancreas. Before the pancreas is reper- renal failure. Br Med Bull 1971;27(2):128–35.
fused, glucose should be checked hourly and treated as 9. Prasad GVR, Ruzicka M, Burns KD, et al. Hypertension in dialysis
and kidney transplant patients. Can J Cardiol 2009;25(5):309–14.
appropriate. Hyperglycemia may cause depressed immune 10. Gavras H, Oliver JA,PJ. Cannon, Interrelations of renin, angioten-
function and impaired wound healing and puts patients at sin II, and sodium in hypertension and renal failure. Annu Rev Med
risk for more severe neurologic injury should brain isch- 1976;27:485–521.
emia occur.114 After the pancreas is reperfused, glucose 11. Craig RG, Hunter JM. Recent developments in the perioperative
management of patients with chronic kidney disease. Br J Anaesth
should be checked every 30 minutes to monitor for both 2008;101(3):296–310.
hyperglycemia, which may harm pancreatic graft beta 12. Converse Jr RL, Jacobsen RN, Toto RD, et al. Sympathetic over-
cells, and hypoglycemia. Typically, glucose concentrations activity in patients with chronic renal failure. N Engl J Med
decrease by approximately 50 mg/dL/h after the pancreas 1992;327(27):1912–8.
is unclamped.115 13. Toto RD. Treatment of hypertension in chronic kidney disease.
Semin Nephrol 2005;25(6):435–9.
A randomized trial of intraoperative glucose management 14. Sear JW, Jewkes C, Tellez JC, et al. Does the choice of antihyperten-
in insulin-dependent type 2 diabetics compared a continu- sive therapy influence haemodynamic responses to induction, laryn-
ous glucose-insulin infusion with intermittent intravenous goscopy and intubation? Br J Anaesth 1994;73(3):303–8.
insulin injections during both major and minor surgery. No 15. Comfere T, Sprung J, Kumar MM, et al. Angiotensin inhibitors in a
general surgery population. Anesth Analg 2005;100:636–44.
difference was found in the ability to control intraoperative 16. Kheterpal S, Khodaparast O, Shanks A, et al. Chronic angiotensin-
and postoperative glucose levels and metabolism.116 Simi- converting enzyme inhibitor or angiotensin receptor blocker therapy
larly, a study of intraoperative glucose management dur- combined with diuretic therapy is associated with increased episodes
ing pancreas transplantation and SPK found no significant of hypotension in noncardiac surgery. J Cardiothorac Vasc Anesth
difference in glycemic control when comparing continu- 2008;22(2):180–6.
17. Smith I, Jackson I. Beta-blockers, calcium channel blockers, angio-
ous insulin infusion with intermittent intravenous insulin tensin converting enzyme inhibitors and angiotensin receptor block-
boluses.117 Therefore the manner of lowering glucose does ers: should they be stopped or not before ambulatory anaesthesia?
not appear to be as critical as the glucose level itself. Curr Opin Anesthesiol 2010;23:687–90.
13 • Perioperative Care of Patients Undergoing Kidney Transplantation 195

18. Florens N, Calzada C, Lyasko E, et al. Modified lipids and lipopro- 44. Grayburn PA, Hillis LD. Cardiac events in patients undergoing non-
teins in chronic kidney disease: a new class of uremic toxins. Toxins cardiac surgery: shifting the paradigm from noninvasive risk stratifi-
2016;8(12):376–402. cation to therapy. Ann Intern Med 2003;138(6):506–11.
19. Goldberg IJ. Lipoprotein metabolism in normal and uremic patients. 45. Schmidt A, Stefenelli T, Schuster E, et al. Informational contribu-
Am J Kidney Dis 1993;21(1):87–90. tion of noninvasive screening tests for coronary artery disease in
20. Wolfe RA, Ashby VB, Milford EL, et al. Comparison of mortality patients on chronic renal replacement therapy. Am J Kidney Dis
in all patients on dialysis, patients on dialysis awaiting transplan- 2001;37(1):56–63.
tation, and recipients of a first cadaveric transplant. N Engl J Med 46. Lee TH, Marcantonio ER, Mangione CM, et al. Derivation and
1999;341(23):1725–30. prospective validation of a simple index for prediction of cardiac
21. Grundy SM. Hypertriglyceridemia, insulin resistance, and the meta- risk of major noncardiac surgery. Circulation 1999;100(10):
bolic syndrome. Am J Cardiol 1999;83(9B):25F-9F. 1043–9.
22. Lentine KL, Hurst FP, Jindal RM, et al. Cardiovascular risk assess- 47. Hoftman N, Prunean A, Dhillon A, et al. Revised Cardiac Risk
ment among potential kidney transplant candidates: approaches Index (RCRI) is a useful tool for evaluation of perioperative car-
and controversies. Am J Kidney Dis 2010;55(1):152–67. diac morbidity in kidney transplant recipients. Transplantation
23. Rigatto C. Clinical epidemiology of cardiac disease in renal trans- 2013;96(7):639–43.
plant recipients. Semin Dial 2003;16(2):106–10. 48. Lentine KL, Costa SP, Weir MR, et al. Cardiac disease evaluation and
24. Gallon LG, Leventhal JR, Kaufman DB. Pretransplant evaluation of management among kidney and liver transplantation candidates:
renal transplant candidates. Semin Nephrol 2002;22(6):515–25. a scientific statement from the american heart association and
25. Humar A, Kerr SR, Ramcharan T, et al. Peri-operative cardiac mor- the american college of cardiology foundation. JACC 2012;60(5):
bidity in kidney transplant recipients: incidence and risk factors. Clin 434–80.
Transplant 2001;15(3):154–8. 49. Boden WE, O’Rourke RA, Teo KK, et al. Optimal medical therapy
26. Gill JS, Pereira BJ. Death in the first year after kidney transplanta- with or without PCI for stable coronary disease. N Engl J Med
tion: implications for patients on the transplant waiting list. Trans- 2007;356(15):1503–16.
plantation 2003;75(1):113–7. 50. McFalls EO, Ward HB, Moritz TE, et al. Coronary-artery revascu-
27. Foley RN, Parfrey PS, Harnett JD, et al. Clinical and echocardio- larization before elective major vascular surgery. N Engl J Med
graphic disease in patients starting end-stage renal disease therapy. 2004;351(27):2795–804.
Kidney Int 1995;47(1):186–92. 51. Sedlis SP, Jurkovitz CT, Hartigan PM, et al. Optimal medical therapy
28. Ansari A, Kaupke CJ, Vaziri ND, et al. Cardiac pathology in patients with or without percutaneous coronary intervention for patients
with end-stage renal disease maintained on hemodialysis. Int J Artif with stable coronary artery disease and chronic kidney disease. Am J
Organs 1993;16(1):31–6. Cardiol 2009;104(12):1647–53.
29. Burke SW, Solomon AJ. Cardiac complications of end-stage renal. 52. Dunkelgrun M, Boersma E, Schouten O, et al. Bisoprolol and fluv-
Adv Ren Replace Ther 2000;7(3):210–9. astatin for the reduction of perioperative cardiac mortality and
30. Eschbach JW, Kelly MR, Haley NR, et al. Treatment of the anemia of myocardial infarction in intermediate-risk patients undergoing non-
progressive renal failure with recombinant human erythropoietin. N cardiovascular surgery: a randomized controlled trial (DECREASE-
Engl J Med 1989;321(3):158–63. IV). Ann Surg 2009;249:921–6.
31. Marsh JT, Brown WS, Wolcott D, et al. rHuEPO treatment improves 53. Mittel AM, Wagener G. Anesthesia for kidney and pancreas trans-
brain and cognitive function of anemic dialysis patients. Kidney Int plantation. Anesthesiol Clin 2017;35(3):439–52.
1991;39(1):155–63. 54. Peterson P, Hayes TE, Arkin CF, et al. The preoperative bleeding
32. Lietz K, Lao M, et al. The impact of pretransplant erythropoietin time test lacks clinical benefit: College of American Pathologists’ and
therapy on late outcomes of renal transplantation. Ann Transplant American Society of Clinical Pathologists’ position article. Arch Surg
2003;8(2):17–24. 1998;133(2):134–9.
33. Pivalizza EG, Abramson DC, Harvey A. Perioperative hyperco- 55. Eng M, Brock G, Li X, et al. Perioperative anticoagulation and anti-
agulability in uremic patients: a viscoelastic study. J Clin Anesth platelet therapy in renal transplant: is there an increase in bleeding
1997;9(6):442–5. complication? Clin Transplant 2011;25(2):292–6.
34. Ando M, Iwata A, Ozeki Y, et al. Circulating platelet-derived mic- 56. Fahy BG, Gouzd VA, Atallah JN. Pregnancy tests with end-stage
roparticles with procoagulant activity may be a potential cause of renal disease. J Clin Anesth 2008;20(8):609–13.
thrombosis in uremic patients. Kidney Int 2002;62(5):1757–63. 57. Committee on Quality Management and Departmental Administra-
35. Schoonjans R, Van B, Vandamme W, et al. Dyspepsia and gastro- tion. Pregnancy Testing Prior to Anesthesia and Surgery Committee
paresis in chronic renal failure: the role of Helicobacter pylori. Clin of Origin: Quality Management and Departmental Administration.
Nephrol 2002;57(3):201–7. American Society of Anesthesiology; 2016. Available online at:
36. Strid H, Simren M, Stotzer PO, et al. Delay in gastric empty- http://www.asahq.org/quality-and-practice-management/prac-
ing in patients with chronic renal failure. Scand J Gastroenterol tice-guidance- resource-documents/pregnancy-testing-prior-to-
2004;39(6):516–20. anesthesia-and-surgery.
37. Goldfarb-Rumyantzev A, Hurdle JF, Scandling J, et al. Duration of 58. Reissell E, Taskinen MR, Orko R, et al. Increased volume of gastric
end-stage renal disease and kidney transplant outcome. Nephrol contents in diabetic patients undergoing renal transplantation:
Dial Transplant 2005;20(1):167–75. lack of effect with cisapride. Acta Anaesthesiol Scand 1992;36(7):
38. Lentine KL, Costa SP, Weir MR, et al. Cardiac disease evaluation and 736–40.
management among kidney and liver transplantation candidates. 59. Orko Heino AR, Rosenberg PH. Anaesthesiological complications in
JACC 2012;60(5):434–80. renal transplantation: a retrospective study of 500 transplantations.
39. Herzog CA, Marwick TH, Pheley AM, et al. Dobutamine stress echo- Acta Anaesthesiol Scand 1986;30(7):574–80.
cardiography for the detection of significant coronary artery disease in 60. Sear JW. Kidney transplants: induction and analgesic agents. Int
renal transplant candidates. Am J Kidney Dis 1999;33(6):1080–90. Anesthesiol Clin 1995;33(2):45–68.
40. Sharma R, Pellerin D, Gaze DC, et al. Dobutamine stress echocar- 61. Sandid MS, Assi MA, Hall S. Intraoperative hypotension and pro-
diography and cardiac troponin T for the detection of significant longed operative time as risk factors for slow graft function in kidney
coronary artery disease and predicting outcome in renal transplant transplant recipients. Clin Transplant 2006;20:762–8.
candidates. Eur J Echocardiogr 2005;6(5):327–35. 62. Miller RD, Eriksson LI, Fleisher LA, et al. Philadelphia, PA: Churchill
41. Wang LW, Fahirn MA, Hayen A, et al. Cardiac testing for coronary Livingstone/Elsevier. 7th ed. Miller’s Anesthesia, ;831. 2010. p.
artery disease in potential kidney transplant recipients. Cochrane 2112–6. 880.
Database Syst Rev 2011;12:CD008691. 63. Feng CK, Chan KH, Liu KN, et al. A comparison of lidocaine, fen-
42. Wong CF, Little MA, Vinjamuri S, et al. Technetium myocardial tanyl, and esmolol for attenuation of cardiovascular response
perfusion scanning in prerenal transplant evaluation in the United to laryngoscopy and tracheal intubation. Acta Anaesthesiol Sin
Kingdom. Transplant Proc 2008;40:1324–8. 1996;34(2):61–7.
43. Rabbat CG, Treleaven DJ, Russell JD, et al. Prognostic value of myo- 64. Goyal P, Puri GD, Pandey CK, et al. Evaluation of induction doses
cardial perfusion studies in patients with end-stage renal disease of propofol: comparison between end-stage renal disease and nor-
assessed for kidney or kidney-pancreas transplantation: a meta- mal renal function patients. Anaesth Intensive Care 2002;30(5):
analysis. J Am Soc Nephrol 2003;14:431–9. 584–7.
196 Kidney Transplantation: Principles and Practice

65. Kirvela M, Olkkola KT, Rosenberg PH, et al. Pharmacokinetics of study of renal function and graft survival after preservation with
propofol and haemodynamic changes during induction of anaesthe- University of Wisconsin solution in multi-organ donors. European
sia in uraemic patients. Br J Anaesth 1992;68(2):178–82. Multicenter Study Group. Transplantation 1997;63(11):1620–8.
66. Robertson EN, Driessen JJ, Booij LH. Pharmacokinetics and pharma- 91. Grundmann R, Kindler J, Meider G, et al. Dopamine treatment of
codynamics of rocuronium in patients with and without renal fail- human cadaver kidney graft recipients: a prospectively randomized
ure. Eur J Anaesthesiol 2005;22(1):4–10. trial. Klin Wochenschr 1982;60(4):193–7.
67. Powell DR, Miller R. The effect of repeated doses of succinylcholine 92. Carmellini M, Romagnoli J, Giulianotti PC, et al. Dopamine lowers the
on serum potassium in patients with renal failure. Anesth Analg incidence of delayed graft function in transplanted kidney patients
1975;54(6):746–8. treated with cyclosporine A. Transplant Proc 1994;26(5):2626–9.
68. Thapa S, Brull SJ. Succinylcholine-induced hyperkalemia in 93. Kadieva VS, Friedman L, Margolius LP, et al. The effect of dopamine
patients with renal failure: an old question revisited. Anesth Analg on graft function in patients undergoing renal transplantation.
2000;91(1):237–41. Anesth Analg 1993;76(2):362–5.
69. Staals LM, Snoeck MM, Driessen JJ, et al. Reduced clearance of 94. Sandberg J, Tyden G, Groth CG. Low-dose dopamine infusion fol-
rocuronium and sugammadex in patients with severe to end-stage renal lowing cadaveric renal transplantation: no effect on the incidence of
failure: a pharmacokinetic study. Br J Anaesth 2010;104(1):31–9. ATN. Transplant Proc 1992;24(1):357.
70. Neale J, Smith AC. Cardiovascular risk factors following renal trans- 95. Spicer ST, Gruenewald S, O’Connell PJ, et al. Low-dose dopamine
plant. World J Transplant 2015;5(4):183–95. after kidney transplantation: assessment by doppler ultrasound. Clin
71. Ebert TJ, Frink Jr EJ, Kharasch ED. Absence of biochemical evidence Transplant 1999;13(6):479–83.
for renal and hepatic dysfunction after 8 hours of 1.25 minimum 96. Dean M. Opioids in renal failure and dialysis patients. J Pain Symp-
alveolar concentration sevoflurane anesthesia in volunteers. Anes- tom Manage 2004;28(5):497–504.
thesiology 1998;88(3):601–10. 97. Halawa A, Rowe S, Roberts F, et al. A better journey for patients,
72. Ebert TJ, Messana LD, Uhrich TD, et al. Absence of renal and hepatic a better deal for the NHS: the successful implementation of an
toxicity after four hours of 1.25 minimum alveolar anesthetic con- enhanced recovery program after renal transplant surgery. Exp Clin
centration sevoflurane anesthesia in volunteers. Anesth Analg Transplant 2018;16(2):127–32. Available online at: https://doi.
1998;86(3):662–7. org/10.6002/ect.2016.0304.
73. Eger 2nd EI, Gong D, Koblin DD, et al. Dose-related biochemical 98. Forbes RC, Concepcion BP, King AB. Intraoperative management of
markers of renal injury after sevoflurane versus desflurane anesthe- the kidney transplant recipient. Curr Transpl Rep 2017;4:75–81.
sia in volunteers. Anesth Analg 1997;85(5):1154–63. 99. Lo C, Jun M, Badve SV, et al. Glucose-lowering agents for treating
74. Eger 2nd EI, Koblin DD, Bowland T, et al. Nephrotoxicity of sevo- pre-existing and new-onset diabetes in kidney transplant recipients.
flurane versus desflurane anesthesia in volunteers. Anesth Analg Cochrane Database Syst Rev 2017;27(2):1–54.
1997;84(1):160–8. 100. Buchleitner AM, Martínez-Alonso M, Hernández M, et al. Periop-
75. Frink Jr EJ, Malan P, Morgan S, et al. Renal concentrating function erative glycaemic control for diabetic patients undergoing surgery.
with prolonged sevoflurane or enflurane anesthesia in volunteers. Cochrane Database Syst Rev 2012;12(9):1–96.
Anesthesiology 1994;80(5):1019–25. 101. Kogan A, Singer P, Cohen J, et al. Readmission to an intensive care
76. Frink Jr EJ, Malan TP, Morgan SE, et al. Sevoflurane degradation unit following liver and kidney transplantation: a 50-month study.
product concentrations with soda lime during prolonged anesthesia. Transplant Proc 1999;31(4):1892–3.
J Clin Anesth 1994;6(3):239–42. 102. Yadav B, Prasad N, Agrawal V, et al. Urinary kidney injury mol-
77. Munday IT, Stoddart PA, Jones RM, et al. Serum fluoride concentra- ecule-1 can predict delayed graft function in living donor renal
tion and urine osmolality after enflurane and sevoflurane anesthesia allograft recipients. Nephrology 2015;20(11):801–6.
in male volunteers. Anesth Analg 1995;81(2):353–9. 103. Gill JS, Tonelli M, Mix CH, et al. The change in allograft function
78. Artru AA. Renal effects of sevoflurane during conditions of possible among long-term kidney transplant recipients. J Am Soc Nephrol
increased risk. J Clin Anesth 1998;10(7):531–8. 2003;14(6):1636–42.
79. Conzen PF, Kharash ED, Czerner SF, et al. Low-flow sevoflurane 104. Park JS, Oh IH, Lee CH, et al. The rate of decline of glomerular filtra-
compared with low-flow isoflurane anesthesia in patients with stable tion rate is a predictor of long-term graft outcome after kidney trans-
renal insufficiency. Anesthesiology 2002;97(3):578–84. plantation. Transplant Proc 2013;45(4):1438–41.
80. Conzen PF, Nuscheler M, Melotte A, et al. Renal function and serum 105. Schankel K, Robinson J, Bloom R, et al. Determinants of coronary
fluoride concentrations in patients with stable renal insufficiency artery calcification progression in renal transplant recipients. Am J
after anesthesia with sevoflurane or enflurane. Anesth Analg Transplant 2007;7(9):2158–64.
1995;81(3):569–75. 106. Bittar J, Arenas P, Chiurchiu C, et al. Renal transplantation in
81. Nishimori A, Tanaka K, Ueno K, et al. Effects of sevoflurane anaes- high cardiovascular risk patients. Transplant Rev (Orlando)
thesia on renal function. J Int Med Res 1997;25(2):87–91. 2009;23(4):224–34.
82. Teixeira S, Costa G, Costa F J, et al. Sevoflurane versus isoflurane: does 107. Cho WH, Kim HT, Park CH, et al. Renal transplantation in advanced
it matter in renal transplantation? Transplant Proc 2007;39:2486– cardiac failure patients. Transplant Proc 1997;29(1-2):236–8.
8. 108. Kasiske BL, Maclean JR, Snyder JJ. Acute myocardial infarction and
83. O’Malley CM, Frumento RJ, Hardy MA, et al. A randomized, double- kidney transplantation. J Am Soc Nephrol 2006;17(3):900–7.
blind comparison of lactated Ringer’s solution and 0.9% NaCl during 109. Kasiske BL, Zeier MG, Craig JC, et al. Special issue: KDIGO clinical
renal transplantation. Anesth Analg 2005;100(5):1518–24. practice guideline for the care of kidney transplant recipients. Kid-
84. Wan S, Roberts MA, Mount P. Normal saline versus lower-chloride ney Disease: Improving Global Outcomes (KDIGO) transplant work
solutions for kidney transplantation. Cochrane Database Syst Rev group. Am J Transplant 2009;9(Suppl 3):1–155.
2016;8:1–27. 110. Ponticelli C, Villa M. Role of anaemia in cardiovascular mortal-
85. Othman MM, Ismael AZ, Hammouda GE. The impact of timing of ity and morbidity in transplant patients. Nephrol Dial Transplant
maximal crystalloid hydration on early graft function during kidney 2002;17(Suppl 1):41–6.
transplantation. Anesth Analg 2010;110(5):1440–6. 111. Kandaswamy R, Stock PG, Gustafson SK, et al. OPTN/SRTR 2015
86. Hadimioglu N, Ertug Z, Yegin A, et al. Correlation of peripheral annual data report: pancreas. Am J Transplant 2017;17(Suppl
venous pressure and central venous pressure in kidney recipients. 1):117–73.
Transplant Proc 2006;38(2):440–2. 112. Gruessner AC, Gruessner RW. Pancreas transplantation of US and
87. Gabriels G, August C, Grisk O, et al. Impact of renal transplantation non-US cases from 2005 to 2014 as reported to the United Network
on small vessel reactivity. Transplantation 2003;75(5):689–97. for Organ Sharing (UNOS) and the International Pancreas Trans-
88. Morita K, Seki T, Nonomura K, et al. Changes in renal blood flow plant Registry (IPTR). Rev Diabet Stud 2016;13(1):35–58.
in response to sympathomimetics in the rat transplanted and dener- 113. Oppert M, Schneider U, Boksch W, et al. Improvement of left ven-
vated kidney. Int J Urol 1999;6(1):24–32. tricular function and arterial blood pressure 1 year after simul-
89. Day KM, Beckman RM, Machan JT, et al. Efficacy and safety of phen- taneous pancreas kidney transplantation. Transplant Proc
ylephrine in the management of low systolic blood pressure after 2002;34(6):2251–2.
renal transplantation. J Am Coll Surg 2014;218(6):1207–13. 114. Sieber FE. The neurologic implications of diabetic hyperglycemia
90. Koning OH, Ploeg RJ, van Bockel JH, et al. Risk factors for delayed during surgical procedures at increased risk for brain ischemia. J Clin
graft function in cadaveric kidney transplantation: a prospective Anesth 1997;9(4):334–40.
13 • Perioperative Care of Patients Undergoing Kidney Transplantation 197

115. Spiro MD, Eilers H. Intraoperative care of the transplant patient. 117. Halpern H, Miyoshi E, Kataoka LM, et al. Glycemic control during
Anesth Clin 2013;31:705–21. pancreas transplantation: continuous infusion versus bolus. Trans-
116. Hemmerling TM, Schmid MC, Schmidt J, et al. Comparison of a plant Proc 2004;36(4):984–5.
continuous glucose-insulin-potassium infusion versus intermittent 118. Gruessner AC. 2011 update on pancreas transplantation: compre-
bolus application of insulin on perioperative glucose control and hensive trend analysis of 25,000 cases followed up over the course of
hormone status in insulin-treated type 2 diabetics. J Clin Anesth twenty-four years at the International Pancreas Transplant Registry
2001;13(4):293–300. (IPTR). Rev Diabet Stud 2011;8(1):6–16.
14 Early Course of the Patient
With a Kidney Transplant
PAUL M. SCHRODER, LORNA P. MARSON, and STUART J. KNECHTLE

CHAPTER OUTLINE Overview Hyperacute Rejection


Perioperative Management Antibody-Mediated Rejection
Graft Dysfunction Acute Rejection
Surgical Complications Borderline Rejection
Urinary Problems Medical Complications
Urinary Obstruction Delayed Graft Function
Bleeding Into the Urinary System Nephrotoxicity From Calcineurin Inhibitors
Urine Leak Prerenal Azotemia and Volume Depletion
Vascular Problems Other Drug Toxicity
Arterial Stenosis Recurrent Disease
Arterial Thrombosis Infection
Renal Vein Thrombosis Hypertension
Postoperative Bleeding Management of Graft Dysfunction
Graft Loss and Transplant Nephrectomy Summary
Rejection During the Early Postoperative
Period

A successful long-term outcome for a new kidney trans- to transplantation exist. For instance, the presence of sig-
plant recipient depends on the early perioperative manage- nificant cardiac disease may preclude successful surgery.
ment and course after surgery. Important factors affecting Characteristics such as tobacco use, diabetes, obesity,
long-term outcome include the occurrence of delayed graft hypertension, and dyslipidemia have all been shown to be
function (DGF); episodes of acute rejection; early surgical independent predictors of cardiovascular disease in kidney
complications, such as urinary obstruction, urine leak, or transplant recipients and should prompt further cardiac
vascular complications; and sepsis. Toxicity from calcineu- evaluation, particularly in the symptomatic preoperative
rin inhibitors (CNIs) can lead to chronic transplant damage candidate.1–3 The Revised Cardiac Risk Index has also been
later in the posttransplantation course. Donor and recipi- demonstrated to be a useful perioperative tool for evaluat-
ent factors affect long-term outcome, particularly the use ing adverse cardiac event risk in kidney transplant recipi-
of high kidney donor profile index (KDPI) donors or highly ents, particularly for those older than age 50.4 In addition,
sensitized recipients. The early recognition and manage- peripheral vascular disease and vascular insufficiency are
ment of risk factors in the immediate postoperative period more common in end-stage renal disease (ESRD) patients
may lessen their long-term negative effect and improve and represent a barrier to successful transplantation,
outcome. with a higher incidence of postoperative renal transplant
artery stenosis, graft failure, and mortality. Thus a simple
pulse examination before surgery with ankle brachial indi-
ces in select patients can help stratify perioperative risks
Overview of vascular morbidity in kidney transplant recipients.5
Assessment of the recipient’s pretransplant fluid status
PERIOPERATIVE MANAGEMENT
and electrolyte levels to determine the need for dialysis is
A patient’s journey to a successful kidney transplant also important in the perioperative period. However, rou-
begins long before the patient meets the surgical and tine hemodialysis immediately before transplantation is
anesthesiology teams, at the time of diagnosis of chronic not warranted except in cases of metabolic derangements
kidney disease where the patient and his or her nephrolo- (e.g., hyperkalemia) or fluid overload because preopera-
gist discuss and initiate the process of waitlist candidacy. tive hemodialysis has been associated with an increased
Surgical management of the kidney transplant recipient risk of delayed graft function.6 Knowledge of the donor
begins in the immediate preoperative period. The initial status is also helpful in the early postoperative manage-
evaluation includes a careful history and physical exami- ment of the transplant recipient. With an ideal deceased
nation to determine whether potential contraindications donor or a living related donor, the expected outcome is
198
14 • Early Course of the Patient With a Kidney Transplant 199

an immediately functioning transplant that may preclude TABLE 14.1 Early Surgical and Medical Complications
posttransplant dialysis. Kidneys procured from high KDPI After Transplantation
donors or donation after circulatory death (DCD) donors
have a higher likelihood of DGF, which can lead to volume Surgical/Mechanical Medical
overload and the need for urgent dialysis.7 Technical con- Ureteral obstruction Acute rejection
siderations include the need for vascular reconstruction, Hematuria Delayed graft function
which may prolong surgery and contribute to postopera- Urine leak Acute calcineurin inhibitor
Arterial thrombosis/stenosis nephrotoxicity
tive DGF. Recipient factors also affect the early postopera- Renal vein thrombosis Prerenal/volume contraction
tive course. Significant risk factors for early posttransplant Postoperative hemorrhage Drug toxicity
dysfunction include pretransplant sensitization, obesity, Lymphocele Infection
younger or older age, and anatomic considerations that Recurrent disease
complicate the surgery.
In the early perioperative period, attention to fluid and
electrolyte balance is crucial. Careful monitoring of urine (Table 14.1). The most common early posttransplant medi-
output is essential, and any decrease in urine flow must be cal problem is DGF, which occurs in 20% of patients who
evaluated. The Kidney Disease: Improving Global Outcomes received kidneys from ideal deceased donors and in nearly
(KDIGO) guidelines recommend measuring urine volume 40% of patients in whom the donors were older than age
every 1 to 2 hours for at least 24 hours after transplanta- 55 years.13 After or concomitant with DGF, acute rejection
tion and daily until graft function is stable.8 In addition, may become a significant clinical problem. Other reasons
serum creatinine should be measured daily until hospital for early medical complications include acute cyclospo-
discharge, and frequently thereafter. For example, creati- rine or tacrolimus nephrotoxicity, prerenal azotemia,
nine should be measured two to three times per week for a other drug toxicity, infection, and early recurrent disease.
month, and a tapering frequency of measurements in ensu- An uncommon but serious posttransplantation medical
ing weeks.8 A decrease in urine volume may be a result of problem is thrombotic microangiopathy (discussed later).
acute tubular necrosis, hypovolemia, urinary leak, ureteric Thrombotic microangiopathy may be induced by rejection
obstruction or, most significantly, vascular thrombosis or or as a secondary consequence of cyclosporine, tacrolimus,
acute rejection. Assessment of the patient’s volume status or sirolimus therapy.14 CNI blood levels should be measured
may help eliminate hypovolemia as a cause of decreasing regularly during the immediate postoperative period until
urine output. DGF can be ascertained further with duplex target levels are reached,8 as this measurement may indi-
ultrasonography to assess perfusion of the graft and to cate the likelihood of CNI toxicity versus rejection in the
exclude renal artery or vein thrombosis. Duplex ultraso- diagnosis of graft dysfunction. The level of mammalian tar-
nography also allows the diagnosis of a urinary complica- get of rapamycin (mTOR) inhibitor should also be measured
tion such as obstruction or leak. regularly if this class of drugs is used.
Measures to decrease the likelihood of DGF often are used Mechanical problems usually are the result of compli-
during the operative procedure and in the perioperative cations of surgery or specific donor factors, such as mul-
period. Maintenance of adequate blood pressure and fluid tiple arteries, that lead to posttransplantation dysfunction.
status may be accomplished with intravenous albumin or Mechanical/surgical factors include obstruction of the
crystalloid.9 There is no evidence for the superiority of one transplant, hematuria, urine leak or urinoma, and vascular
type of fluid during kidney transplant; however, the use problems such as renal artery or vein stenosis or thrombo-
of normal saline is associated with a higher incidence of sis. Postoperative bleeding is another potential complica-
acidosis.10 Shorter cold ischemia or pulsatile perfusion of tion that may cause compression of the transplant because
the donor organ also may decrease the likelihood of post- the transplant usually is placed in the retroperitoneal space.
operative DGF, and there is ongoing evaluation of normo- Posttransplant lymphoceles are another common cause of
thermic perfusion techniques in this context. Some centers early transplant dysfunction. Lymph drainage from tran-
have used intraarterial calcium channel blockers, such as sected lymphatic channels accumulates in the perivascular
verapamil, to improve renal blood flow.11 It is common and periureteral space and can cause ureteral obstruction
practice to administer mannitol (12.5 g) about 10 min- or lower-extremity swelling from iliac vein compression.
utes before the kidney is reperfused, which helps trigger an
osmotic diuresis and might be protective. Loop diuretics are
also commonly used at the time of renal reperfusion. Oral
calcium channel blockers have been used to decrease the Surgical Complications
incidence of DGF.12 There is controversy about the early ini-
tiation of CNIs because of the potential for nephrotoxicity. URINARY PROBLEMS
Some centers delay the use of CNIs until there is established
diuresis. If additional immunosuppression is desired, poly- Urinary Obstruction
clonal or monoclonal anti–T-cell antibodies may be used as After implantation of a living donor kidney transplant, urine
induction therapy. output begins immediately or within minutes. (See Chap-
ter 29 for a more complete discussion of urinary problems.)
The same is not generally true of deceased donor kidneys, in
GRAFT DYSFUNCTION
which urine output may not be apparent for 1 hour or more
Early complications of renal transplantation may be after implantation and may be sluggish or nonexistent for
mechanical/surgical or medical. Early medical problems days if the kidney has been injured (DGF) by donor factors
are more common than posttransplant surgical problems or preservation. If a kidney that was formerly making urine
200 Kidney Transplantation: Principles and Practice

A B C
Fig. 14.1 This patient presented with an elevated creatinine level. Ultrasound showed pelvicaliceal dilation. (A) A percutaneous nephrostomy tube was
placed, and the next day a nephrostogram was obtained. (B) The midureteral stenosis was crossed successfully with a guidewire, and the ureter was
dilated with a balloon (the waist of the dilated balloon corresponds to the stricture). (C) Subsequently, a double-J stent was placed from the renal pelvis
into the bladder across the dilated stricture.

slows down or stops and does not respond to fluid administra- stent placed during surgery is significantly lower compared
tion, urinary obstruction must be considered in the differen- with those who did not have a ureteral stent placed during
tial diagnosis. The initial evaluation is to check the patient’s the transplant.15 However, placement of ureteral stents
vital signs and physical examination to ensure adequate during transplant carries a higher risk of infection so it is
hydration and to check that the Foley catheter is functioning recommended that a sulfa-based antibiotic prophylaxis be
correctly. Obstruction of the Foley catheter by blood clots eas- administered to these patients.16 Possible explanations for
ily may occur and can be cleared by gentle irrigation. If these obstruction are a twisted ureter or anastomotic narrowing.
problems are not present, renal transplant ultrasound is the Generally, obstructions appear several weeks postopera-
fastest, most accurate, and least expensive method to assess tively, after the stent has been removed, and occur most fre-
the renal pelvis for obstruction. Pelvicaliceal and/or ureteral quently at the anastomosis between ureter and bladder.17
dilation seen by ultrasound implies distal obstruction. If the Usually, these obstructions can be crossed by a guidewire
bladder is collapsed rather than full, the problem is likely and dilated percutaneously by an interventional radiolo-
to be ureteral obstruction. Treatment should be immediate gist (Fig. 14.1). If the nephrostogram shows a long (>2 cm)
decompression of the renal transplant pelvis by percutane- stricture, especially a proximal or midureteral stricture, it is
ous insertion of a nephrostomy tube. Subsequently (usually likely to be a result of ischemia and is not usually amenable
1 or 2 days later to allow blood and edema to clear after neph- to balloon dilation, necessitating surgical repair (Fig. 14.2).
rostomy tube placement), a nephrogram can be obtained to The operation of choice for a long stricture or one that has
evaluate the ureter for stenosis or obstruction. The diagnosis failed balloon dilation is ureteroureterostomy or ureteropy-
is confirmed by a decline in the serum creatinine level after elostomy using the ipsilateral native ureter. The spatulated
decompression of the renal pelvis. ends of the transplant and native ureters are anastomosed
After the Foley catheter is removed, the most common using running 5-0 absorbable suture. This anastomosis
cause of urinary obstruction is not ureteral stenosis, but can be done over a 7 French double-J stent, which is left in
rather bladder dysfunction. This problem is particularly place for 4 to 6 weeks. If no ipsilateral ureter is available, it
common in diabetic patients with neurogenic bladders. may be necessary to use the contralateral ureter. If neither
Initial management is replacement of the Foley catheter the ipsilateral ureter nor the contralateral ureter is avail-
and a trial of an alpha-blocker, such as tamsulosin, doxa- able, alternatives include bringing the bladder closer to the
zosin, or terazosin. If bladder dysfunction persists after one kidney using a psoas hitch or fashioning a Boari flap, but
or two such trials, it may be necessary to start intermittent these measures are seldom necessary.18 Another method
self-catheterization. In rare instances in which bladder is endoureterotomy; experience with this method is grow-
dysmotility is severe and urinary tract infections are com- ing.19 Even if urinary obstruction is clinically silent (i.e., the
mon, it may be preferable to drain the transplant ureter into patient is asymptomatic with a normal creatinine value),
an ileal conduit to the anterior abdominal wall. Ideally, a urinary obstruction manifested by dilation of the pelvis
patient with a neurogenic bladder should have been evalu- and calices on ultrasound should be treated because it ulti-
ated before transplantation with urodynamic studies, and a mately leads to thinning of the renal cortex and loss of renal
decision should have been made about management at that function. Urinary obstruction should be treated immedi-
time (see Chapters 4 and 12). ately to minimize damage to the transplanted kidney.
During the first 1 or 2 weeks after transplantation,
obstruction usually is caused by a technical problem related Bleeding Into the Urinary System
to surgery (see Chapter 29). If a ureteral stent was placed Gross hematuria is common immediately postoperatively
at the time of surgery, it is highly unusual to have obstruc- because of surgical manipulation of the bladder. The
tion. Indeed, the incidence of major urologic complications Leadbetter–Politano procedure for ureteroneocystostomy
after kidney transplant in patients who had a prophylactic is associated with more hematuria compared with the
14 • Early Course of the Patient With a Kidney Transplant 201

retroperitoneal position, a urinoma collects around the


kidney and bladder and causes a bulge in the wound and
pain with direct displacement of adjacent viscera, includ-
ing the bladder. The diagnosis should be suspected if the
serum creatinine level is increasing (or not decreasing
appropriately). Adjunctive tests to help make the diag-
nosis of urine leak, if it is not obvious clinically, include a
renal scan, which would show urine in the retroperitoneal
space surrounding the bladder or around loops of bowel,
or an ultrasound, which would show a fluid collection out-
side the bladder and when aspirated has a high creatinine
level. Urine leak generally is because of a surgical problem
with the ureteroneocytostomy or ischemic necrosis of the
distal ureter. Other causes include postbiopsy injury and
ureteral obstruction. Such a leak should be immediately
repaired surgically because the risk of wound infection
increases with delay in treatment.

VASCULAR PROBLEMS
Arterial Stenosis
Transplant renal artery stenosis is a relatively common
vascular complication after kidney transplant with an inci-
dence of 1% to 23%.21 It may manifest in the early postop-
erative period by: (1) fluid retention, (2) elevated creatinine
levels, and (3) hypertension.22 (See Chapters 28 and 30
for a more complete discussion of vascular problems.)
Commonly, the patient does not tolerate cyclosporine or
tacrolimus because these drugs exacerbate the preexisting
ischemia at the glomerular arteriolar level. The aforemen-
tioned triad of clinical findings need not all be present, and
Fig. 14.2 This intraabdominal kidney transplant was found by ultra- the diagnosis should be suspected for any one of the three
sound to be obstructed. A nephrostomy tube was placed, and a neph- clinical signs. Cytomegalovirus (CMV) infection and DGF
rostogram was obtained the next day. The kidney had rotated medially have been described as risk factors for transplant renal
and twisted the ureter proximally. The patient was managed opera-
tively by placing the kidney laterally in a retroperitoneal pocket and
artery stenosis.23 If the creatinine level is greater than 2
performing ureteroureterostomy using the ipsilateral native ureter. mg/dL, renal arteriography is best avoided because of the
nephrotoxicity of the contrast dye. Magnetic resonance
angiography usually can give an accurate delineation of
extravesical approach typified by the Lich-Gregoir tech- the arterial anatomy. Ultrasound also is safe, but less dis-
nique or the technique described by us (see Chapter 11).20 criminatory, and may be helpful if jetting of flow beyond a
The advantage of the latter technique is that it effectively stricture is seen.
prevents reflux and can be done with excellent long-term As the population of renal transplant recipients has
results. Occasionally, continuous bladder irrigation is nec- become older and includes more diabetic patients and
essary if gross hematuria is associated with clots, although patients with vascular disease, transplant renal artery
intermittent manual irrigation usually is adequate. Bladder pseudostenosis has become increasingly common. Pseu-
outlet obstruction by a blood clot is an emergency; vigilant dostenosis refers to arterial stenosis in the iliac artery
nursing care is required to ensure that it does not occur. It is proximal to the implantation of the transplant renal artery.
preferable not to distend the bladder in the immediate post- Although the anastomosis and renal artery may be com-
operative period to avoid disrupting the bladder sutures or pletely normal, a more proximal iliac artery stenosis can
causing a leak, and continuous bladder irrigation and cys- lead to hypoperfusion and resulting high renin output by
toscopy ideally are avoided. Minor hematuria without clots the transplanted kidney.
is common in the first 1 or 2 days regardless of the surgi- Treatment of transplant renal artery stenosis and pseudo-
cal method of ureteroneocystostomy and does not require stenosis includes both percutaneous interventions and sur-
treatment; it resolves over time without specific treatment. gery. Generally, ostial stenosis, long areas of stenosis, and
stenosis in tortuous arteries difficult to access radiographi-
Urine Leak cally are not treated as successfully with percutaneous
A leak of urine from the transplanted kidney in the early interventions (balloon dilation or stenting) as with surgery.
postoperative period may be clinically obvious if the Stenoses within smaller branches of the renal artery may
patient presents with abdominal pain, an increasing cre- be treatable only by angioplasty. Iliac artery disease caus-
atinine level, and a decrease in urine output. Urine in ing pseudostenosis may be treated by angioplasty, but risks
the peritoneal cavity causes peritonitis and pain. More embolization or dissection, leading to thrombosis or further
commonly, assuming that the kidney was placed in the ischemia. A recent systematic review of transplant renal
202 Kidney Transplantation: Principles and Practice

A B
Fig. 14.3 This patient presented with fluid retention, hypertension, and an elevated creatinine level. (A) An arteriogram showed that the artery to the
lower pole arising from a common aortic patch was stenotic proximally. (B) This stenosis was successfully treated with balloon angioplasty with resolu-
tion of the patient’s symptoms.

artery stenosis treated with either percutaneous angio- In cases of more than one renal transplant artery in
plasty or stenting demonstrated equivalent outcomes with which arterial reconstruction is performed at implantation,
success rates ranging from 65% to 94%.24 Surgical options there may be increased risk of thrombosis of one or more
include bypass of the stenosis using autologous saphenous arteries. This increased risk is a particular concern if there
vein, a prosthetic graft, or an allogeneic arterial graft pro- is a small accessory renal artery supplying the lower pole of
cured from a deceased donor. The risk of the procedure the kidney and providing the ureteral blood supply. Throm-
has to be weighed against the potential benefit of improv- bosis of a branch artery may manifest as an increase in
ing renal transplant blood flow. In addition to the serum serum creatinine levels associated with hypertension. Angi-
creatinine determination, a biopsy may be useful to assess ography shows partial thrombosis and loss of perfusion of a
the quality of the renal parenchyma. In advanced chronic wedge-shaped section of renal parenchyma. The risk of this
rejection with an elevated creatinine level for more than 1 situation, in addition to potential long-term hypertension,
month, it may not be prudent to repair such arteries, but is caliceal infarction and urine leak in the early postopera-
this problem is not generally encountered in the early post- tive period. Such kidneys, with partial infarction, generally
transplant course. Fig. 14.3 shows a renal artery stenosis can be salvaged. Urine leaks occurring through the outer
in the lower pole artery that was managed successfully by cortex of the kidney after partial infarction may be man-
balloon angioplasty. aged by nephrostomy tube placement for urinary drain-
age and placement of another drain adjacent to the kidney
Arterial Thrombosis to prevent urinoma. When the transplant ureter necroses
Renal transplant arterial thrombosis usually occurs early as a result of arterial ischemia, alternative urinary drain-
(within 30 days) in the posttransplant period, but is a rare age needs to be provided surgically; this would be man-
event and is generally caused by a technical error at the aged most often by ureteropyelostomy using the ipsilateral
time of surgery.25 It usually is related to an intimal injury native ureter.
to the donor kidney during procurement or to anastomotic
narrowing or iliac artery injury during implantation. The Renal Vein Thrombosis
incidence of renal transplant arterial thrombosis is around Renal vein thrombosis occurs in between 0.1% and 4.2%
1% to 2%.26 Kidneys from donors younger than 5 years of recipients and is more common in deceased donor trans-
old have been associated with a higher risk of thrombo- plants.29 Thrombosis may occur as a technical complica-
sis.27 The kidney tolerates only 30 to 60 minutes of warm tion when the donor renal vein was narrowed by repair of
ischemia before it is irreversibly injured, making it difficult an injury or when the vein was twisted or compressed exter-
to diagnose and correct this problem before it is too late to nally, but it may occur in the absence of a technical compli-
salvage the kidney. The diagnosis should be suspected in cation. Risk factors for renal vein thrombosis include use
a patient who has had a transplant hours to days before of the right donor kidney, prolonged ischemic time, older
and has had good urine output but who suddenly has a donors, older recipients, use of peritoneal dialysis pretrans-
decrease in urine output. A high degree of suspicion has to plant, hypercoagulable states in the recipient, and peri-
be present, and the patient should be returned to the oper- operative hypotension in the recipient.30 The diagnosis is
ating room promptly. Although some reports of catheter- indicated by sudden onset of gross hematuria and decrease
based thrombolysis for renal artery thrombosis have been in urine output, associated with pain and swelling over the
described, the majority of cases require operative inter- graft. Ultrasound shows absence of flow in the renal vein,
vention and if unsuccessful require transplant nephrec- diastolic reversal of flow in the renal artery (Fig. 14.4), and
tomy.28 If the patient had urine output preoperatively an enlarged kidney, often with surrounding blood. Ultra-
from the native kidneys, the diagnosis is difficult to make sound can point to this diagnosis definitively. Only if it is
in a timely manner because urine output may continue immediately recognized and repaired can this problem be
after the renal transplant has thrombosed. The advantage reversed. Immediate surgical repair of the vein and con-
of diagnostic ultrasound has to be weighed against the trol of bleeding are required, and it is generally necessary
disadvantage of delaying a return to the operating room. to remove the kidney and revise the venous anastomosis.
Almost all kidney transplants with arterial thrombosis are However, some instances of catheter-directed thrombolytic
lost because of ischemic injury. therapy or thrombectomy have been successfully reported
14 • Early Course of the Patient With a Kidney Transplant 203

long the kidney has been in place. If nephrectomy is per-


formed within 4 weeks, there are minimal adhesions, and
the vessels are exposed easily for ligation and transplant
nephrectomy. At later times, it is usually easiest to reopen
the transplant incision and enter the subcapsular plane
around the kidney. The kidney is dissected free in the sub-
capsular space, and a large vascular clamp is placed across
the hilum. The kidney is amputated above the clamp, and
3-0 polypropylene (Prolene) is used to oversew the hilar
vessels. The ureter also is oversewn (see Chapter 11).

Rejection During the Early


Postoperative Period
Fig. 14.4 Ultrasound shows absence of flow in the renal vein and HYPERACUTE REJECTION
reversal of diastolic flow in the renal artery. This kidney was enlarged
to 14 cm in length with a surrounding fluid collection that represented Hyperacute rejection is the immediate rejection of the donor
blood. These ultrasound findings were pathognomonic of transplant kidney upon reperfusion and is mediated by preformed anti-
renal vein thrombosis. The condition was treated surgically with exci- bodies against the donor. The risk of hyperacute rejection
sion of the kidney, placement of a venous extension graft using donor or of antibody-mediated rejection (AMR) is increased when
iliac vein obtained from a third-party donor, and reimplantation of the
kidney. Three weeks later, the patient had a normally functioning kid-
a renal transplant is performed in the setting of ABO mis-
ney transplant. match or a positive lymphocytotoxic crossmatch (see Chap-
ter 22). Hyperacute rejection is now a rare event because
of our understanding of transplant immunology and the
in the setting of early postoperative renal vein thrombo- implementation of more stringent immunologic testing of
sis.31 Bleeding from the swollen and cracked kidney surface donors and recipients to prevent such occurrences. Cur-
usually can be controlled with hemostatic agents. rent guidelines recommend molecular human leukocyte
antigen (HLA) typing both the recipient and donor before
POSTOPERATIVE BLEEDING kidney transplant because HLA matching for HLA-A, HLA-
B, and HLA-DR, with an emphasis on HLA-DR matching,
As with all surgery, postoperative bleeding may complicate has been shown to improve kidney transplant outcomes.34
renal transplant outcomes. Bleeding generally occurs dur- For nonsensitized patients with no preformed antibodies,
ing the first 24 to 48 hours after transplantation and is diag- it is reasonable to proceed with transplantation with no
nosed by a decreasing hematocrit, swelling over the graft prospective crossmatch and this strategy of “virtual cross-
with a bulging incision, or significant blood seepage from matching” has been developed to minimize cold ischemic
the incision. Postoperative bleeding occurs in roughly 12% time.35 For sensitized patients that have preformed anti-
of kidney transplant recipients and is more likely to occur HLA antibodies, the selection of donors toward whom the
in those with calcified iliac vessels who receive higher doses patient has no preformed antibodies is critical for the suc-
of heparin as prophylactic anticoagulation and in patients cess of the transplant. Therefore highly sensitized patients
taking anticoagulation agents for other medical problems should undergo further serologic typing of the donor and
such as coronary artery or cerebrovascular disease.32 high-resolution HLA typing of both the donor and recipi-
Patients treated with clopidogrel for underlying cardiac dis- ent.36 In addition, sensitized kidney transplant candidates
ease are at significant risk for postoperative bleeding; this should undergo a complement dependent cytotoxicity
class of medications should be avoided or discontinued 1 crossmatch and a flow cytometric crossmatch with the
week before renal transplantation if acceptable from a car- putative donor to prevent hyperacute and acute rejec-
diac perspective.33 If the hematoma is not clinically obvi- tion.34 Although this rigorous immunologic testing of
ous, an ultrasound or computed tomography (CT) scan can donor and recipient has drastically reduced the incidence of
define its size and help determine whether or not surgical hyperacute rejection, there are reports of hyperacute rejec-
evacuation is appropriate. Treatment includes immediate tion occurring even in the setting of negative crossmatch
surgery and blood transfusions as necessary. results.37 A hyperacutely rejected kidney has no perfusion
on renal scan because of microvascular thrombosis and
GRAFT LOSS AND TRANSPLANT NEPHRECTOMY should be removed. The introduction of solid-phase assays
based on the Luminex platform may help identify those with
During the early posttransplant period, if a renal transplant donor-specific antibody (DSA) that may cause a problem in
loses perfusion because of thrombosis or because of hyper- the absence of positive crossmatch results, but these assays
acute, acute, or accelerated vascular rejection, it must be require more rigorous validation and standardization. The
removed. Otherwise, the systemic toxicity of a necrotic incidence of hyperacute rejection is not 100% in those with
kidney may cause fever, graft swelling or tenderness, and preformed antibodies, presumably because some antibodies
generalized malaise. Loss of perfusion can be assessed by have lower affinity, lower density, do not bind complement,
nuclear scan or duplex ultrasound. The technically easiest or cause accommodation.38 In some cases, blood type A2
way to perform a transplant nephrectomy depends on how donors may be transplanted successfully to type O recipients
204 Kidney Transplantation: Principles and Practice

because type A2 expresses less of the putative antigen, but transplantation, but it can occur at virtually any later time.
this strategy also has increased risk of graft loss.39 Desensi- The highest incidence of acute rejection is within the first 3
tization is a strategy to remove preformed antibody before months, and overall rates of rejection vary from 5% to 25%
transplantation to prevent hyperacute and antibody-medi- within the first 6 months, depending on HLA matching and
ated rejection. Desensitization regimens include the use of the immunosuppressive protocol. The clinical harbingers of
plasmapheresis combined with intravenous immunoglobu- acute rejection include an increasing creatinine level, weight
lin and/or rituximab and are a growing area of research.40 gain, and graft tenderness. Often, there are no physical signs
If at all possible, a crossmatch-negative, ABO-compatible or symptoms, making the diagnosis largely dependent on
recipient should be identified for the transplant candidate or laboratory assessment of renal function. Better diagnos-
the kidney can be shipped to a center that has such a patient tics based on urine or blood molecular analysis have been
awaiting a kidney, potentially in exchange for a kidney to developed but are at early stages of clinical application.47–49
which the intended recipient has a negative crossmatch. A The current diagnosis of acute rejection is based on kidney
strategy such as this will maximize outcomes and utility for transplant biopsy and histopathologic changes, including
the kidney transplant community. tubulitis (invasion of tubules by lymphocytes), glomeruli-
tis, and arteritis, which are classified according to the Banff
Classification System.50 The importance of biopsy confirma-
ANTIBODY-MEDIATED REJECTION
tion of rejection relates to the risk of increasing immunosup-
Despite a negative T-cell crossmatch test preoperatively, pression in patients whose graft dysfunction is not caused by
some patients may develop an early aggressive form of rejec- rejection but perhaps infection or other causes that might be
tion, termed antibody-mediated rejection (AMR).38 The inci- exacerbated by increased immunosuppression.
dence of AMR is greater than 20% in sensitized patients.41 First-line treatment of acute cellular rejection is bolus
The diagnosis of AMR is based on the presence of DSA in steroid therapy with methylprednisolone sodium succi-
recipient serum and biopsy of the transplanted kidney dem- nate (Solu-Medrol). Many regimens are used successfully,
onstrating microvascular inflammation (glomerulitis or but typical dose and duration are 10 mg/kg intravenously
peritubular capillaritis), immune cell infiltration, and usu- daily for 3 days (up to a maximum single dose of 500 mg/
ally evidence of complement activation by C4d staining of day). About 85% to 90% of acute cellular rejection episodes
peritubular capillaries.42 are steroid-responsive. If the patient’s serum creatinine
AMR is seen most often in sensitized patients with DSA of level has not begun to decrease by day 4 of therapy, alter-
high mean channel fluorescence by flow crossmatch. Often native treatment must be considered, such as antilympho-
such patients have had a previous transplant. The time cytic globulin, alemtuzumab (Campath-1H), or rituximab
course of this type of rejection is typically within days to (anti-CD20) as lymphocytotoxic therapy. Many centers use
weeks of the transplant, although it may occur at any time; antibody-depleting therapy first line for all severe vascular
it tends to be poorly responsive to steroids and occasionally rejections (Banff 2A and 3), particularly if anti-IL-2 induc-
resistant to all forms of antirejection therapy. Indeed, renal tion was used. However, antibody-depleting therapies may
transplant patients who develop DSA have 60% 5-year graft be associated with an increase in infectious complications
survival compared with 80% graft survival in those who when used to treat rejection compared with when used
do not develop DSA.43 Although successful prophylaxis of for induction.51 A recent systematic review of randomized
rejection has been described using intravenous immuno- trials in treating acute rejection demonstrated that anti-
globulin, rituximab, plasmapheresis, or thymoglobulin in lymphocyte antibody therapies are likely superior to ste-
highly sensitized patients, when this form of rejection has roids in the initial treatment of acute cellular rejection in
started there is no standard treatment.44 The KDIGO guide- terms of reversing rejection and preventing graft loss, but
lines recommend that such rejection be treated with one or there was no difference in subsequent rejection episodes or
more of the following, with or without steroids: plasmapher- patient survival, and the antibody therapies carry a higher
esis, intravenous immunoglobulin, anti-CD20 antibody, or rate of adverse events compared with steroid treatment.52
other lymphocyte-depleting antibody.8 Other novel strate- Rejection that does not respond to treatment with steroids
gies to treat AMR include targeting complement, targeting or antibody therapy occurs in less than 5% of patients,
plasma cells with proteasome inhibitors, targeting the ger- although more frequently in sensitized patients or repeat
minal center reaction with anticytokine or cytokine recep- transplants with significant DSA present.
tor antibodies or costimulatory blockade (belatacept), or The effect of acute cellular rejection on graft survival
directly targeting antibodies by enzymatic cleavage.45 Ran- depends on the response to treatment, with minimal effect
domized controlled trials are needed to determine which of if treatment results in return to baseline function but nega-
these therapies (or combination of therapies) is most effec- tive effect with incomplete response or repeated rejection
tive in treating AMR.46 episodes.53 Whether or not an early rejection episode pre-
disposes the kidney to chronic rejection is controversial but
likely depends on the complete resolution of the rejection
ACUTE REJECTION
and associated DSA.
The most common form of immunologic rejection in the
early posttransplant period is acute cellular rejection, medi- BORDERLINE REJECTION
ated predominantly by host T lymphocytes responding to
the allogeneic major histocompatibility complex (MHC) The entity of borderline rejection on kidney transplant
antigens on the donor kidney. Without adequate immuno- biopsy is of uncertain significance. Whereas some studies
suppression, acute rejection typically occurs 5 to 7 days after demonstrate that treatment of borderline rejection with an
14 • Early Course of the Patient With a Kidney Transplant 205

increase in the patient’s immunosuppression can improve problems with the vascular or ureteral anastomoses. The
graft function, others have found little benefit in treating major differential diagnostic considerations in a recipient
those with borderline rejection on biopsy results.54 There- with decreasing or absent urine output are volume deple-
fore if a kidney biopsy is interpreted as “borderline” by Banff tion or an acute vascular or urologic complication. Other
criteria, we suggest the decision to treat be made on an indi- conditions that can mimic DGF are rejection and recurrent
vidual basis according to the clinical picture of the recipient. focal segmental glomerulosclerosis (FSGS). This differential
diagnosis can be evaluated with urgent ultrasound or radio-
nuclide renal scanning. Typically, a transplant with DGF
shows good renal perfusion and good parenchymal uptake of
Medical Complications orthoiodohippurate (123I-OIH) or mercaptoacetyltriglycine
(99mTc-MAG3) with poor or no renal excretion. A biopsy
DELAYED GRAFT FUNCTION
of the kidney transplant should be performed to assess for
DGF is the most common early complication after kidney rejection and acute tubular necrosis and is the gold standard
transplantation. The definition of DGF has been the subject for diagnosis. Hemodialysis should be used when clinically
of much debate but has been traditionally defined as the indicated in the posttransplant period in recipients with DGF;
need for dialysis during the first week after transplanta- however, careful attention to fluid status is paramount to
tion. A more recent and objective definition of DGF termed decrease the frequency and necessity for dialysis. The usual
functional DGF is defined as the failure of serum creatinine time course of DGF is 10 to 14 days.
to drop by 10% on three consecutive days in the first post- A major concern for transplant recipients with DGF is the
transplant week.55 The most recent data from the Organ potential for early acute rejection. DGF may lead to activa-
Procurement and Transplantation Network (OPTN) and tion of the immune system with release of cytokines and
Scientific Registry of Transplant Recipients (SRTR) Annual adhesion molecules (see Chapter 25). This situation may
Data Report 2012 reports rates of DGF in the United States result in an alloimmune response, leading to an increased
at 15% to 20%. This rate is higher for deceased donors at frequency of acute rejection. A recent study of pooled data
around 20% to 25%, with recipients of DCD kidney trans- reported 49% of DGF patients experienced an episode of
plants carrying the highest rates of DGF at about 40%. The acute rejection, compared with 35% of those without
long-term clinical consequences of DGF are significant. DGF.59 This increase in rejection rate may contribute to the
Recent reports have demonstrated that DGF is associated reported decrease in allograft survival in DGF patients.60
with a 38% increase in acute rejection and a 53% increased The diagnosis of rejection in patients with DGF may be
risk of mortality in recipients of deceased donor kidneys.56 hindered because the primary clinical monitoring tool is a
Both donor and recipient factors contribute to the risk for decrease in serum creatinine levels. For this reason it is often
DGF. The most notable risk factors associated with DGF are useful to obtain periodic biopsies of the transplanted kidney
cold ischemic time, receipt of a DCD kidney, donor age, body in recipients with DGF to assess for rejection, and this may
mass index (BMI), and terminal creatinine level. Other donor be performed as often as every 7 to 10 days while on dialysis
factors associated with increased risk for DGF include anoxic for DGF.8 Although the use of antilymphocyte agents such
or cerebrovascular cause of death, a history of hypertension, as thymoglobulin or alemtuzumab will not necessarily
increased warm ischemia time, and greater HLA mismatch. treat the DGF, some centers use these agents to prevent or
Recipient characteristics associated with increased risk of decrease the incidence of rejection in patients with DGF.61
DGF include male sex, black race, history of diabetes, lon- Prevention of DGF and early recognition of rejection are
ger time on dialysis, increased BMI, frailty, elevated Panel important goals to help improve early and long-term graft
Reactive Antibody (PRA), prior transplant, previous blood survival.
transfusions, elevated pretransplant phosphate levels, and
a mismatch in body size between the donor and the recipi- NEPHROTOXICITY FROM CALCINEURIN
ent.57 Despite this knowledge, a reliable method for predict- INHIBITORS
ing DGF in kidney transplant recipients remains elusive.
Several methods are available to help reduce the risk for DGF. Early institution of the CNIs cyclosporine and tacrolimus
For instance, it appears that the incidence of DGF may be at the time of transplantation is important to prevent acute
reduced by the use of pulsatile perfusion of the procured renal rejection episodes. Because of the potential for additive
allograft; however, the effect of this technology on long-term nephrotoxicity, however, some centers avoid instituting
graft outcomes is unclear.58 CNIs until there is adequate function of the transplanted
The pathophysiology of DGF is not completely under- kidney. Centers that delay the onset of CNIs usually use
stood. It is thought to be a multifactorial process attributed antibody induction therapy to lower the incidence of early
to multiple donor and recipient factors that together medi- acute rejection.60 Other centers, including ours, begin
ate an ischemia-reperfusion injury that ultimately results administering CNIs early in the posttransplant course,
in oxidative stress and an inflammatory cascade resulting whether or not the allograft is functioning well or in DGF.
in cell injury and death within the graft.57 The diagnosis of Both of the CNIs, cyclosporine and tacrolimus, are effective
DGF is usually apparent during the first 24 hours after trans- in preventing acute rejection episodes, but they can lead
plantation; the most common clinical scenario is a decline to nephrotoxicity. Acute CNI-induced nephrotoxicity is a
in urine output unresponsive to a fluid challenge. The dif- dose-dependent and reversible process that occurs early
ferential diagnosis of DGF includes the same prerenal, renal, after the initiation of CNI therapy. This acute nephrotoxic-
and postrenal causes of acute kidney injury as for native ity after initiation of CNI therapy has been reported to occur
kidneys with additional emphasis on potential technical in roughly 20% of renal allograft recipients.62 Development
206 Kidney Transplantation: Principles and Practice

of CNI nephrotoxicity may have long-term consequences a transplant biopsy if clinical suspicion for acute rejection
for the kidney allograft because it has been associated with is high. Antihypertensive medications should be used care-
the later development of chronic allograft nephropathy.63 fully in the posttransplant period to avoid hypotension,
Clinical risk factors associated with the development of which may further worsen renal blood flow. Meticulous
acute CNI nephrotoxicity include the following: systemic attention to daily weights, intake and output, and assess-
overexposure to cyclosporine or tacrolimus, interactions ment of orthostatic blood pressure changes can diagnose
with other drugs such as mTOR inhibitors that interfere volume depletion as a contributing factor to renal allograft
with ABCB1-mediated transport in tubular epithelial cells, dysfunction. Volume repletion with intravenous or oral
interactions with drugs such as ketoconazole that alter the fluids is indicated in the setting of prerenal azotemia from
metabolism of CNIs by CYP3A enzymes, older kidney age, volume depletion in the postoperative kidney transplant
use of NSAIDs, use of diuretics, and salt depletion.64 The recipient.
pathophysiology of this acute nephrotoxicity is related to
vasoconstriction of the afferent arterioles, which is medi- OTHER DRUG TOXICITY
ated by an increase in the production of vasoconstrictors
such as endothelin and thromboxane, and activation of Transplant patients often have complex pharmacologic
the renin angiotensin system by supratherapeutic levels of regimens at the time of transplantation, which may include
cyclosporine or tacrolimus. Increased free radical formation nephrotoxic medications or medications that may cause con-
and activation of the sympathetic nervous system in native comitant nephrotoxicity with CNIs. Examples of the former
kidneys also play a part in acute CNI nephrotoxicity.64 include nonsteroidal antiinflammatory drugs and nephro-
Although there are many clinical parameters that have toxic antibiotics such as amphotericin and aminoglycosides.
been advocated to differentiate CNI nephrotoxicity from Tacrolimus and cyclosporine are metabolized by the cyto-
rejection, most clinical parameters are of insufficient sen- chrome P-450-3A4 system (CYA P-450-3A4), and medi-
sitivity to predict confidently the cause of the transplant cations that are also metabolized by CYA P-450-3A4 may
dysfunction. In patients with DGF, it may be more difficult increase their blood levels. Examples of drugs that may inter-
to diagnose acute rejection or CNI nephrotoxicity reliably. act with the metabolism of CNIs include nondihydropyridine
Suspicion of acute CNI nephrotoxicity should be raised calcium channel blockers such as diltiazem and verapamil,
when renal allograft function declines and there is evidence erythromycin, ketoconazole, and fluconazole.64 Grapefruit
of elevated drug levels of cyclosporine or tacrolimus. The juice also has been shown to increase the gastrointestinal
most reliable way of differentiating CNI nephrotoxicity from absorption of cyclosporine (see Chapters 16 and 17). Selec-
rejection is percutaneous renal allograft biopsy. Generally, tive serotonin reuptake inhibitor antidepressants are another
biopsies can be performed safely soon after transplantation, class of pharmacologic agents that need to be used with care.
using real-time ultrasound imaging and automated biopsy In particular, nefazodone and fluvoxamine are metabolized
needle devices. Although it can be difficult to differentiate by CYA P-450-3A4 and may increase CNI blood levels.66
between CNI nephrotoxicity and other potential diagnoses Routine drug level monitoring is important when drugs
in the renal allograft, there is a scoring system based on that are metabolized by CYA P-450-3A4 are used. Adjust-
histologic features of CNI toxicity that can aid in the diag- ment of the daily dose of cyclosporine and tacrolimus to
nosis and that correlates with future graft function.65 Most attain therapeutic blood levels helps prevent episodes of
cases of acute CNI nephrotoxicity are reversible by lowering nephrotoxicity from the concomitant use of these agents.
the dose of the CNI. Because of the variability of intestinal Avoidance of medications that interfere with drug metabo-
absorption in the early transplant period, underdosing and lism is desirable. Some studies have demonstrated a person-
overdosing of these agents are common, which can lead alized approach to deciding the best immunosuppressive
to rejection episodes or CNI nephrotoxicity. Monitoring regimens for kidney transplant recipients as associations
cyclosporine and tacrolimus levels is valuable in prevent- of drug toxicities with some CYP3A genotypes or single-
ing significant increases in blood levels, which may lead to nucleotide polymorphisms in certain genes can influence
nephrotoxicity.8 Some centers routinely use a high-dose the metabolism of these drugs and the interaction between
CNI protocol to prevent rejection and accept a certain level the immunosuppressants and other drugs.62
of nephrotoxicity as a consequence.
RECURRENT DISEASE
PRERENAL AZOTEMIA AND VOLUME DEPLETION
Most causes of renal failure do not recur in the transplanted
Prerenal azotemia from volume depletion may lead to dete- kidney; when they do, it is usually later in the posttrans-
rioration of allograft function during the immediate postop- plant course. (See Chapters 4 and 32 for further discussion of
erative period. Overuse of diuretics and uncontrolled blood recurrent disease.) Two diseases may occur in the immediate
glucose are two common causes of volume depletion and posttransplant period and lead to significant graft dysfunc-
prerenal azotemia in the posttransplant kidney recipient. tion or graft loss if not treated aggressively. FSGS is the most
Because most patients are already receiving CNIs, which common glomerular disease that can recur in the immedi-
decrease renal blood flow, the concurrent insult of volume ate postoperative period.67 The overall recurrence rate is
depletion may lead to elevated blood urea nitrogen and approximately 30% to 40%, with most cases recurring in the
serum creatinine levels. It may be difficult to distinguish first posttransplant year.68 The pathogenesis of recurrent
prerenal azotemia from an episode of acute rejection, but FSGS, like primary FSGS, remains poorly understood, but evi-
persistent elevation in serum creatinine even after appro- dence points to a circulating factor or factors present in the
priate volume repletion should prompt further workup with serum as the likely culprit mediating this disease.69 Several
14 • Early Course of the Patient With a Kidney Transplant 207

risk factors have been associated with an increased risk of or thrombocytopenia) or a variety of tissue invasive diseases
developing recurrent FSGS after kidney transplant: these (pneumonia, gastrointestinal disease, hepatitis, encephalitis,
include younger age at primary diagnosis, rapid progression retinitis, nephritis, cystitis, myocarditis, and pancreatitis).77
of primary FSGS to ESRD (<3 years), white race, and loss of Universal CMV prophylaxis with oral ganciclovir or valgan-
a prior transplant because of recurrent FSGS.70 The diag- ciclovir is recommended for all CMV+ recipients and for all
nosis is established by the development of nephrotic range recipients of CMV+ donor kidneys for at least 3 months after
proteinuria in a patient with a pretransplant diagnosis of transplant and for 6 weeks after treatment with antilympho-
FSGS and is confirmed on biopsy. Electron microscopy of the cyte antibody; however, after stopping valganciclovir CMV
allograft biopsy shows diffuse podocyte foot process efface- may still occur.8 CMV disease should be treated with thera-
ment, which is diagnostic in this setting.71 Various strategies peutic doses of valganciclovir or intravenous ganciclovir for
have been used to treat recurrent FSGS, but the mainstays of at least 2 weeks or until CMV replication is no longer detect-
treatment include high-dose CNIs, prednisone, plasmapher- able.78 Epstein–Barr virus (EBV) infection may occur early
esis, and rituximab. Plasmapheresis alone or in combination after transplantation and usually is related to heightened
with any of those treatments seems to be most effective with immunosuppression in a previously seronegative patient. The
a 60% to 70% success rate for this strategy.67 In some cases, type of immunosuppression may also increase the risk of EBV
a course of plasmapheresis may need to be repeated if there is reactivation because replication of EBV occurs more often in
an initial response and subsequent relapse. patients receiving belatacept compared with cyclosporine.79
Another recurrent disease of concern in the immediate In the past, Pneumocystis jirovecii pneumonia was a frequent
postoperative period is thrombotic microangiopathy, which complication of transplantation; however, most centers now
can result from recurrent disease, endothelial injury from use routine prophylaxis with trimethoprim/sulfamethoxazole,
CNIs, hypercoagulable disorders, or AMR.72 Thrombotic which has nearly eliminated the occurrence of this infection
microangiopathy is multifactorial in origin, but recurrent in transplant patients. Other prophylactic agents include anti-
thrombotic microangiopathy is usually associated with a fungal agents, such as fluconazole or clotrimazole troches,
primary diagnosis of atypical hemolytic uremic syndrome which can reduce the risk of mucosal Candida superinfection.
(HUS), particularly those with mutations in genes related to Highly resistant organisms have been detected with
the complement system and its regulation.73 It is character- increasing frequency in transplant patients. Vancomycin-
ized clinically by a decrease in hematocrit or platelet count, resistant Enterococcus (VRE)80 and Candida infections81
or both, with evidence of a microangiopathic process on are becoming significant causes of morbidity in hospital-
peripheral blood smear, increased lactate dehydrogenase ized transplant patients with an incidence of 4% to 11%
levels, and transplant allograft dysfunction. Kidney biopsy in transplant recipients. Risk factors for VRE include pro-
specimens show fibrin deposition in the small arterioles of longed hospitalization, indwelling invasive devices, close
the kidney. Recurrent thrombotic microangiopathy after proximity to a VRE colonized or infected patient, placement
kidney transplant occurs primarily in patients whose pri- in a contaminated room, use of broad spectrum antibiotics,
mary disease was atypical HUS. The incidence of recurrence hepatitis C infection, kidney pancreas transplant, posttrans-
in this population ranges from 50% to 60% and generally plant hemodialysis, reoperation, and presence of a nephros-
occurs in the first month posttransplant.74 Treatment of tomy.80 Treatment options for this infection are limited.
recurrent thrombotic microangiopathy has included dis- Quinupristin/dalfopristin (Synercid), linezolid (Zyvox), and
continuation of the CNI and plasmapheresis. Eculizumab, daptomycin (Cubicin) may be useful for the control of serious
an anti-C5 monoclonal antibody, has proven effective in VRE infections. The investigational drugs oritavancin and
treating renal transplant recipients with recurrent atypi- telavancin are have been shown to have bactericidal activ-
cal HUS related to genetic defects in complement regulatory ity against enterococci resistant to linezolid, daptomycin,
proteins, and with thrombotic microangiopathy related to and vancomycin. The increase in Candida infection seems to
antiphospholipid antibodies.75 be caused by the routine use of clotrimazole or fluconazole
to prevent Candida infection. Intravenous antibiotic use pre-
disposes patients to fungal infection after transplantation.
INFECTION
Infection with extended-spectrum beta-lactam-producing
In the immediate postoperative period most infections are gram-negative bacteria is becoming more common, par-
related to the surgical procedure and usually involve sur- ticularly in the urinary tract.17,82 Many of these patients
gical site infection, bacteremia from a central line, urinary require combination therapy that includes aminoglycoside
tract infection, or pneumonia. (See Chapter 31 for a com- antibiotics; the resulting nephrotoxicity can contribute to
plete discussion of infection.) Prevention of these infections graft loss and many of these antibiotics also influence the
involves meticulous surgical technique, careful line care and levels of immunosuppressant medications.83
use, removal of central lines and the Foley catheter as soon Clostridium difficile colitis is becoming an increasing prob-
as possible, and early mobilization of the patient to prevent lem in solid organ transplant patients; one study found the
atelectasis or pneumonia. Most opportunistic infections do peak incidence occurred 6 to 10 days posttransplant.84 Risk
not occur until after the first 30 days. Of the opportunistic factors associated with the development of Clostridium dif-
infections, CMV is still common after transplantation, par- ficile infection after kidney transplant include VRE coloniza-
ticularly in recipients who are seronegative for CMV and tion before transplant, receipt of an organ from a Centers
who receive seropositive organs, and in those that receive for Disease Control high-risk donor, and administration
antilymphocyte agents such as antithymocyte globulin.76 of high-risk antibiotics within 30 days after transplant.85
CMV disease can manifest as CMV syndrome (characterized Early testing of stool for toxin and treatment with oral vanco-
by fever, malaise, atypical lymphocytosis, and neutropenia mycin or fidaxomicin should be instituted as soon as possible.
208 Kidney Transplantation: Principles and Practice

In persistent or recurrent cases, oral rifaximin or parenteral The high incidence of hypertension in kidney transplant
metronidazole may be required.86 Hands should be washed recipients and the fact that hypertension is an independent
with soap and water, because alcohol-based hand scrubs risk factor for cardiovascular disease in recipients warrants
may not kill the spores and prevent nosocomial transmission. close monitoring and screening for hypertension in this
When an infection occurs in the kidney transplant high-risk population. For this reason, the KDIGO guidelines
recipient, aggressive management is indicated. This man- recommend screening for hypertension at every clinic visit
agement may include removal or replacement of central in kidney transplant recipients.8 Elevated blood pressure
venous catheters or Foley catheters. Any intraabdominal also has long-term consequences for recipients of kidney
fluid collections should be aspirated and drained if clinical allografts: each 10 mmHg increase in systolic blood pres-
suspicion for abscess is high. Urinary tract infections should sure over the upper limit of normal has been associated with
be treated promptly, and the Foley catheter and ureteral a higher risk of graft failure and death.91 In addition, con-
stent should be removed as soon as possible. trolling blood pressure to <140/90 mmHg correlates with
improvement in recipient and graft survival.92 Therefore
kidney transplant patients should be diagnosed with hyper-
HYPERTENSION
tension according to the usual guidelines (elevated blood sys-
Hypertension develops in nearly 80% of renal transplant tolic pressure >140 mmHg or diastolic pressure >90 mmHg
recipients (see Chapter 30). The development of hyper- on at least two occasions) and treated aggressively to a goal
tension after kidney transplant may be related to allograft blood pressure of <130/80 mmHg. Although there are no
dysfunction resulting from DGF, rejection, or immuno- strong data or specific recommendations regarding the types
suppressive medications. Cyclosporine, tacrolimus, and of antihypertensives to use in this population, some obser-
corticosteroids all may contribute to the development of vational studies have suggested avoiding calcium channel
hypertension. CNIs primarily cause hypertension through blockers in specific cases.93 Table 14.2 presents the advan-
widespread arterial vasoconstriction, which increases sys- tages and potential side effects of antihypertensive agents
temic vascular resistance.87 Vasoconstriction in the kidney in transplant recipients. Because of the complexity of this
activates the renin–angiotensin system, promotes sodium patient population, an individualized approach to blood pres-
reabsorption and volume expansion, and increases endo- sure management and selection of the optimal antihyper-
thelin release. CNIs also result in adverse changes in ara- tensive for treatment is important. As discussed previously,
chidonic acid metabolites, nitrous oxide production, and persistently elevated or difficult to control blood pressures,
activation of the sympathetic nervous system.88 Cortico- along with signs of allograft dysfunction in the kidney trans-
steroids cause peripheral vasoconstriction, resulting in an plant recipient, should prompt an additional workup for vas-
acute rise in blood pressure; furthermore, they activate cular compromise of the allograft with imaging.
the mineralocorticoid receptor, resulting in volume expan-
sion.89 Of note, up to 90% of patients are hypertensive before MANAGEMENT OF GRAFT DYSFUNCTION
transplant; preexisting vascular calcification and arterial
stiffness may persist and contribute to ongoing blood pres- The diagnosis and treatment of graft dysfunction are inte-
sure elevation.90 Allograft vascular compromise such as gral components of the successful long-term manage-
posttransplant renal artery stenosis is a treatable cause of ment of the renal transplant recipient. Early diagnosis and
hypertension and should be ruled out in patients who have directed therapy are crucial in the early posttransplant
new-onset hypertension or do not respond readily to antihy- period to initiate appropriate therapy and to avoid potential
pertensive therapy (see previous section). It may be difficult overimmunosuppression. Urine output, serum creatinine,
to ascertain the specific cause of hypertension after renal and urine protein are all useful parameters to measure
transplantation, because multiple factors may be working postoperatively to help identify graft dysfunction in the kid-
in tandem to cause elevated blood pressure in the recipient. ney transplant recipient. Urine output should be measured

TABLE 14.2 Advantages and Potential Side Effects of Antihypertensive Agents in Transplant Recipients
Class Advantages/Indications Side Effects
Diuretics Salt-sensitive hypertension, volume expansion Hyperuricemia
Volume depletion
Beta-blockers Large selection Adverse effect on lipids
Selective agents preferred Relative contraindication with asthma, diabetes,
Added benefit for those with CHF or post MI or peripheral vascular disease
Alpha-blockers Useful with prostatic hypertrophy Postural hypotension (first dose)
Central alpha-agonists Clonidine useful in diabetic patients Dry mouth
Clonidine available as transdermal patch Rebound hypertension
Fatigue
Calcium channel blockers Improve renal blood flow Drug interaction with CNI
May ameliorate cyclosporine nephrotoxicity Edema
(verapamil and diltiazem)
ACE inhibitors and ARBs Proteinuria May cause renal insufficiency
Added benefit in those with CHF and post MI Hyperkalemia
Anemia

ACE, Angiotensin-converting enzyme; ARB, angiotensin II receptor blocker; CHF, congestive heart failure; CNI, calcineurin inhibitor; MI, myocardial infarction.
14 • Early Course of the Patient With a Kidney Transplant 209

closely in the first 24 hours postoperatively and daily until 4. Hoftman N, Prunean A, Dhillon A, et al. Revised Cardiac Risk Index
graft function is stable because this may be one of the earli- (RCRI) is a useful tool for evaluation of perioperative cardiac morbidity
in kidney transplant recipients. Transplantation 2013;96(7):639–43.
est signs of graft dysfunction and has been correlated with 5. Patel SI, Chakkera HA, Wennberg PW, et al. Peripheral arterial dis-
graft outcomes.94 Serum creatinine is also a useful param- ease preoperatively may predict graft failure and mortality in kidney
eter to monitor because it is a reliable method for detect- transplant recipients. Vasc Med 2017;22(3):225–30.
ing changes in kidney function, and it is recommended to 6. Kikic Z, Lorenz M, Sunder-Plassmann G, et al. Effect of hemodialysis
before transplant surgery on renal allograft function—a pair of ran-
monitor serum creatinine daily until discharge after trans- domized controlled trials. Transplantation 2009;88(12):1377–85.
plant and two to three times per week for the first month 7. Gandolfini I, Buzio C, Zanelli P, et al. The Kidney Donor Profile Index
posttransplant.95 Urine protein excretion should also be (KDPI) of marginal donors allocated by standardized pretransplant
measured at least once in the first month posttransplant donor biopsy assessment: distribution and association with graft out-
and then every 3 months for the first year. Proteinuria can comes. Am J Transplant 2014;14(11):2515–25.
8. Kidney Disease: Improving Global Outcomes (KDIGO) Transplant
be an indication of recurrent disease (such as FSGS) and is Work Group. KDIGO clinical practice guideline for the care of kidney
a risk factor for graft failure.76 Aberrancy in any of these transplant recipients. Am J Transplant 2009;9(Suppl. 3):S1–155.
parameters should potentiate a thorough workup for graft 9. Dawidson IJ, Sandor ZF, Coorpender L, et al. Intraoperative albumin
dysfunction and potential causes. administration affects the outcome of cadaver renal transplantation.
Transplantation 1992;53(4):774–82.
Evaluation of graft dysfunction should start with a careful 10. O’Malley CM, Frumento RJ, Hardy MA, et al. A randomized, double-
history to see if there is a potential for nephrotoxicity from blind comparison of lactate Ringer’s solution and 0.9% NaCl during
drugs or if there is any likelihood of volume contraction con- renal transplantation. Anesth Analg 2005;100(5):1518–24.
tributing to the elevation of serum creatinine levels. This 11. Dawidson I, Rooth P, Fry WR, et al. Prevention of acute cyclosporine-
evaluation should include measurements of therapeutic drug induced renal blood flow inhibition and improved immunosuppres-
sion with verapamil. Transplantation 1989;48(4):575–80.
levels, particularly for the CNIs. A vigorous search for poten- 12. Dawidson I, Rooth P, Alway C, et al. Verapamil prevents posttrans-
tial infection should follow, and if there is no obvious cause plant delayed function and cyclosporine A nephrotoxicity. Transplant
for deterioration in graft function, an ultrasound followed by Proc 1990;22(4):1379–80.
a renal biopsy should be performed. If there is any clinical 13. Cecka JM. The UNOS scientific renal transplant registry. In: Cecka JM,
Terasaki PI, editors. Clinical transplants. 1998. Los Angeles: UCLA
suspicion of renal artery or iliac artery stenosis, a magnetic Tissue Typing Laboratory; 1999:1–16.
resonance angiogram or arteriogram should be performed. 14. Oyen O, Strom EH, Midtvedt K, et al. Calcineurin inhibitor-free
We agree with the KDIGO indications for a kidney transplant immunosuppression in renal allograft recipients with thrombotic
biopsy, which are a persistently unexplained elevation in the microangiopathy/hemolytic uremic syndrome. Am J Transplant
serum creatinine, new-onset or unexplained proteinuria, 2006;6(2):412–8.
15. Wilson CH, Bhatti AA, Rix DA, et al. Routine intraoperative ureteric
and the failure of the serum creatinine to return to baseline stenting for kidney transplant recipients. Cochrane Database Syst Rev
values after treatment of acute rejection.8 The KDIGO guide- 2005;(4):CD004925.
lines also recommend a kidney allograft biopsy every 7 to 10 16. Fayek SA, Keenan J, Haririan A, et al. Ureteral stents are asso-
days during DGF to evaluate for evidence of acute rejection. ciated with reduced risk of ureteral complications after kidney
transplantation: a large single center experience. Transplantation
A biopsy can also help differentiate between CNI toxicity and 2012;93(3):304–8.
rejection. Nephrotic-range proteinuria in a patient whose 17. Ghasemian SM, Guleria AS, Khawand NY, et al. Diagnosis and man-
original disease was FSGS or thrombotic microangiopathy agement of the urologic complications of renal transplantation. Clin
should prompt an immediate biopsy for diagnosis and poten- Transplant 1996;10(2):218–23.
tial treatment with plasmapheresis. Prompt diagnosis and 18. del Pizzo JJ, Jacobs SC, Bartlett ST, et al. The use of bladder for total
transplant ureteral reconstruction. J Urol 1998;159(3):750–2.
management of the cause of early graft dysfunction are criti- discussion 2-3.
cal to the long-term success of the kidney transplant. 19. Kristo B, Phelan MW, Gritsch HA, et al. Treatment of renal transplant
ureterovesical anastomotic strictures using antegrade balloon dila-
tion with or without holmium:YAG laser endoureterotomy. Urology
2003;62(5):831–4.
Summary 20. Knechtle SJ. Ureteroneocystostomy for renal transplantation. J Am
Coll Surg 1999;188(6):707–9.
Optimization of outcomes after renal transplantation 21. Bruno S, Remuzzi G, Ruggenenti P. Transplant renal artery stenosis.
J Am Soc Nephrol 2004;15(1):134–41.
depends on the rapid diagnosis and treatment of surgical 22. Fervenza FC, Lafayette RA, Alfrey EJ, et al. Renal artery stenosis in
and medical complications. In view of the invasiveness of kidney transplants. Am J Kidney Dis 1998;31(1):142–8.
the transplant procedure itself, the complexity of medical 23. Audard V, Matignon M, Hemery F, et al. Risk factors and long-term
problems in this patient population, and the side effects of outcome of transplant renal artery stenosis in adult recipients after
nonspecific immunosuppressive therapy, close attention treatment by percutaneous transluminal angioplasty. Am J Trans-
plant 2006;6(1):95–9.
to the problems outlined in this chapter is crucial to avoid 24. Ngo AT, Markar SR, De Lijster MS, et al. A systematic review of out-
graft loss and patient death. Vigilance should be highest in comes following percutaneous transluminal angioplasty and stenting
the early posttransplant period, because this is when the in the treatment of transplant renal artery stenosis. Cardiovasc Inter-
frequency of complications is the greatest. vent Radiol 2015;38(6):1573–88.
25. Penny MJ, Nankivell BJ, Disney AP, et al. Renal graft thrombosis: a
survey of 134 consecutive cases. Transplantation 1994;58(5):565–9.
References 26. Bakir N, Sluiter WJ, Ploeg RJ, et al. Primary renal graft thrombosis.
1. Kasiske BL, Guijarro C, Massy ZA, et al. Cardiovascular disease after Nephrol Dial Transplant 1996;11(1):140–7.
renal transplantation. J Am Soc Nephrol 1996;7(1):158–65. 27. Singh A, Stablein D, Tejani A. Risk factors for vascular thrombosis in
2. Lentine KL, Schnitzler MA, Abbott KC, et al. De novo congestive heart pediatric renal transplantation: a special report of the North Ameri-
failure after kidney transplantation: a common condition with poor can Pediatric Renal Transplant Cooperative Study. Transplantation
prognostic implications. Am J Kidney Dis 2005;46(4):720–33. 1997;63(9):1263–7.
3. National Kidney Foundation. K/DOQI clinical practice guidelines for 28. Rouviere O, Berger P, Beziat C, et al. Acute thrombosis of renal trans-
managing dyslipidemias in chronic kidney disease. Am J Kidney Dis plant artery: graft slavage by means of intra-arterial fibrinolysis.
2003;41(Suppl. 3):S1–91. Transplantation 2002;73(3):403–9.
210 Kidney Transplantation: Principles and Practice

29. El Zorkany K, Bridson JM, Sharma A, et al. Transplant renal vein 56. Yarlagadda SG, Coca SG, Formica Jr RN, et al. Association
thrombosis. Exp Clin Transplant 2017;15(2):123–9. between delayed graft function and allograft and patient survival:
30. Luna E, Cerezo I, Collado G, et al. Vascular thrombosis after kidney a systematic review and meta-analysis. Nephrol Dial Transplant
transplantation: predisposing factors and risk index. Transplant Proc 2009;24(3):1039–47.
2010;42(8):2928–30. 57. Siedlecki A, Irish W, Brennan DC. Delayed graft function in the kidney
31. Harraz AM, Shokeir AA, Soliman SA, et al. Salvage of grafts with vas- transplant. Am J Transplant 2011;11(11):2279–96.
cular thrombosis during live donor renal allotransplantation: a criti- 58. Matsuoka L, Almeda JL, Mateo R. Pulsatile perfusion of kidney
cal analysis of successful outcome. Int J Urol 2014;21(10):999–1004. allografts. Curr Opin Organ Transplant 2009;14(4):365–9.
32. Okidi OO, Van Dellen D, Sobajo C, et al. Kidney transplant recipients 59. Yarlagadda SG, Coca SG, Formica Jr RN, et al. Association
requiring critical care admission within one year of transplant. Exp between delayed graft function and allograft and patient survival:
Clin Transplant 2017;15(1):40–6. a systematic review and meta-analysis. Nephrol Dial Transplant
33. Dempsey CM, Lim MS, Stacey SG. A prospective audit of blood 2009;24(3):1039–47.
loss and blood transfusion in patients undergoing coronary artery 60. Ortiz J, Parsikia A, Mumtaz K, et al. Early allograft biopsies performed
bypass grafting after clopidogrel and aspirin therapy. Crit Care Resus during delayed graft function may not be necessary under thymoglob-
2004;6(4):248–52. ulin induction. Exp Clin Transplant 2012;10(3):232–8.
34. European Renal Best Practice Transplantation Guideline Development 61. Humar A, Johnson EM, Payne WD, et al. Effect of initial slow graft
Group. ERBP guideline on the management and evaluation of the kid- function on renal allograft rejection and survival. Clin Transplant
ney donor and recipient. Nephrol Dial Transplant 2013;28(Suppl. 2): 1997;11(6):623–7.
ii1–71. 62. Jacobson PA, Schladt D, Israni A, et al. Genetic and clinical deter-
35. Taylor CJ, Kosmoliaptsis V, Sharples LD, et al. Ten-year experience of minants of early, acute calcineurin inhibitor-related nephrotoxic-
selective omission of the pretransplant crossmatch test in deceased ity: results from a kidney transplant consortium. Transplantation
donor kidney transplantation. Transplantation 2010;89(2):185–93. 2012;93(6):624–31.
36. Claas FH, Rahmel A, Doxiadis II . Enhanced kidney allocation to highly 63. Solez K, Vincenti F, Filo RS. Histopathologic findings from 2-year
sensitized patients by the acceptable mismatch program. Transplanta- protocol biopsies from a U.S. multicenter kidney transplant trial com-
tion 2009;88(4):447–52. paring tacrolimus versus cyclosporine: a report of the FK506 Kidney
37. Pereira M, Guerra J, Goncalves J, et al. Hyperacute rejection in a kid- Transplant Study Group. Transplantation 1998;66(12):1736–40.
ney transplant with negative crossmatch: a case report. Transplant 64. Naesens M, Kuypers DR, Sarwal M. Calcineurin inhibitor nephrotox-
Proc 2016;48(7):2384–6. icity. Clin J Am Soc Nephrol 2009;4(2):481–508.
38. Racusen LC, Haas M. Antibody-mediated rejection in renal allografts: 65. Kambham N, Nagarajan S, Shah S, et al. A novel, semiquantitative,
lessons from pathology. Clin J Am Soc Nephrol 2006;1(3):415–20. clinically correlated calcineurin inhibitor toxicity score for renal
39. Hanto DW, Brunt EM, Goss JA, et al. Accelerated acute rejection of an allograft biopsies. Clin J Am Soc Nephrol 2007;2(1):135–42.
A2 renal allograft in an O recipient: association with an increase in 66. Vella JP, Sayegh MH. Interactions between cyclosporine and newer
anti-A2 antibodies. Transplantation 1993;56(6):1580–3. antidepressant medications. Am J Kidney Dis 1998;31(2):320–3.
40. Sethi S, Choi J, Toyoda M, et al. Desensitization: overcoming the 67. Artero ML, Sharma R, Savin VJ, et al. Plasmapheresis reduces protein-
immunologic barriers to transplantation. J Immunol Res 2017;2017: uria and serum capacity to injure glomeruli in patients with recurrent
6804678. focal glomerulosclerosis. Am J Kidney Dis 1994;23(4):574–81.
41. Lefaucheur C, Nochy D, Hill GS, et al. Determinants of poor graft out- 68. Choy BY, Chan TM, Lai KN. Recurrent glomerulonephritis after kid-
come in patients with antibody-mediated acute rejection. Am J Trans- ney transplantation. Am J Transplant 2006;6(11):2535–42.
plant 2007;7(4):832–41. 69. Savin VJ, Sharma R, Sharma M, et al. Circulating factor associated
42. Haas M, Sis B, Racusen LC, et al. Banff 2013 meeting report: inclusion with increased glomerular permeability to albumin in recurrent
of C4d-negative antibody-mediated rejection and antibody-associated focal segmental glomerulosclerosis. N Engl J Med 1996;334(14):
arterial lesions. Am J Transplant 2014;14(2):272–83. 878–83.
43. Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Rates and determinants of 70. Vinai M, Waber P, Seikaly MG. Recurrence of focal segmental glomer-
progression to graft failure in kidney allograft recipients with de novo ulosclerosis in renal allograft: an in-depth review. Pediatr Transplant
donor-specific antibody. Am J Transplant 2015;15(11):2921–30. 2010;14(3):314–25.
44. Jordan SC, Vo AA, Tyan D, et al. Current approaches to treatment of 71. Chang JW, Pardo V, Sageshima J, et al. Podocyte foot process efface-
antibody-mediated rejection. Pediatr Transplant 2005;9(3):408–15. ment in postreperfusion allograft biopsies correlates with early
45. Valenzuela NM, Reed EF. Antibody-mediated rejection across solid recurrence of proteinuria in focal segmental glomerulosclerosis.
organ transplants: manifestations, mechanisms, and therapies. J Clin Transplantation 2012;93(12):1238–44.
Invest 2017;127(7):2492–504. 72. Chiurchiu C, Ruggenenti P, Remuzzi G. Thrombotic microangiopathy
46. Turgeon NA, Kirk AD, Iwakoshi NN. Differential effects of donor- in renal transplantation. Ann Transplant 2002;7(1):28–33.
specific alloantibody. Transplant Rev (Orlando) 2009;23(1): 73. Noris M, Remuzzi G. Thrombotic microangiopathy after kidney trans-
25–33. plantation. Am J Transplant 2010;10(7):1517–23.
47. Bloom RD, Bromberg JS, Poggio ED, et al. Cell-free DNA and active 74. Loirat C, Fremeauz-Bacchi V. Hemolytic uremic syndrome recurrence
rejection in kidney allografts. J Am Soc Nephrol 2017;28(7):2221–32. after renal transplantation. Pediatr Transplant 2008;12(6):619–29.
48. Garg N, Samaniego MD, Clark D, et al. Defining the phenotype of 75. Nester C, Stewart Z, Myers D, et al. Pre-emptive eculizumab and plas-
antibody-mediated rejection in kidney transplantation: advances in mapheresis for renal transplant in atypical hemolytic uremic syn-
diagnosis of antibody injury. Transplant Rev 2017;31(4):257–67. drome. Clin J Am Soc Nephrol 2011;6(6):1488–94.
49. Kim SC, Page EK, Knechtle SJ. Urine proteomics in kidney transplan- 76. Bataille S, Moal V, Gaudart J, et al. Cytomegalovirus risk factors in
tation. Transplant Rev 2014;28(1):15–20. renal transplantation with modern immunosuppression. Transpl
50. Sis B, Mengel M, Haas M, et al. Banff '09 meeting report: antibody Infect Dis 2010;12(6):480–8.
mediated graft deterioration and implementation of Banff working 77. De Keyzer K, Van Laecke S, Peeters P, et al. Human cytomegalovirus
groups. Am J Transplant 2010;10(3):464–71. and kidney transplantation: a clinician’s update. Am J Kidney Dis
51. Morris PJ, Russell NK. Alemtuzumab (Campath-1H): a sys- 2011;58(1):118–26.
tematic review in organ transplantation. Transplantation 78. Asberg A, Humar A, Rollag H, et al. Oral valganciclovir is noninfe-
2006;81(10):1361–7. rior to intravenous ganciclovir for the treatment of cytomegalovi-
52. Webster AC, Wu S, Tallapragada K, et al. Polyclonal and monoclonal rus disease in solid organ transplant recipients. Am J Transplant
antibodies for treating acute rejection episodes in kidney transplant 2007;7(9):2106–13.
recipients. Cochrane Database Syst Rev 2017;20(7):CD004756. 79. Bassil N, Rostaing L, Mengelle C, et al. Prospective monitoring of
53. Nankivell BJ, Alexander SI. Rejection of the kidney allograft. N Engl J cytomegalovirus, Epstein-Barr virus, BK virus, and JC virus infections
Med 2010;363(15):1451–62. on belatacept therapy after a kidney transplant. Exp Clin Transplant
54. Beimler J, Zeier M. Borderline rejection after renal transplantation—to 2014;12(3):212–9.
treat or not to treat. Clin Transplant 2009;23(Suppl. 21):19–25. 80. Munoz P, AST Infectious Diseases Community of Practice. Multiply
55. Moore J, Shabir S, Chand S, et al. Assessing and comparing rival defi- resistant gram-positive bacteria: vancomycin-resistant enterococcus
nitions of delayed renal allograft function for predicting subsequent in solid organ transplant recipients. Am J Transplant 2009;9(Suppl. 4):
graft failure. Transplantation 2010;90(10):1113–6. S50–56.
14 • Early Course of the Patient With a Kidney Transplant 211

81. Nampoory MR, Khan ZU, Johny KV, et al. Invasive fungal infections in 89. Goodwin JE, Zhang J, Geller DS. A critical role for vascular smooth
renal transplant recipients. J Infect 1996;33(2):95–101. muscle in acute glucocorticoid-induced hypertension. J Am Soc
82. Linares L, Cervera C, Cofan F, et al. Risk factors for infection with Nephrol 2008;19(7):1291–9.
extended-spectrum and AmpC beta-lactamase-producing Gram-nega- 90. Young JB, Neumayer HH, Gordon RD. Pretransplant cardiovascular
tive rods in renal transplantation. Am J Transplant 2008;8(5):1000–5. evaluation and posttransplant cardiovascular risk. Kidney Int Suppl
83. Paterson DL, Singh N. Interactions between tacrolimus and antimi- 2010;118:S1–7.
crobial agents. Clin Inf Dis 1997;25(6):1430–40. 91. Kasike BL, Anjum S, Shah R, et al. Hypertension after kidney trans-
84. Boutros M, Al-Shaibi M, Chan G, et al. Clostridium difficile colitis: plantation. Am J Kidney Dis 2004;43(6):1071–81.
increasing incidence, risk factors, and outcomes in solid organ trans- 92. Opelz G, Dohler B, Collaborative Transplant Study. Improved long-
plant recipients. Transplantation 2012;93(10):1051–7. term outcomes after renal transplantation associated with blood pres-
85. Neofytos D, Kobayashi K, Alonso CD, et al. Epidemiology, risk factors, sure control. Am J Transplant 2005;5(11):2725–31.
and outcomes of Clostridium difficile infection in kidney transplant 93. Tutone VK, Mark PB, Stewart GA, et al. Hypertension, antihyperten-
recipients. Transpl Infect Dis 2013;15(2):134–41. sive agents and outcomes following renal transplantation. Clin Trans-
86. Surawicz CM, Alexander J. Treatment of refractory and recurrent Clostrid- plant 2005;19(2):181–92.
ium difficile infection. Nat Rev Gastroenterol Hepatol 2011;8(6):330–9. 94. Lai Q, Pretagostini R, Poli L, et al. Early urine output predicts graft
87. Wadei HM, Textor SC. Hypertension in the kidney transplant recipient. survival after kidney transplantation. Transplant Proc 2010;42(4):
Transplant Rev (Orlando) 2010;24(3):105–20. 1090–2.
88. Klein IH, Abrahams AC, van Ede T, et al. Differential effects of acute 95. Perrone RD, Madias NE, Levey AS. Serum creatinine as an index
and sustained cyclosporine and tacrolimus on sympathetic nerve of renal function: new insights into old concepts. Clin Chem
activity. J Hypertens 2010;28(9):1928–34. 1992;38(10):1933–53.
15 Azathioprine and
Mycophenolates
ROBERT S. GASTON, GAURAV AGARWAL, and Sir PETER J. MORRIS

CHAPTER OUTLINE Introduction Infections


History Neoplastic Diseases
Azathioprine Pregnancy and Reproduction
Mycophenolates (Mycophenolic Acid, MPA) Early Clinical Trials of Mycophenolates in
Mechanism of Action Kidney Transplantation
Clinical Pharmacology and Blood Combination Therapy Utilizing MMF or
Monitoring Azathioprine in Kidney Transplantation
Azathioprine Cyclosporines
Dosing, Metabolism, and Monitoring Tacrolimus
Side Effects mTOR Inhibitors
Mycophenolates Belatacept
Dosing Use of Mycophenolates to Minimize or
Absorption Avoid Other Immunosuppressants
Metabolism and Clearance Calcineurin Inhibitor Minimization
Assays and Blood Monitoring CNI Withdrawal
Drug–Drug Interactions CNI Avoidance
Clinical Toxicities of Mycophenolates Corticosteroid Avoidance or Withdrawal
Gastrointestinal Adverse Reactions Therapeutic Drug Monitoring
Myelosuppression

Introduction History
Whereas calcineurin inhibitors (cyclosporine and tacrolimus) AZATHIOPRINE
block early signaling events and cytokine production, both
azathioprine and mycophenolates, often termed antiprolifer- 6-Mercaptopurine was developed by Elion and Hitchings at
atives, exert effects downstream in the cell cycle, interfering Burroughs Wellcome as an anticancer agent in the 1950s.3,4
with cytokine-dependent signals and lymphocyte prolifera- Subsequently, 6-mercaptopurine was shown to be immuno-
tion.1,2 Azathioprine, usually in combination with cortico- suppressive by Schwartz and Dameshek5,6; it attenuated the
steroids, was the backbone of immunosuppression in renal humoral response to a foreign protein and prolonged sur-
transplantation from the early 1960s to the early 1980s, vival of skin allografts in rabbits, termed drug-induced immu-
after which cyclosporine (CsA) was added to the mix. “Triple” nologic tolerance. This work was noted by Calne in the UK
therapy, with CsA, azathioprine, and steroids then became and Hume in the US; independently, these investigators found
standard in most centers through the mid-1990s. Myco- that 6-mercaptopurine delayed or prevented rejection of renal
phenolate mofetil (MMF) was developed in the early 1990s allografts in dogs.7,8 In Calne’s laboratory, although only two
as an alternative to azathioprine, supplanting it as standard dogs survived the kidney transplant operation, at death a little
therapy (in combination with calcineurin inhibitors and ste- more than a month later, there was no histologic evidence of
roids) by the end of the decade in most developed countries. rejection, a unique finding at the time. Almost simultane-
At the time of this writing, MMF is the most widely prescribed ously, similar results in a much larger series of canine trans-
transplant immunosuppressant in the world, although some plants emerged from Hume’s unit. Soon thereafter, Elion and
would maintain its superiority compared with azathioprine is colleagues4,9 produced azathioprine, an imidazolyl derivative
modest. In addition, because azathioprine is less expensive, it of 6-mercaptopurine that appeared somewhat less toxic than
will continue to have a role in transplantation, in particular, 6-mercaptopurine.10 Azathioprine was first used in humans
in developing countries where cost is a greater determinant in Boston at the Peter Bent Brigham Hospital in 1961 and 11,12
of immunosuppressive protocols. Thus both remain relevant then subsequently utilized (at doses of 2.5–3 mg/kg/day) in a
to clinical practice in transplantation. rapidly increasing number of kidney transplant units globally.
212
15 • Azathioprine and Mycophenolates 213

In those early days, rejection seemed inevitable, and to be difficult to administer to dogs and primates, with sub-
almost every patient experienced at least one episode. At stantial gastrointestinal toxicity, especially at higher doses
first, corticosteroids were used to treat rejection in patients (>20 mg/kg).20,21 Optimal kidney graft outcomes in dogs
on azathioprine;13 later they were added to azathioprine- occurred when RS61443 was combined with low-dose CsA
based prophylaxis by Starzl at the time of transplantation.14 and prednisone. MMF monotherapy, even at substantially
In this so-called azathioprine era, arbitrarily large doses of higher doses, demonstrated only limited efficacy. The ini-
steroids were administered from the time of transplanta- tial human experience was in rheumatoid arthritis, where
tion, with a gradual reduction over 6 to 12 months to main- a dose of 2 g/day was associated with “significant clinical
tenance levels. These high doses of steroids may have been improvement” among refractory patients.22 Side effects
primarily responsible for significant morbidity (discussed in were minimal and primarily of gastrointestinal origin (nau-
Chapter 16); only in the 1970s, after a series of randomized sea, vomiting, abdominal pain, diarrhea). No other signifi-
trials and clinical observations documenting the efficacy cant toxicities (including bone marrow suppression) were
of lower doses in preventing rejection, was a major reduc- noted.
tion in steroid-related complications noted (see Chapter In 1988, it was elected to proceed with clinical trials in
16). By the late 1970s, azathioprine and low-dose steroids, kidney transplantation. The initial study was a phase I/II
sometimes used together with an antilymphocyte serum or trial involving 48 kidney recipients at two centers.23 All
globulin for induction (particularly in North America), had the patients received polyclonal antibody induction, CsA,
become standard immunosuppressive therapy. and prednisone. Eight dosing groups of six subjects each
After CsA emerged in the early 1980s, azathioprine received MMF in daily doses ranging from 100 to 3500 mg.
was deemed obsolete. However, it quickly reemerged, uti- The greatest efficacy in preventing acute rejection with an
lizing lower doses (1.5–2 mg/kg/day) as adjunct therapy acceptable adverse event profile was seen in the 2000 and
to enable administration of lower, presumably less-toxic, 3000 mg dosing groups, providing the basis for the design
doses of CsA. For more than a decade, triple therapy with of three large registration trials.
CsA (4–8 mg/kg/day), azathioprine, and low-dose corti- The phase III MMF trials, commencing globally in 1992,
costeroids was considered standard immunosuppressive were the first of their kind: rigorously conducted studies
therapy for kidney recipients in most units in the developed involving large numbers of patients in solid-organ trans-
world. During this era, acute rejection rates fell from 80% plantation. Each study included approximately 500 sub-
or greater to around 50%, with some improvement in graft jects assigned to one of three treatment arms. Two of the
survival and patient survival, and substantially less mor- groups in each study received MMF, either 2 or 3 g/day, in
bidity than in preceding decades. It was the search for even combination with CsA and corticosteroids. In the US trial,
better immunosuppressants in the late 1980s that led to the all the patients underwent polyclonal antibody induction,
introduction of mycophenolates. with the comparator group receiving azathioprine instead
of MMF.24 In the Tricontinental trial, treatment arms were
MYCOPHENOLATES (MYCOPHENOLIC ACID, identical, with an azathioprine control group, but without
antibody induction.25 In the European trial, there was no
MPA)
antibody induction, and comparator patients received pla-
MPA is a fermentation product of Penicillium brevicompac- cebo instead of azathioprine.26 Findings of all three trials
tum and related fungi. It was first isolated by Gosio in 1896, were remarkably similar: a 50% reduction in rates of acute
named in 1913 by Alsberg and Black, and subsequently rejection with MMF (to less than 20% overall) within the
found to have weak antibacterial and antifungal activ- first 6 months after transplantation, with identical graft
ity.15 Raistrick and colleagues published the structure of and patient survival among treatment arms at 1 and 3
MPA in 1952. In the 1960s, it was found to have a strong years (Fig. 15.1) For most patients, the 2-g dose appeared
antimitotic effect in mammalian cells and was regarded as to offer optimal efficacy with fewer adverse effects. Based
a potential antitumor agent. Subsequent work by Franklin on these findings, on May 3, 1995, the US Food and Drug
and Cook documented its major mechanism of action (inhi- Administration (FDA) approved MMF (CellCept) 2 to 3 g/
bition of the do novo pathway of purine synthesis).15 At day for use in combination with CsA and corticosteroids,
about the same time, a Japanese group documented MPA’s with global availability soon to follow.27 A starting dose of
immunosuppressive properties, which were initially viewed 2 g/day became widely accepted in adults, and, by 2002,
as an impediment to clinical utility.16 MMF was the most widely prescribed immunosuppressant
As noted earlier, discovery and development of CsA in the US.28
changed the landscape for transplant immunosuppres- In ensuing years, there have been at least three other
sion. In the search for new agents targeting other path- landmark developments affecting the use of mycopheno-
ways, Allison and Eugui, among others, working first in the lates in transplantation. First, to address the gastrointes-
UK and subsequently at Syntex Pharmaceuticals, began tinal adverse events that plague some patients, Novartis
examining in vitro the effect of MPA on lymphocyte func- developed an enteric-coated formulation, mycophenolate
tion, documenting inhibition of cytotoxic T cell generation, sodium (EC-MPS, Myfortic).29 Designed to delay MPA release
antibody formation, lymphocyte adhesion, and the mixed and absorption in the gut (with the assumption that gastric
lymphocyte reaction.2,17,18 Scientists at Syntex, via mor- and early small-bowel exposure were major determinants
pholinoethyl esterification of MPA, produced a prodrug of gastrointestinal toxicity), clinical trials documented
(RS61443, mycophenolate mofetil, or MMF) with substan- EC-MPS efficacy equivalent to MMF, with trends to fewer
tially improved oral bioavailability in humans and other adverse gastrointestinal events.30,31 EC-MPS became avail-
mammals.19 Initial animal studies demonstrated the agent able in 2004. Second, in 2008, patent protection for the
214 Kidney Transplantation: Principles and Practice

70
PLA/AZA (n = 498)

Cumulative incidence (%)


60 MMF 1.0 g BID (n = 505)
MMF 1.5 g BID (n = 490)
50

40
30

20
10

0
0 1 2 3 4 5 6 7 8 9 10 11 12
Months from transplant to event
Fig. 15.1 Cumulative incidence of first biopsy-proven rejection in first year (excludes 1-year protocol biopsies; Kaplan–Meier estimates). The separation
between the curves occur in the first 2 months and is sustained to the end of the observation period. AZA azathioprine; MMF, mycophenolate mofetil: PLA,
placebo. (Data from the pooled efficacy analysis of the three double-blind clinical studies in prevention of rejection in Halloran P, Mathew T, Tomlanovich S,
et al. Mycophenolate mofetil in renal allograft recipients: a pooled efficacy analysis of three randomized, double-blind, clinical studies in prevention of rejec-
tion. The International Mycophenolate Mofetil Renal Transplant Study Groups. Transplantation 1997;63(1):39–47.)

De novo pathway

Ribose-5P + ATP
RNA RNA
PRPP synthetase
PRPP
Guanosine Adenosine
Salvage pathway
DP/TP TP
HGPRTase
Adenine deaminase
(Lesch-Nyhan) Guanosine IMPDH Inosine
Guanine Adenosine
MP MP MP
Ribonucleotide Ribonucleotide
reductase reductase
Deoxyguanosine Deoxyadenosine
DP/TP DP/TP

DNA MPA DNA

Fig. 15.2 Purine synthesis pathways showing site of inhibition by mycophenolate acid (MPA). ATP, adenosine triphosphate; DP, diphosphate; HGPRTase,
hypoxanthine-guanine phosphoribosyl transferase; IMPDH, inosine monophosphate dehydrogenase; MP, monophosphate; PRPP, phosphoribosyl
pyrophosphate; TP, triphosphate. (Adapted from Budde K, Glander P, Bauer S. Pharmacokinetics and pharmacodynamics of mycophenolate acid: a CME
monograph. Berlin: Walter de Gruyter; 2004:2-13.)

innovator formulation of MMF (CellCept) began expiring in imidazolyl derivative of 6-mercaptopurine. It is transformed
the US and globally.32 The clinical effect of the availability to 6-mercaptopurine in the liver, in turn, converted to thio-
of multiple MMF preparations on patients and the market- inosinic acid and 6-thioguanine. These latter metabolites
place remains controversial.33,34 Finally, in 2009, Tacroli- are incorporated into DNA and RNA, inhibiting lympho-
mus/MMF became a FDA-approved combination in kidney cyte proliferation by suppressing de novo purine synthesis.
transplantation. Previously, use of MMF or EC-MPS with Given the documented immunosuppressive effect of
tacrolimus, the most common immunosuppressant com- azathioprine, a relatively nonspecific inhibitor of nucleo-
bination in the world, had been “off-label,” limiting its use tide synthesis, MPA use in transplantation was based on
as a comparator protocol in drug development.35 This label the observation that inherited deletions in nucleic acid
change altered the landscape for new drug development synthesis resulted in immunodeficiencies: children lack-
and provided guidance for MPA dosing in tacrolimus-based ing adenosine deaminase show combined T cell and B cell
protocols. deficits.36 In contrast, subjects with absence of hypoxan-
thine-guanine phosphoribosyl transferase display essen-
tially normal immune function,17 indicating the purine
Mechanism of Action salvage pathway as relatively unimportant in lymphocytes
(Fig. 15.2). MPA acts as a reversible, noncompetitive inhibi-
Although both azathioprine and mycophenolate are consid- tor of inosine 5′-monophosphate dehydrogenase (IMPDH),
ered antiproliferative agents, their effects result from differ- the rate-limiting enzyme in the de novo synthesis of gua-
ing mechanisms of action. Azathioprine is a thiopurine, an nine nucleotides.2 This effect arrests new DNA synthesis in
15 • Azathioprine and Mycophenolates 215

proliferating cells at the G1/S interface, with guanosine tri- the face of evidence of marrow suppression. Analysis of data
phosphate (GTP) decreasing to 10% of levels measured in from the Collaborative Transplant Study suggested that, for
unstimulated T cells.1 Addition of guanosine or deoxyguano- patients receiving azathioprine and steroids only, long-term
sine reverses the inhibition, confirming the IMPDH target. T doses greater than 1.5 mg/kg/day were associated with
and B lymphocytes are highly dependent on this pathway for superior graft survival.51 When azathioprine is adminis-
proliferation in response to stimulation. There are two iso- tered with a calcineurin inhibitor (CNI) and steroids (triple
forms of IMPDH, with type I expressed in many cell lines, but therapy), lower doses are appropriate. A fairly standard
type II the predominant isoform in activated T and B lympho- dose of azathioprine in a triple protocol is 1.5 mg/kg, 100
cytes; IMPDH type II is four times more sensitive to MPA.37 mg/day. At this level, hematologic toxicity is uncommon.
These two factors impart some degree of lymphocyte-specific Xanthine oxidase has an important role in the catabo-
selectivity to the antiproliferative activity of MPA. Although lism of 6-mercaptopurine, and, therefore, azathioprine.
monocyte and dendritic cell replication may also be affected Concomitant use of xanthine oxidase inhibitors (e.g., allo-
by MPA, there is relatively little effect on neutrophils.1,38 purinol) requires significant azathioprine dose reduction to
The action of MPA at the G1/S interface is also selective. avoid excessive toxicities, particularly in the bone marrow.9
Neither the production of interleukin-2 nor the expression of Although the metabolites are excreted in the urine, these
its receptor is affected, showing a lack of influence on signal are inactive, and no reduction in dosage is required in acute
1 of lymphocyte activation. The primary antiproliferative kidney failure.52
effect also retards induction of cytotoxic T cells.2 Similarly, Regular monitoring of hematologic parameters is an
MPA mitigates primary and ongoing B cell responses, pre- important aspect of azathioprine therapy; common practice
sumably as a result of blockade of cell division, with subjects is to reduce the dose when the leukocyte count decreases
receiving MMF in the US pivotal trial demonstrating far less to less than 3 × 109/L. As already mentioned, if xanthine
production of xenoantibody toward equine antithymocyte oxidase inhibitors are used, the azathioprine dose should be
globulin (ATG) than those on azathioprine.39,40 MPA treat- reduced by at least 25% or consideration given to conver-
ment also blunts the synthesis of natural xenoantibodies sion to MPA. Blood levels of azathioprine or its metabolites
after plasma exchange and splenectomy in rats and inhibits are not routinely monitored in clinical practice. It has been
the humoral response to influenza vaccine in humans.41,42 noted, however, that the development of leukopenia can
Another distinct mechanism of MPA activity may relate also result from viral infection, leading to the suggestion that
to the requirement for GTP to activate fucose and man- erythrocyte 6-thioguanine nucleotide levels may be a better
nose transfer and glycoprotein synthesis, thus diminishing indicator of azathioprine activity in transplant patients.53
expression of adhesion molecules.1,18 When GTP is depleted A number of genetic variations in the thiopurine S-
as a consequence of MPA, adhesion of lymphocytes and methyltransferase (TMPT) gene have been identified, which
monocytes to activated endothelial cells is decreased in a have been related to azathioprine-induced toxicities.54,55
dose-dependent fashion. Polymorphisms in the TMPT enzyme, which catalyzes the
Cell types other than lymphocytes may also be sensitive S-methylation of 6-mercaptopurine and azathioprine, may
to MPA inhibition, with some models indicating vascular be associated with an increased likelihood of myelotoxicity
smooth muscle cells,43 mesangial cells,44 and myofibro- and leukopenia and require dose modification.54,55 Geno-
blasts45 to display dampened proliferation. In a subhuman typing for this polymorphism before commencing azathio-
primate model of orthotopic allograft vasculopathy, intra- prine might allow the appropriate azathioprine dose for an
vascular ultrasound documented dose-dependent inhi- individual to be determined,56 however, the cost/benefit
bition of the progression of intimal volume changes.20 ratio of this approach remains controversial.57
The efficacy of MMF was confirmed using aortic allograft
in another subhuman primate model46 and in a rodent Side Effects
chronic rejection system.47 The effects were potentiated The major complication of azathioprine therapy is bone
when MMF was administered with sirolimus, a combina- marrow suppression, most commonly manifest as leu-
tion that also reduced transforming growth factor-β1 and kopenia, although, in more severe settings, anemia and
its effects on extracellular matrix synthesis and degrada- thrombocytopenia may also occur. Megaloblastic ane-
tion.48,49 There may also be a more direct antifibrotic effect mia has been described in association with the use of aza-
of MPA mediated by upregulation of neutral endopeptidase thioprine.58 Although hepatotoxicity has been attributed
in the kidney.50 to azathioprine for many years; it is probably rarer than
thought, with many cases likely reflective of unrecognized
viral hepatitis. Hair loss is an uncommon complication.
Increased risk of nonmelanoma skin cancers and other
Clinical Pharmacology and Blood malignancies in transplant recipients has been attributed to
Monitoring azathioprine. Evolving experience, with both skin and colon
cancer, does indeed indicate azathioprine-treated patients
AZATHIOPRINE to be at greater risk, with a recent meta-analysis indicating
increased skin cancers like squamous cell, but not basal cell
Dosing, Metabolism, and Monitoring carcinoma.59–62 Mechanistically, Hofbauer and colleagues
Azathioprine is given as a single daily dose. If used with ste- have shown that mercaptopurine metabolites intercalated
roids alone, a suitable starting dose is 2.5 mg/kg/day, with into DNA of skin increase sensitivity to ultraviolet irradia-
careful monitoring of leukocyte counts. The dose of azathio- tion, providing a plausible mechanistic explanation for the
prine may be reduced somewhat over time, particularly in clinical observations.63
216 Kidney Transplantation: Principles and Practice

OH CH3
O
O
MMF N
O
O O
OCH3
CH3

OH CH3
O
MPA COOH
O
OCH3
CH3

EHC
OH OH
OH CH2OH OH COOH

O O
OH OH
O CH3 O CH3 OH CH3 OH
O O O
O OH
COOH COCH
O O O
O O
OCH3 OCH3 OCH3 OH
CH3 CH3 CH3 COOH
7-O-Glucoside Acyl glucuronide
metabolite MPAG metabolite
Fig. 15.3 Metabolism of parent compound mycophenolate mofetil (MMF) to mycophenolic acid (MPA) by cleavage of mofetil group (encircled) with
primary metabolism to its glucuronide (MPAG), which may undergo enterohepatic recycling (EHC), and secondary acyl glucuronide and 7-O-glucoside
metabolites. (Adapted from Shaw LM, Korecka M, Venkataramanan R, et al. Mycophenolate acid pharmacodynamics and pharmacokinetics provide the basis
for rational monitoring strategies. Am J Transplant 2003;3:534.)

MYCOPHENOLATES for 24 hours (AUC0–24) is proportionate to dose in healthy


subjects,64 although a recent study of MMF kinetics in kid-
Dosing ney transplant recipients revealed a nonlinear relationship,
In adults with kidney transplants, MMF is most commonly with reduced bioavailability as doses increase from 250
administered orally at a dose of 1 g twice daily.27 In pedi- mg to 2000 mg.65 The magnitude of the nonlinearity was
atric patients, the recommended dose of MMF oral suspen- greater (176%–76%) in combination with tacrolimus than
sion is 600 mg/m2 at 12-hour intervals up to a maximum with CsA (123%–90%). A recent multicenter crossover
of 2 g daily. Intravenous MMF is administered at the same study comparing pharmacokinetics of two MMF prepara-
dose and frequency as the oral preparation, infused over 2 tions (Teva and Roche) in 43 stable kidney recipients found
hours. EC-MPS is administered orally at a dose of 720 mg them to be comparable in all parameters studied.66
twice daily in adults.29 This suggested dose of EC-MPS in a EC-MPS does not release MPA under acidic conditions
stable pediatric patient is 400 mg/m2 twice daily, but its use (pH <5) as in the stomach but is highly soluble in a neutral-
is not recommended in patients less than 5 years old. pH environment like the intestine.67 Consistent with this
property is a Tmax of 1.5–2.75 hours, substantially delayed
Absorption compared with MMF.29 Gastrointestinal absorption is 93%,
Orally delivered MMF is rapidly and almost completely and absolute bioavailability of MPA after administration
absorbed in the stomach and upper intestine and then effi- of EC-MPS is 72%. There is a substantial effect of food on
ciently hydrolyzed to MPA (Fig. 15.3). Drug absorption, as MPA absorption with EC-MPS, although overall AUC is not
assessed by area under the curve (AUC), is not significantly affected; to avoid variability, it should always be taken on
altered by coadministered food, although the maximal con- an empty stomach. Like MMF, EC-MPS pharmacokinetics
centration (Cmax) is 40% lower in simultaneously fed renal are dose-proportional over its administration range, and
transplant recipients.27 The time to maximal concentration its profile regarding metabolism and clearance (see later)
(Tmax) of less than 1 hour is independent of hepatic or renal is similar as well. An EC-MPS dose of 720 mg most closely
function. The Tmax is slightly delayed, however, in the period approximates the MPA exposure of 1000 mg of MMF.
immediately after transplantation (1.31 ± 0.76 hours) and
in diabetic patients (1.59 ± 0.67 hours). In contrast, by 3 Metabolism and Clearance
months it is 0.90 ± 0.24 hours. Metabolism of MMF to MPA MPA is rapidly metabolized to an inactive glucuronide
yields 94% bioavailability. Over a range of 100 to 3000 (MPAG) via one or more isoforms of the UGT1 gene fam-
mg/day, the MPA area under the concentration-time curve ily of uridine diphosphate–glucuronosyl transferases in the
15 • Azathioprine and Mycophenolates 217

Tacrolimus MPAG is the major urinary excretion product (93% of the


radioactive parent compound); urinary excretion of MPA is
negligible (1%). Fecal excretion accounts for 6%. Neither
100 hemodialysis nor peritoneal dialysis significantly affects
MPA plasma concentrations, although in multiple-dose
studies either dialysis method may remove some MPAG.
80 With renal dysfunction, there is a moderate increase in
Estimated MPA AUC (mg•h/L)

plasma MPA and a marked accumulation of MPAG. MPA


binds tightly and extensively (97%), but reversibly, to
60 serum albumin, decreasing its ability to inhibit IMPDH.71
The free fraction, constituting 1% to 3% of the total amount
in the blood of stable patients, is cleared by biliary and renal
40
routes.
Hypoalbuminemia is associated with increased free MPA
and greater MPA clearance. Renal allograft dysfunction
20
also increases the MPA-free fraction because it decreases
Intercept: 23.016 the binding of acidic drugs; in contrast, hepatic oxida-
Slope: 9.8287
R squared: 0.68636 tive impairment produces no effect. Estimates of free MPA
0
in ultrafiltrate samples have not been shown to offer any
0 1 2 3 4 5 6 7 advantage over measurements of total MPA to predict ther-
A MPA trough (mg/L) apeutic versus adverse reactions,69,72 except in the pres-
ence of impaired graft function.68
Cyclosporine
Assays and Blood Monitoring
The MPA parent compound is readily measured in plasma
100 by high-performance liquid chromatography.73 In con-
trast, the widely available automated enzyme multiplier
immunoassay technique (EMIT)74 yields 15% to 20%
80 higher results because its antibody reagent cross-reacts
Estimated MPA AUC (mg•h/L)

with the acyl-MPAG metabolite (see Fig. 15.3), which


may contribute to both immunosuppressive and toxic
60 effects.75 Because of these properties, some have sug-
gested the EMIT assay may be preferable and is the most
widely utilized.
40 The gold-standard pharmacokinetic measure of MPA
exposure is a frequent sampling of blood over a 12-hour
period, with the calculation of the AUC concentration-time
20 exposure. MPA AUC can be calculated from either full (8 or
Intercept: 30.921 more samples over 12 hours) or limited (2–5 samples over
Slope: 7.6355
R squared: 0.32874
4 hours) sampling strategies.76,77 Although the evidence
0 is conflicting regarding utility, 12-hour predose (C0) MPA
0 1 2 3 4 5 6 7
levels are convenient to obtain and have been utilized effec-
B tively in some studies.78,79 Correlation (r2) of the predose
MPA trough (mg/L)
level with AUC of MMF ranges from 0.32 to 0.68 and may
Fig. 15.4 Linear regression analysis of the estimated mycopheno- differ by concomitant CNI usage79,80 (Fig. 15.4). For EC-
lic acid (MPA) area under the concentration-time curve (AUC) versus
trough in patients in the Opticept study receiving (A) tacrolimus and (B)
MPS, variability in absorption means significantly poorer
cyclosporine. (From Gaston RS, Kaplan B, Shah T, et al. Fixed- or controlled- correlation between predose MPA level and AUC (r2 =
dose MMF with standard- or reduced-dose calcineurin inhibitors: the Opti- 0.02). Long-term, intrapatient variability of both AUC and
cept trial. Am J Transplant 2009;1613.) trough levels is relatively low.77
After the first few weeks after transplantation, dose-
gastrointestinal tract, liver, and possibly kidney. Two minor normalized MPA exposure increases by 30% to 50%, per-
metabolites also are formed: the acyl glucuronide and the haps reflecting a decline in drug clearance, MPA satura-
phenolic glucoside68 (see Fig. 15.3) tion of tissues, or other factors.81,82 MPA exposure/dose
The apparent elimination half-life of MPA in healthy vol- relationships may also differ in settings other than kidney
unteers is 17.9 hours – a clearance of 11.6 L/h.69 At 8 to transplantation (e.g., liver and small-bowel transplan-
12 hours after oral drug administration, 37% of patients tation, bone marrow transplantation).83,84 Typically,
(range 10%–61%) display a secondary peak in plasma MPA younger children require higher MMF doses per body mass
concentrations representing enterohepatic recirculation. index than older children or adults to achieve comparable
The additional peak results from excreted MPAG in bile MPA exposure.85
undergoing deglucuronidation by intestinal bacteria with Considerable interpatient pharmacokinetic variability
subsequent reabsorption of MPA, and may account for up of MMF has been documented to be due to differences in
to 40% to 60% of the total dose interval MPA AUC.64,70 hepatic/renal function,86 concurrent drug administration,
218 Kidney Transplantation: Principles and Practice

and the presence of diarrhea, but not to ethnicity or gen- and gastrointestinal side effects were reportedly “modest.”23
der.87,88 Pharmacodynamic monitoring of patients receiving In general, adverse events, including invasive cytomegalovi-
MPA therapy has been studied. Peripheral blood lympho- rus (CMV) infections, were more common in patients receiv-
cytes (PBL) from patients on MMF demonstrate reduced ing 3 g (vs. 2 g) of MMF daily, suggesting a dose-dependent
response to stimulation assays and diminished antibody relationship. In the pivotal trials, adverse events accounted
production.40,89 However, so many other factors affect the for study withdrawal in 15% of subjects in the MMF 3 g/day
proliferative activity of PBL in this setting that the clinical dose, 9% from MMF 2 g/day dose, and 5% from the compara-
utility of these assays is modest. IMPDH assays are techni- tor cohorts.100 Early on, these side effects were thought to
cally demanding and difficult to reproduce; in addition, the be related to the MMF dose rather than to the plasma con-
whole-blood matrix may not reflect the effect in activated centration of parent compound or its metabolites,82 and
lymphocytes, which are the cells of interest. Estimates of indeed adverse events often respond to MMF dose reduction.
IMPDH activity in isolated peripheral blood mononuclear However, whereas findings have been somewhat inconsis-
cells indicate considerable interindividual variability. The tent, more recent evidence has correlated gastrointestinal
time course of IMPDH inhibition, as measured by the produc- and hematologic adverse events to MPA exposure as well as
tion of xanthine monophosphate by isolated mononuclear dose.78,101 The frequency and severity of MMF-related adverse
cells, parallels the MPA plasma concentration.90 Patients events are also influenced by the other immunosuppressants
with lower IMPDH levels before transplantation, indicative of with which it is combined. The common clinical practice has
genetic susceptibility to the drug, more frequently underwent been to reduce MMF dosing in response to adverse events;
dosage reductions within 6 months; pretransplant IMPDH there is some evidence, however, that dose reduction can be
activity has been suggested as a guide to MPA dosing in pedi- associated with increased risk of rejection and graft failure, and
atric patients.91,92 It is unclear, however, whether levels of should be approached cautiously.102
fluctuations in IMPDH activity can be of utility in predicting
either efficacy or toxicity of MMF in clinical transplantation. GASTROINTESTINAL ADVERSE REACTIONS
Drug–Drug Interactions One or more gastrointestinal symptoms from a constella-
In general, important drug–drug interactions with myco- tion that includes diarrhea, indigestion, bloating, nausea,
phenolates are rare. Cholestyramine may reduce MPA vomiting, and abdominal pain, are the most commonly
absorption.27 Antibiotic therapy disrupting the gastroin- reported adverse effects of MMF therapy. In the European
testinal flora may interfere with deglucuronidation and trial, the overall incidence of any gastrointestinal complaint
enterohepatic recirculation, decreasing drug levels. Because was 53% and 46% for the MMF 3-g and 2-g doses, respec-
acyclovir and ganciclovir compete with MPAG for tubular tively, versus 42% in the placebo comparator arm.103 These
secretion, they may increase drug concentration slightly, adverse effects often improve with MMF dose reduction, or,
particularly among patients experiencing renal impair- in some cases, with the maintenance of the same total daily
ment. Coadministration of proton pump inhibitors has been dose administered more frequently in smaller increments
reported to reduce exposure to MPA by 25% to 35%, but (e.g., 500 mg four times daily rather than 1000 mg twice
the clinical effect of this interaction is uncertain.27 daily).
Clearly, however, the most significant drug–drug interac- The most common and troublesome gastrointestinal
tion is with CsA: its importance is magnified because most dos- adverse event is diarrhea, experienced by up to 35% of sub-
ing regimens of MMF and EC-MPS were established utilizing jects in the pivotal trials using CsA, almost twice the fre-
this combination. When either mycophenolate preparation is quency as in the comparator arms.100 Diarrhea occurs even
administered with CsA, subjects display lower MPA concen- more often, and with greater severity, when MMF is used
trations than those not receiving CsA or in whom the drug in combination with tacrolimus (45% vs. 25% with CsA in
was discontinued.93,94 This effect is likely the result of reduced one study).104 Because diarrhea also increases tacrolimus
enterohepatic recirculation.95 In contrast, coadministration absorption and blood levels, its clinical consequence may
with tacrolimus does not alter MPA exposure compared with be greater with this combination as well.105 In a registry
placebo, and MPA exposure in combination with sirolimus is analysis, Bunnapradist and colleagues found significantly
likewise higher than observed with CsA.96,94,97 There appears increased risk of diarrhea (hazard ratio [HR] 1.37) associ-
to be no drug–drug interaction between belatacept and MMF. ated with the tacrolimus/MMF combination, which, in
Similarly, the experimental Janus kinase inhibitor tofacitinib turn, was linked to increased risk of graft failure (HR 2.13)
does not affect MPA pharmacokinetics.72 This property resulted and patient death (HR 2.04).106 After renal transplanta-
in 37% greater MPA exposure in tofacitinib-treated (versus tion, however, diarrhea can be due to a variety of causes
CsA) subjects in a phase IIb trial, with a profound influence other than immunosuppressant therapy, including preex-
on the outcomes in kidney transplant recipients.98 Finally, the isting diabetic or uremic conditions, intercurrent infectious
corticosteroid complement of most regimens increases MPAG diseases, and concurrent antibiotic treatment.107 Maes and
slightly by inducing hepatic glucuronosyl transferase.99 colleagues reported 60% of 26 cases of diarrhea were due
to an infectious origin: CMV, Campylobacter, bacterial over-
growth, or, in another report, microsporidiosis.107,108 In
Clinical Toxicities of the other 40%, erosive enterocolitis was attributed to MMF,
Mycophenolates characterized by faster colonic transit, crypt distortion, and
focal inflammation, thought to represent a toxic action
The initial clinical trials of MMF indicated the absence of nep­ of the acyl-MPAG metabolite on absorptive cells, leading
hrotoxicity, neurotoxicity, or hepatotoxicity. Myelotoxicity to a predominance of goblet cells.109 In the presence of
15 • Azathioprine and Mycophenolates 219

persistent diarrhea, colonic biopsy specimens have shown decreased donor, and CMV seronegativity with a CMV-
apoptosis of intestinal gland epithelial cells110 and atrophy positive donor (and lengthier antiviral prophylaxis).113
of the intestinal villi, which has been documented to disap- MMF-related leukopenia may be associated with markedly
pear a few months after MMF withdrawal.111 abnormal neutrophil morphology.119 As with anemia,
EC-MPS was developed specifically to mitigate the gas- some, but not all, studies have shown MPA plasma concen-
trointestinal toxicities associated with MMF by delaying trations and particularly free MPA AUC0–12h to be signifi-
absorption in the gastrointestinal tract with the goal of cantly related to severe infections and leukopenia.120,121 The
reducing peak levels and delivering the active drug to more leukopenia has been associated with stomatitis, particularly
distal portions of the bowel. Compared with MMF, EC-MPS when combined with a sirolimus-based regimen.122
showed equivalent efficacy and overall equivalent MPA
drug exposure in kidney transplant patients, but very simi- INFECTIONS
lar gastrointestinal adverse events when administered in a
randomized fashion (with CsA and corticosteroids) to either In the pivotal trials of CsA with MMF, whereas viral infec-
de novo or stable recipients.30,31 Some conversion patients, tions (herpes simplex and zoster, and tissue-invasive CMV)
however, showed marked improvement in EC-MPS, and, in were more common in MMF-treated recipients than con-
another study of gastrointestinal intolerance of MMF, fewer trols, other opportunistic infections were not.24–26 More
dose changes of EC-MPS were required, with an apparently recently, several studies have indicated significantly fewer
reduced symptom burden, better functioning, and improved herpes viral infections (especially CMV) with mammalian
health-related quality of life.112 target of rapamycin (mTOR) inhibitors than with mycophe-
nolates.123 However, widespread use of antiviral prophy-
laxis is now the norm, and most centers now view CMV as a
MYELOSUPPRESSION
manageable problem.124
Dose-dependent myelosuppression has been observed with Recognition of BK virus infection and nephropathy coin-
MMF. This was particularly notable in the European trial, cided with the rapid assimilation of mycophenolates and
as subjects receiving 3-g or 2-g doses of MMF had more tacrolimus into clinical practice, and most attribute its rise
anemia or leukopenia (26% and 24%, respectively) than to more potent immunosuppression associated with these
placebo-treated controls (13%).24,26 However, in the other combinations.125 Aggressive screening of BK viruria or
pivotal trials, hematopoietic adverse events may have viremia, with immunosuppressant dose reduction (focused
occurred slightly less frequently with MMF than in azathio- on mycophenolates), has diminished the effect of BK
prine controls.24,25 As with gastrointestinal side effects, it nephropathy on risk of allograft failure.126 Progressive mul-
should be remembered that among kidney recipients there tifocal leukoencephalopathy (PML) is a rare but debilitat-
may be multiple causes of anemia or leukopenia, includ- ing central nervous system manifestation of polyomavirus
ing renal dysfunction and infection, as well as concomitant infection, and it has been associated with mycophenolate
immunosuppressants and other drugs. use. Registry analysis of PML in the US found all reported
A prospective study involving 978 patients at seven North cases in kidney recipients to be taking MMF, although MMF
American transplant centers documented anemia (hemoglo- use was so ubiquitous that no statistical association could
bin ≤10 g/dL) in 10% of MMF-treated subjects and an asso- be documented.127 Another analysis of reported cases of
ciation with recipient age, use of antiviral prophylaxis, and PML in the US found a strong association with not only
posttransplant dialysis.113 In the Fixed Dose-Concentration mycophenolates, but also azathioprine, CsA, cyclophos-
Controlled (FDCC) trial in Europe, anemia was reported in phamide, efalizumab, rituximab, and tacrolimus, among
38% of subjects.114 Greater MMF dose and MPA predose (C0) others, indicating a potential role for interactions among
plasma concentrations have been correlated with decreased components of combination therapy and overall intensity
hemoglobin values among stable renal transplant recipi- of immunosuppression.128
ents.115 Another study suggested an even better correlation Introduction of MMF therapy has been reported to pro-
of anemia with MPA metabolites, MPAG and acyl-MPAG duce a significant increase in hepatitis C virus viremia in
than with MPA itself.116 Conversely, others have found no some, but not all, patients receiving concomitant CsA.129
association.75,117 In a single-nucleotide polymorphism anal- In contrast, MMF seems to show no significant effect on
ysis of kidney recipients with MMF-related anemia, one study hepatitis B virus viremia despite in vitro studies that sug-
found a protective effect of the HUS1 allele, with interleukin- gested that it inhibited viral replication.130 Recent analysis
12A and CYP2C8A genes associated with an increased risk of outcomes in kidney recipients with hepatitis C indicated
for anemia.83 Other investigators, however, while confirm- no adverse effect of mycophenolate therapy on long-term
ing an association with CYP2C8A (but not the other geno- graft or patient survival.131
types) noted that expression of the “at-risk” phenotype is
so rare (0.5% of population) as not to explain the observed NEOPLASTIC DISEASES
frequency of anemia (or leukopenia).114 In the current era,
leukopenia (fewer than 3000 leukocyte/mm3) is reported in Early experience with MMF was associated with a numerical
14% to 23% of kidney transplant recipients.113,114 A mul- increase in subjects with lymphoma compared with placebo
tivariable analysis (Cox regression) of Medicare-covered or azathioprine control groups.100 However, large case-
kidney recipients found a significant association between control studies now indicate little or no additional adverse
mycophenolate use and leukopenia (adjusted HR 1.21).118 effect of mycophenolates on the risk of lymphoma or other
Other documented risk factors for leukopenia included corti- malignancy in patients receiving pharmacologic immuno-
costeroid-free regimens, weight, previous kidney transplant, suppression.132,133 Registry data also indicate malignancy
220 Kidney Transplantation: Principles and Practice

risk associated with MMF use to be similar to other immu- azathioprine (or placebo) in the prevention of early rejec-
nosuppressants.134 Despite an emerging consensus that tion episodes, (2) fewer withdrawals in MMF-treated sub-
mTOR inhibitors may be associated with reduced malig- jects due to treatment failure, and (3) more adverse effects
nancy risk, much of the purported benefit is in comparison in the 3-g than the 2-g MMF groups.
to CNIs rather than mycophenolates.135,136 Although none of the studies individually was powered to
test efficacy in terms of graft survival, a planned pooled anal-
ysis of 12-month data from the three studies, with a total of
PREGNANCY AND REPRODUCTION
1493 randomized subjects, confirmed the benefit of MMF
MMF demonstrated teratogenic effects at subclinical doses on acute rejection episodes: 20% and 17% in the two MMF
in animal studies,137 which generated caution regarding its arms versus 41% among controls, yielding a relative risk
use in both males and females of reproductive age. Avoid- (RR) ratio of 0.46.100 There was significantly less steroid-
ance of mycophenolates in males fathering children is no resistant rejection with MMF, and renal function at 1 year
longer recommended. However, fetal malformations have was substantially better (despite identical CsA exposure) in
been reported in the offspring of female kidney recipients the MMF-treated patients. Despite a trend indicating fewer
taking MMF.138 Mycophenolates are now recognized by graft losses over time with MMF, the combined incidence of
the US FDA as category D agents in pregnancy (positive evi- graft loss and death was 10%, 11%, and 12%, respectively,
dence of human fetal risk, but the benefits from use in preg- at 1 year (P = not significant). Longer-term follow-up failed
nant women may be acceptable despite risk).139 In a female to suggest a benefit of MMF versus azathioprine or placebo
recipient with stable kidney allograft function at least 1 year in the US or Tricontinental studies, although data from the
posttransplant who desires pregnancy, common practice is European trial revealed that the 2-g, but not that 3-g, dose
to convert from mycophenolate to azathioprine at least 6 of MMF reduced death-censored graft loss compared with
weeks before discontinuing contraception.139 placebo: 9%, 13%, and 16%, respectively (P = 0.03).103
After FDA approval in 1995, MMF was rapidly incorpo-
rated into clinical practice in the US. By 1999, over 80%
Early Clinical Trials of of the US kidney recipients were on MMF, usually in com-
bination with CsA, and now its use is ubiquitous.28 From
Mycophenolates in Kidney the beginning, pharmacoeconomic analyses documented
Transplantation benefit; the cost of adding MMF to a regimen was more
than offset by reduced costs associated with diagnosing and
As mycophenolate emerged as a potential immunosuppres- treating acute rejection and its consequences.142,143 Subse-
sant in the late 1980s, azathioprine was the antiprolifera- quent analyses of clinical outcomes of almost 50,000 kidney
tive agent of choice, most commonly utilized with CsA and recipients in the US Renal Data System (USRDS) suggested
corticosteroids as triple therapy. A phase I/II dose-finding that prescription of MMF yielded significantly better early
study of MMF in combination with polyclonal antibody and late patient and graft survivals. The risk of late acute
induction, CsA, and corticosteroids in de novo kidney recipi- rejection episodes was reduced by 65%,144 with the risk of
ents indicated the therapeutic benefit of MMF at doses of 2 to long-term deterioration of graft function reduced by 20%
3 g/day.23 There was also suggested efficacy at these doses, to 34%.144,145 More recently, Wagner and colleagues con-
demonstrated in the treatment settings of an ongoing or ducted a meta-analysis of MMF versus azathioprine.146 They
steroid-resistant acute renal rejection episode.140,141 Three included 23 studies with 3301 patients and demonstrated
blinded, randomized phase III trials were initiated in 1992. that MMF significantly reduced death-censored graft loss
Patients in each trial were assigned to one of three treat- (RR 0.78, 95% confidence interval [CI] 0.62–0.99, P < 0.05)
ment arms: a control group or one of two groups receiving (Fig. 15.5) and acute rejection (RR 0.65, 95% CI 0.57–0.73,
MMF 2 g or 3 g daily, respectively. All patients also received P < 0.01), (Fig. 15.6). Adverse events also had a slightly dif-
CsA and prednisone. The primary end-point of each study ferent profile with gastrointestinal symptoms being more
was the prevention of acute rejection episodes during the common with MMF and thrombocytopenia and elevated
first 6 months after transplantation, thought to be indica- liver enzymes with azathioprine.
tive of poorer 1-year and possibly long-term graft survival.
The European Mycophenolate Mofetil Study compared
MMF with placebo with no antibody induction therapy.26
At 6 months, 46% of those receiving placebo had experi- Combination Therapy Utilizing
enced biopsy-proven acute rejection (BPAR), versus only MMF or Azathioprine in Kidney
17% (2 g) and 14% (3 g) of those randomized to MMF. The
US Renal Transplant Study, which required polyclonal anti-
Transplantation
body induction for all patients and included an azathioprine CYCLOSPORINES
control arm, yielded corresponding acute rejection rates of
38%, 20%, and 18%, respectively.24 The Tricontinental Shortly after the introduction of mycophenolates, CsA
study, which included an azathioprine comparator but microemulsion (Neoral and generic, CsA-ME) began replac-
without antibody induction, demonstrated BPAR in 36%, ing its original oil-based formulation (Sandimmune). Data
20%, and 15% of subjects, respectively.25 Each study also reported with the newer CsA-ME/MMF combinations
found significantly fewer histologic and clinically severe generally confirmed the initial experience cited previ-
rejections among the MMF-treated cohorts. Thus each trial ously.147–149 The ELITE-Symphony trial, enrolling more
documented: (1) similar benefit of MMF compared with than 1600 subjects, included two CsA-ME cohorts, normal
15 • Azathioprine and Mycophenolates 221

Study or subgroup MMF AZA Risk Ratio Weight Risk Ratio


M- M-
H,Random,95% H,Random,95%
n/N n/N CI CI
4 Longest duration of follow-up
MMF US Study 1995 50/331 28/164 31.3 % 0.88 [ 0.58, 1.35 ]

MMF TRI Study 1996 39/335 25/162 25.8 % 0.75 [ 0.47, 1.20 ]

Ling 1998 0/11 0/5 Not estimable

Mendez 1998 1/117 3/59 1.1 % 0.17 [ 0.02, 1.58 ]

Egfjord 1999 1/25 5/25 1.3 % 0.20 [ 0.03, 1.59 ]

Suhail 2000 0/20 0/20 Not estimable

Johnson 2000 9/72 9/76 7.5 % 1.06 [ 0.44, 2.51 ]

Ji 2001 0/56 2/50 0.6 % 0.18 [ 0.01, 3.64 ]

Busque 2001 0/23 0/23 Not estimable

Folkmane 2001 2/23 3/25 1.9 % 0.72 [ 0.13, 3.96 ]

Weimer 2002 2/31 0/25 0.6 % 4.06 [ 0.20, 80.94 ]

Sadek 2002 16/162 16/157 13.0 % 0.97 [ 0.50, 1.87 ]

Miladipour 2002 0/40 1/40 0.6 % 0.33 [ 0.01, 7.95 ]

Tuncer 2002 5/38 11/38 6.1 % 0.45 [ 0.17, 1.18 ]

Merville 2004 0/37 5/34 0.7 % 0.08 [ 0.00, 1.46 ]

MYSS Study 2004 7/124 7/124 5.4 % 1.00 [ 0.36, 2.77 ]

Joh 2005 4/34 6/34 4.1 % 0.67 [ 0.21, 2.15 ]

Subtotal (95% CI) 1479 1061 100.0 % 0.78 [ 0.62, 0.99 ]

0.002 0.1 1 10 500


Less with MMF Less with AZA

Fig. 15.5 Forest plot shows the relative risk of graft loss: censored for death comparing mycophenolate mofetil versus azathioprine. Boxes represent
relative risk in individual studies; diamonds represent summary effects. Horizontal bars represent 95% confidence intervals (CI). AZA, azathioprine; MMF,
mycophenolate mofetil. (Adapted from Wagner M, Earley AK, Webster AC, et al. Mycophenolic acid versus azathioprine as primary immunosuppression for
kidney transplant recipients. Cochrane Database Syst Rev 2015(12):CD007746.)

and reduced-dose (with MMF) and daclizumab induction.150 (3% vs. 17% of subjects) using higher-than-standard doses
Rejection rates after 1 year approximated 25%, similar to of EC-MPS (2880 then 2160 mg/day) for 6 weeks.154
those observed previously. Alternatively, a multicenter African Americans are generally considered at greater
European study using CsA-ME demonstrated only modest immunologic risk than others, and a subgroup analysis of
insignificant reductions in acute rejection episodes with the US pivotal trial documented a benefit of MMF for black
MMF compared with azathioprine (34% and 35%, respec- recipients only in the 3-g dose group: an acute rejection rate
tively, after 1 year) and questioned the value of a 10- to of 12% compared with 32% in the 2-g group and 48% in
15-fold cost differential.151 Five-year follow-up likewise doc- the azathioprine cohort.155 These findings were seemingly
umented no evidence of a long-term benefit of MMF versus confirmed in a single-center study: the standard 2-g dose of
azathioprine with CsA-ME.152 MMF produced no benefit in rejection risk among African
Studies comparing MMF with sirolimus on everolimus (in Americans (relative risk 0.88), whereas white recipients
combination with CsA) have typically shown comparable fared much better (RR 0.35).149 Registry data, however,
outcomes, with a differing side effect profile.153 With a sig- supported an overall benefit for African American recipi-
nificant percentage of CsA-treated recipients on standard ents.156 Review of data from the pivotal trial and other stud-
doses of MMF or EC-MPS not achieving adequate exposure ies found that the excess acute rejections among African
to MPA early after transplantation (see section on thera- Americans, and others, occurred quite early after transplan-
peutic drug monitoring later), a European trial examined tation, a time when MPA pharmacokinetics are not stable
the effect of a “loading dose” approach. Investigators found and MPA exposure at standard doses is low in a substantial
more adverse events but substantially less early rejection number of recipients on CsA-based protocols.79,117,155,157
222 Kidney Transplantation: Principles and Practice

Study or subgroup MMF AZA Risk Ratio Weight Risk Ratio


M- M-
H,Random,95% H,Random,95%
n/N n/N CI CI
4 Longest duration of follow-up
MMF US Study 1995 87/331 77/164 17.0 % 0.56 [ 0.44, 0.71 ]

MMF TRI Study 1996 105/335 81/162 19.2 % 0.63 [ 0.50, 0.78 ]

Mendez 1998 24/117 19/59 5.1 % 0.64 [ 0.38, 1.07 ]

Ling 1998 2/11 3/5 0.7 % 0.30 [ 0.07, 1.28 ]

Egfjord 1999 8/25 11/25 2.7 % 0.73 [ 0.35, 1.50 ]

Suhail 2000 2/20 7/20 0.7 % 0.29 [ 0.07, 1.21 ]

Johnson 2000 12/72 16/76 3.1 % 0.79 [ 0.40, 1.56 ]

Busque 2001 2/23 8/23 0.7 % 0.25 [ 0.06, 1.05 ]

Folkmane 2001 5/23 8/25 1.6 % 0.68 [ 0.26, 1.78 ]

Ji 2001 11/56 22/50 3.7 % 0.45 [ 0.24, 0.83 ]

Sadek 2002 34/162 51/157 8.8 % 0.65 [ 0.44, 0.94 ]

COSTAMP Study 2002 59/243 83/246 13.7 % 0.72 [ 0.54, 0.95 ]

Tuncer 2002 7/38 13/38 2.2 % 0.54 [ 0.24, 1.20 ]

Weimer 2002 5/31 11/25 1.7 % 0.37 [ 0.15, 0.92 ]

Army Hospital 2002 1/17 3/16 0.3 % 0.31 [ 0.04, 2.71 ]

Sun 2002b 2/40 6/46 0.6 % 0.38 [ 0.08, 1.79 ]

Baltar 2002 1/14 5/12 0.4 % 0.17 [ 0.02, 1.27 ]

Miladipour 2002 4/40 10/40 1.3 % 0.40 [ 0.14, 1.17 ]

Keven 2003 3/24 3/17 0.7 % 0.71 [ 0.16, 3.10 ]

Merville 2004 5/37 7/34 1.3 % 0.66 [ 0.23, 1.87 ]

MYSS Study 2004 56/167 58/167 12.7 % 0.97 [ 0.72, 1.30 ]

Joh 2005 10/34 7/34 2.0 % 1.43 [ 0.62, 3.31 ]

Subtotal (95% CI) 1860 1441 100.0 % 0.65 [ 0.57, 0.73 ]

0.01 0.1 1 10 100


Less with MMF Less with AZA

Fig. 15.6 Forest plot shows the relative risk of acute rejection, comparing mycophenolate mofetil versus azathioprine. Boxes represent relative risk in
individual studies; diamonds represent summary effects. Horizontal bars represent 95% confidence intervals (CI). MMF, mycophenolate mofetil; AZA,
azathioprine. (Adapted from Wagner M, Earley AK, Webster AC, et al. Mycophenolic acid versus azathioprine as primary immunosuppression for kidney trans-
plant recipients. Cochrane Database Syst Rev 2015(12):CD007746.)

A subsequent study, conducted in stable renal allograft substantially fewer changes suggestive of chronic allograft
recipients, indicated no difference in MPA pharmacokinet- nephropathy and interpreted the data as indicating that
ics based on race or gender.87 In current practice, there is MMF may attenuate CsA nephrotoxicity.159 Indeed, data
no evidence supporting long-term use of a 3-g dose in non- from the earliest days of MMF use have suggested it may
CsA-based regimens. mitigate chronic allograft nephropathy.145 Patients in the
A 2003 paper regarding late kidney allograft failure USRDS registry who were maintained on MMF for at least 2
identified chronic CsA-induced nephrotoxicity as its prin- years were reported to show a 34% reduced risk of worsening
cipal etiology.158 The study was based on serial surveil- renal function, with significantly less late rejection.145,160 A
lance biopsies in recipients treated with CsA, azathioprine, Spanish study indicated that MMF demonstrated a benefit
and corticosteroids. A subsequent study from the same for patients with chronic allograft nephropathy even when
group, however, substituted MMF for azathioprine, found introduced late after transplantation, though contradictory
15 • Azathioprine and Mycophenolates 223

data exist.161,162 It now appears that most late allograft than patients on CsA, and many of the side effects of the two
failure is the result of immunologic mechanisms, with drugs are similar (e.g., diarrhea, myelosuppression), mak-
antibody-mediated injury playing a major role.163,164 The ing the combination difficult for some patients to tolerate.96
putative benefit of MMF in preserving renal function and However, an initial clinical study suggested that MMF
preventing late graft failure may actually reside in its effi- paired with sirolimus was at least as effective as MMF/CsA
cacy as an immunosuppressant with both anti-T cell and to prevent acute rejection.170 In the Symphony study, one of
anti-B cell effects. four treatment groups received sirolimus after daclizumab
induction in combination with MMF and corticosteroids.150
Almost 40% of the patients experienced acute rejection,
TACROLIMUS
with a high rate of study discontinuation, compromised
Studies evaluating the tacrolimus/MMF combination began graft survival, and relatively impaired glomerular filtration
appearing in the mid-1990s. A single-center, randomized rate (GFR) in 1 year. In a conversion trial involving 830
study evaluated adding MMF to a tacrolimus/steroid regi- patients switched between 6 and 120 months posttrans-
men and documented a reduction of acute rejection rates plant from CNIs to sirolimus (in combination with MMF and
at 1 year from 44% to 27%.109 A subsequent multicenter corticosteroids), only subjects with baseline estimated GFR
trial157 reported 2 g/day of MMF reduced the incidence of ≥40 mL/min demonstrated improved GFR 2 years later.171
acute rejection episodes compared with 1 g/day or azathi- Again, adverse effects were problematic, with converted
oprine (9%, 32%, and 32%, respectively). The low acute patients experiencing more hyperlipidemia, anemia, diar-
rejection rate with the higher MMF dose, without overt evi- rhea, stomatitis, and discontinuation from the study than
dence indicating greater risk of infection or malignancy, was those remaining on CNI-based therapy. A study of con-
thought to support the 2-g dose as optimal with tacrolimus; version from CsA-ME to everolimus demonstrated similar
however, the mean time to rejection was 79 days, there was results: more acute rejection, with disappointing benefit in
almost no rejection beyond 180 days in the 1-g MMF group, terms of GFR, adverse effects, and study discontinuation.172
and, by 1 year, patients in the 2-g group had undergone dose Nonetheless, some studies have demonstrated, in patients
reduction to a mean of 1.6 g/day (see section on therapeutic able to tolerate the mTOR/mycophenolate combination,
drug monitoring, later). In contrast, despite a 1-year rejec- stability of kidney function over time and a trend to fewer
tion rate of 15% with the tacrolimus/MMF combination, malignancies.173,174
another study failed to show a significant benefit of tacroli-
mus versus CsA-ME in combination with MMF on the inci- BELATACEPT
dence of acute rejection episodes, graft survival, or patient
survival in 220 subjects.165 A subsequent analysis of the Sci- Belatacept (Nulojix) is a monoclonal antibody that blocks
entific Registry of Transplant Recipients data in recipients of costimulation of lymphocytes by inhibiting the interac-
living donor kidneys found greater risk of graft failure with tion between CD80/86 and CD28. Approved for use in
the tacrolimus/MMF than with the CsA-ME/MMF combina- kidney recipients by US and European authorities in 2011,
tion.166 Although the Symphony trial documented fewer it is the first biologic agent to be utilized as maintenance
rejection episodes (12%) and better graft survival with tacro- therapy. It is administered intravenously at varying inter-
limus/MMF than two CsA-ME-based control groups,150 a vals after transplantation, and, from its earliest trials, has
trial of modified-release tacrolimus documented efficacy and been combined with mycophenolate and corticosteroids.175
safety only similar to the CsA-ME/MMF control group.104 Two treatment regimens (moderate and low intensity) of
Eventually, efficacy and safety data obtained in two stud- belatacept have been studied in three large multicenter tri-
ies that compared tacrolimus with CsA-ME in combination als involving almost 1500 patients. Efficacy in preventing
with MMF led to the FDA’s approval of the tacrolimus/MPA acute rejection and promoting graft and patient survival is
regimen in 2009, at a recommended starting dose of 2g/day. comparable to CsA, with better preservation of renal func-
With tacrolimus/MMF established as a new standard, tion.175–177 There may also be a benefit of the belatacept/
several reports documented similar efficacy of tacrolimus- MMF combination in reducing the formation of de novo
based protocols with sirolimus.167,168 In a randomized, anti-HLA antibodies.177 In 2016, 7-year data from the BEN-
multicenter clinical trial comparing patients treated with EFIT study documented improved patient and graft survival
MMF (n = 176) versus sirolimus (n = 185) (along with among patients who remained on belatacept for the dura-
tacrolimus and steroids), there was no difference in rejec- tion.178 Adverse effects were more common in a “moder-
tion, graft survival, or patient survival. However, the MMF ate-intensity” belatacept cohort that received more of the
cohort displayed better renal function, less hypertension, agent early after transplantation; the “low-intensity” regi-
and reduced hyperlipidemia.168 Among patients on a tacro- men is now approved for clinical use. The Emory group has
limus-based regimen, improvements in renal function were recently published a novel protocol to reduce early acute
observed in 19 patients converted from sirolimus to MMF rejection while capturing the long-term beneficial effect of
compared with 78 recipients remaining on sirolimus.169 a belatacept/MMF combination on renal function by add-
ing tacrolimus for a short duration after transplantation.179
The belatacept/MMF combination was associated with
mTOR INHIBITORS
more posttransplant lymphoproliferative disease than in
Mycophenolates have been utilized with sirolimus in an comparator patients, particularly in those naïve for Epstein-
attempt to avoid CNIs altogether. Patients receiving siro- Barr virus before transplantation; the combination is con-
limus (and presumably everolimus) in combination with traindicated in such patients. A single case of PML has also
mycophenolate have substantially greater MPA exposure been reported.180
224 Kidney Transplantation: Principles and Practice

similar at 3 years with better renal function in those on


Use of Mycophenolates to everolimus. However, given recent evidence linking inade-
Minimize or Avoid Other quate immunosuppression to late graft failure, enthusiasm
Immunosuppressants for these protocols may be diminishing.186

CALCINEURIN INHIBITOR MINIMIZATION CNI AVOIDANCE


Although CNIs have played a major beneficial role in the Experience with an MMF/steroid maintenance regimen
evolution of modern transplantation, their use is associated from the time of transplantation has typically not been good.
with significant toxicities, including glucose intolerance In 98 low-risk kidney recipients, acute rejection episodes
and renal impairment. The efficacy and relative tolerabil- were observed in 53% within 12 months, requiring insti-
ity of mycophenolate have enabled downward recalibra- tution of CNIs.142 A smaller study confined to recipients of
tion of what is considered therapeutic levels or doses of living donor kidneys also observed more rejection and little
both CsA and tacrolimus, potentially reducing their toxic or no overall benefit.187 In contrast, a single-center report
effects. Early on, dose reduction was typically performed described 12 patients older than 50 years who received
late (beyond 6–12 months) in response to declining renal grafts from elderly donors and were successfully treated de
function.181 The CAESAR trial tested full-dose versus low- novo with MMF, steroids, and an induction regimen of rab-
dose CsA-ME (target C0 150–300 ng/mL vs. 50–100 ng/ bit ATG.188 Durability of benefit may also be an issue; in a
mL) in combination with MMF and corticosteroids from Spanish study, 65% of MMF-treated patients remained on
the time of transplantation, with half the low-dose patients an avoidance regimen at 12 months, but only 36% were
randomized to withdraw CsA-ME after 6 months.182 The CNI-free at 60 months.189
withdrawal patients experienced significantly more acute A more successful approach, though not uniformly so,
rejection episodes, but only after CsA-ME was tapered has been the substitution of another agent for CNI at the
and discontinued. The best outcomes were in the patients time of transplantation. This was the basis of the belatacept
who remained on long-term, low-dose CsA-ME treatment, experience (see earlier discussion) and has also provided the
although GFR did not differ significantly from the full-dose underpinnings for the development of tofacitinib.98 Experi-
patients. Likewise, in the Symphony trial, patients on low- ence with mTOR inhibitors from the time of transplantation
dose CsA-ME fared comparably to those on full dose CsA- has been mixed, with Kreis in France and Flechner at the
ME.150 However, patients on low-dose tacrolimus (target Cleveland Clinic reporting good results,96,170 and Larson
C0 3–7 ng/mL) demonstrated fewer rejections, better renal from the Mayo Clinic reporting neither advantage nor dis-
function, and better graft survival than patients in any of advantage from substituting sirolimus for tacrolimus.176
the three comparator groups. This effect remained evident However, other studies, including Symphony, documented
with 3 years of follow-up.183 more rejection, a more problematic adverse event profile,
and little or no benefit in renal function utilizing a MMF/
CNI WITHDRAWAL sirolimus combination in de novo patients.173,190

Mycophenolates have also been used in attempts to elimi- CORTICOSTEROID AVOIDANCE OR


nate CNIs completely from a maintenance regimen, again
WITHDRAWAL
with the goal of averting chronic nephrotoxicity.158 One
multicenter study of patients with deteriorating renal func- Availability of a potent agent like MMF was anticipated, in
tion at a mean of 6 years after transplantation documented combination with CsA, to allow elimination of long-term
significantly improved renal function at 6 and 12 months corticosteroid therapy and its attendant complications. A
after stepwise withdrawal of CsA.184 A multicenter study large National Institutes of Health-sponsored multicenter
compared 1-year outcomes of withdrawal at 3 months of trial of steroid withdrawal at 3 months after transplantation
either CsA (n = 44) or MMF (n = 40) from a three-drug using a CsA-ME/MMF regimen documented an increased
regimen, including steroid therapy.185 Withdrawal of CsA risk of rejection episodes (particularly in minority patients),
was associated with better renal function, decreased blood and resulted in early termination of the study.191 The FREE-
pressure, and more favorable lipid profiles despite a twofold DOM study examined steroid avoidance and withdrawal
increase in acute rejection episodes. Several studies have in patients on CsA and EC-MPS after basiliximab induc-
substituted sirolimus for CNIs in otherwise stable patients, tion; a control group that remained on maintenance ste-
with generally good results. In a trial designated “Spare-the- roids clearly fared best.192 In contrast, steroid withdrawal
Nephron,” stable patients 1 to 6 months posttransplant (on beginning at 3 months after transplantation showed no
either tacrolimus or CsA-ME, as well as MMF and steroids) significant difference in rejection rates versus continued
were randomized to remain on their CNI (mostly tacroli- steroid therapy among European patients prescribed MMF/
mus) or switch to sirolimus.174 Results at 2 years docu- tacrolimus193 or MMF/CsA-ME with antibody induction.194
mented similar renal function between groups and a trend Although the superior results of the later trials might be
to less rejection and graft loss in the converted patients, attributed to more potent baseline therapy, it might also
with a different adverse event profile. The ZEUS trial ran- represent a difference in patient populations.
domized CsA-treated patients on EC-MPS to remain on CNIs After completion of these trials, philosophy regarding
or switched to everolimus at 4.5 months after transplanta- the timing of steroid withdrawal changed: rather than in
tion.173 Despite more rejections among converted patients chronic, stable patients, the most advantageous time point
(13% vs. 5%, P = 0.01), patient and graft survival were might be early after transplantation, either avoiding steroids
15 • Azathioprine and Mycophenolates 225

altogether or withdrawing under the cover of heavier immu- exposure was measured via limited sampling AUC, and
nosuppression with closer monitoring. A European study dose adjustments were made by study nurses according
showed success with only a single methylprednisolone dose to a Bayesian algorithm. There was a significant benefit
at transplant along with daclizumab induction and tacroli- for the CC group at 12 months with less acute rejection
mus/MMF maintenance therapy: 89% of patients were ste- and fewer treatment failures. The MMF dose was higher
roid-free at 6 months, with less posttransplant diabetes and in the CC group at day 14 (P < 0.0001), month 1 (P <
stable kidney function.195 In a large, randomized, blinded 0.0001), and month 3 (P < 0.01), as were median AUCs
study where corticosteroids were withdrawn within 7 days on day 14 (34 vs. 27 mg/h/L; P = 0.00001) and at month
of transplantation after lymphocyte depletion with rabbit 1 (45 vs. 31 mg/h/L; P < 0.0001). Adverse events did not
ATG, tacrolimus/MMF combination was equally effective differ between groups.
with or without maintenance steroids over 5 years post- In contrast were two larger trials in Europe and the
transplant.196 In another large multicenter trial, depletional US. The FDCC study enrolled 901 European kidney trans-
induction with alemtuzumab or rabbit ATG facilitated excel- plant recipients, randomizing to either CC MMF dosing
lent outcomes over 3 years with tacrolimus/MMF/cortico- (target AUC 45 mg/h/L) or standard fixed dosing, in com-
steroid-free combination.197 It should be remembered that, bination with CNIs (predominantly tacrolimus) and cor-
beyond the early avoidance philosophy, use of MMF at stan- ticosteroids.117 Dose changes in the FDCC trial (as well as
dard doses (as mandated in these trials) with tacrolimus is Opticept79) were made by investigators without specific
associated with substantially greater MPA exposure than the guidance as to frequency or scope. The two treatment
CsA-based trials noted in the previous paragraph, with the groups were largely indistinguishable, with similar MPA
enhanced exposure perhaps influencing outcomes. exposure in both at all time points and a similar frequency
of acute rejection episodes and treatment failure. Again,
however, there was a strong relationship between MPA
Therapeutic Drug Monitoring exposure and risk of acute rejection; 37% of subjects had
AUC <30 mg/h/L at day 3, more commonly in those taking
Although fixed doses of MMF and EC-MPS have been the CsA-ME than tacrolimus. In the Opticept trial conducted
norm in most clinical trials and in practice, the marked in the US, 720 patients were randomized to one of three
interindividual variability in pharmacokinetics provides groups: a control group on fixed-dose MMF and standard-
a rationale to implement therapeutic drug monitoring.198 dose CNI (roughly 80% tacrolimus, 20% CsA) or one of two
Despite some interest in pharmacodynamic monitoring, groups with CC MMF administration, and either standard
only pharmacokinetic monitoring has been widely tested or dose or reduced-dose CNI.79 CC dosing of MMF was guided
implemented. In an early study designed to establish a data by trough MPA levels, with a target of 1.3 μL/mL for CsA
set around MPA exposure (AUC) among kidney transplant patients and 1.9 μL/mL for those on tacrolimus. Antibody
recipients, 150 were randomized to one of three dose groups induction was allowed, with approximately a third of
of MMF (in combination with CsA and steroids).82 This study patients receiving rabbit ATG, a third basiliximab, and a
documented poor bioavailability early after transplanta- third no induction. Again, the groups were largely indistin-
tion that seemed to stabilize by week 12, when AUCs were guishable, with similar outcomes, though almost all tacroli-
almost double those seen in the first 2 weeks posttransplant. mus patients (with a starting MMF dose of 2 g daily) quickly
There were substantially fewer acute rejection episodes in displayed therapeutic MPA exposure: there was a strong
patients with AUC > 30 mg/h/L. Patients in the highest relationship in these patients between adequate exposure
MPA target group experienced more adverse events, which (trough MPA level ≥ 1.6 μg/mL) and reduced risk of acute
in this trial resulted in study discontinuation, but no upper rejection (Fig. 15.7).
limit to define toxicity risk could be determined. Although A more recent consensus conference attempted to
full 12-hour AUC measurement is unwieldy for clinical put these data into perspective. 200 AUC targets of 30
monitoring, limited sampling strategies correlate well with to 60 mg/h/L were recommended, with the upper limit
overall MPA exposure. Trough level monitoring correlates established primarily as an indicator of no additional
less well with AUC, but recent studies documented an r2 of efficacy rather than associated with toxicity. Recom-
0.68 for patients also receiving tacrolimus (less so for CsA- mended trough concentrations are C 0 ≥ 1.3 mg/L with
ME), and low intraindividual variability77,79 (see Fig. 15.4). CsA and C 0 ≥1.9 mg/L in patients on tacrolimus. In
It should be reiterated that trough levels and AUC do not both the FDCC and Opticept trials, a standard 2-g daily
correlate (r2 = 0.02) for EC-MPS. Finally, concomitant drug dose of MMF produced adequate early MPA exposure in
therapy matters. Kuypers and colleagues documented the almost all tacrolimus-treated patients, whereas in a sig-
lowest risk for BPAR when both tacrolimus and MPA expo- nificant percentage of patients on the CsA/MMF com-
sure were therapeutic, intermediate risk when one or the bination, it did not. 79,117 In standard patients, there
other (but not both) was therapeutic, and the greatest risk was no indication that therapeutic drug monitoring
if both drugs were subtherapeutic.199 improved clinical outcomes. The greatest clinical bene-
Three large, multicenter studies have prospectively fit of therapeutic drug monitoring might be expected in
tested the effect of therapeutic drug monitoring-based patients on dual immunosuppression, patients under-
MMF dosing on clinical outcomes in kidney transplanta- going CNI- or steroid-sparing regimens, those at high
tion. In a French study (APOMYGRE), all subjects received immunologic risk, those with delayed graft function,
CsA and steroids and were randomized to receive fixed- or those with altered gastrointestinal, hepatic, or renal
dose or concentration-controlled (CC) MMF.75 MMF function. 200,201
226 Kidney Transplantation: Principles and Practice

0.40
MPA trough level ≥1.6 µg/mL
0.35 (P =0.0001) <1.6 µg/mL

Proportion of patients with first biopsy


0.30

proven acute rejection


0.25

0.20

0.15

0.10

0.05

0.00
0 3 6 9 12 15
Time since first trial medication (months)
Fig. 15.7 Mycophenolic acid (MPA) exposure and time to first biopsy-proven acute rejection episode. Cox proportional hazards model estimate with
the abbreviated MPA area under the concentration-time curve, baseline hypertension/diabetes, and treatment effect (controls versus concentration-
controlled dosing) as covariates. Cutoff point of ≥1.6 μg/mL was based on receiver operating characteristic analysis of the study data. (From Gaston RS,
Kaplan B, Shah T, et al. Fixed- or controlled-dose MMF with standard- or reduced-dose calcineurin inhibitors: the Opticept trial. Am J Transplant 2009;1607.)

References 14. Starzl TE, Marchioro TL, Waddell WR. the reversal of rejection in
human renal homografts with subsequent development of homo-
1. Allison AC, Eugui EM. Mechanisms of action of mycophenolate graft tolerance. Surg Gynecol Obstet 1963;117:385–95.
mofetil in preventing acute and chronic allograft rejection. Trans- 15. Franklin TJ, Cook JM. The inhibition of nucleic acid synthesis by
plantation 2005;80(Suppl. 2):S181–90. mycophenolic acid. Biochem J 1969;113(3):515–24.
2. Eugui EM, Almquist SJ, Muller CD, et al. Lymphocyte-selective cyto- 16. Mitsui A, Suzuki S. Immunosuppressive effect of mycophenolic acid.
static and immunosuppressive effects of mycophenolic acid in vitro: J Antibiot (Tokyo) 1969;22(8):358–63.
role of deoxyguanosine nucleotide depletion. Scand J Immunol 17. Allison AC, Hovi T, Watts RW, et al. Immunological observa-
1991;33(2):161–73. tions on patients with Lesch-Nyhan syndrome, and on the role of
3. Elion GB, Burgi E, Hitchings GH. Studies on condensed pyrimidine de-novo purine synthesis in lymphocyte transformation. Lancet
systems. IX. The synthesis of some 6-substituted purines. J Am Chem 1975;2(7946):1179–83.
Soc 1952;74(2):411–4. 18. Allison AC, Kowalski WJ, Muller CJ, et al. Mycophenolic acid and
4. Elion GB, Bieber S, Hitchings GH. A summary of investiga- brequinar, inhibitors of purine and pyrimidine synthesis, block the
tions with 2-amino-6-[(1-methyl-4-nitro-5-imidazolyl)thio]purine glycosylation of adhesion molecules. Transplant Proc 1993;25(3
(B.W. 57-323) in animals. Cancer Chemother Rep 1960;8: Suppl. 2):67–70.
36–43. 19. Lee WA, Gu L, Miksztal AR, et al. Bioavailability improvement of
5. Schwartz R, Dameshek W. Drug-induced immunological tolerance. mycophenolic acid through amino ester derivatization. Pharm Res
Nature 1959;183(4676):1682–3. 1990;7(2):161–6.
6. Schwartz R, Dameshek W. The effects of 6-mercaptopurine on 20. Morris RE, Wang J, Blum JR, et al. Immunosuppressive effects of the
homograft reactions. J Clin Invest 1960;39:952–8. morpholinoethyl ester of mycophenolic acid (RS-61443) in rat and
7. Calne RY. The rejection of renal homografts. Inhibition in dogs by nonhuman primate recipients of heart allografts. Transplant Proc
6-mercaptopurine. Lancet 1960;1(7121):417–8. 1991;23(2 Suppl. 2):19–25.
8. Zukoski CF, Lee HM, Hume DM. The prolongation of functional sur- 21. Platz KP, Sollinger HW, Hullett DA, et al. RS-61443—a new, potent
vival of canine renal homografts by 6-mercaptopurine. Surg Forum immunosuppressive agent. Transplantation 1991;51(1):27–31.
1960;11:470–2. 22. Goldblum R. Therapy of rheumatoid arthritis with mycophenolate
9. Elion GB, Callahan S, Nathan H, et al. Potentiation by inhibition of mofetil. Clin Exp Rheumatol 1993;11(Suppl. 8):S117–9.
drug degradation: 6-substituted purines and xanthine oxidase. Bio- 23. Sollinger HW, Deierhoi MH, Belzer FO, et al. RS-61443—a phase I clini-
chem Pharmacol 1963;12(1):85–93. cal trial and pilot rescue study. Transplantation 1992;53(2):428–32.
10. Calne RY, Alexandre GP, Murray JE. A study of the effects of drugs 24. Sollinger HW. Mycophenolate mofetil for the prevention of acute
in prolonging survival of homologous renal transplants in dogs. Ann rejection in primary cadaveric renal allograft recipients. U.S. Renal
NY Acad Sci 1962;99:743–61. Transplant Mycophenolate Mofetil Study Group. Transplantation
11. Murray JE, Merrill JP, Dammin GJ, et al. Kidney transplantation in 1995;60(3):225–32.
modified recipients. Ann Surg 1962;156:337–55. 25. Tricontinental Mycophenolate Mofetil Renal Transplantation Study
12. Murray JE, Merrill JP, Harrison JH, et al. Prolonged survival of Group. A blinded, randomized clinical trial of mycophenolate mofetil
human-kidney homografts by immunosuppressive drug therapy. for the prevention of acute rejection in cadaveric renal transplanta-
N Engl J Med 1963;268:1315–23. tion. Transplantation 1996;61(7):1029–37.
13. Goodwin W, Mims M, Kaufman J. Human renal transplantation 26. European Mycophenolate Mofetil Cooperative Study Group.
III. Technical problems encountered in six cases of kidney homo- Placebo-controlled study of mycophenolate mofetil combined with
transplantation. Transact Am Assoc of Genito-Urinary Surg. cyclosporin and corticosteroids for prevention of acute rejection.
1961;54:116–25. Lancet 1995;345(8961):1321–5.
15 • Azathioprine and Mycophenolates 227

27. U.S. Food and Drug Administration. CellCept® (mycophenolate 51. Opelz G, Döhler B. Critical threshold of azathioprine dosage for main-
mofetil) full prescribing information. Available online at: www.acce tenance immunosuppression in kidney graft recipients. Collabora-
ssdata.fda.gov/drugsatfda_docs/label/2009/050722s021,050723 tive Transplant Study. Transplantation 2000;69(5):818–21.
s019,050758s019,050759s024lbl.pdf. Accessed October 5, 2017. 52. Bach JF, Dardenne M. The metabolism of azathioprine in renal fail-
28. Kaufman DB, Shapiro R, Lucey MR, et al. Immunosuppression: prac- ure. Transplantation 1971;12(4):253–9.
tice and trends. Am J Transplant 2004;4(Suppl. 9):38–53. 53. Schütz E, Gummert J, Mohr FW, et al. Should 6-thioguanine nucleo-
29. U.S. Food and Drug Administration. Myfortic® (mycophenolic acid) tides be monitored in heart transplant recipients given azathioprine?
delayed release tablets. Prescribing information. Available online at: Ther Drug Monit 1996;18(3):228–33.
https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/050 54. Fabre MA, Jones DC, Bunce M, et al. The impact of thiopurine
791s007lbl.pdf. Accessed October 5, 2017. S-methyltransferase polymorphisms on azathioprine dose 1 year
30. Budde K, Curtis J, Knoll G, et al. Enteric-coated mycophenolate after renal transplantation. Transpl Int 2004;17(9):531–9.
sodium can be safely administered in maintenance renal transplant 55. Kurzawski M, Dziewanowski K, Gawrońska-Szklarz B, et al. The
patients: results of a 1-year study. Am J Transplant 2004;4(2):237– impact of thiopurine s-methyltransferase polymorphism on azathi-
43. oprine-induced myelotoxicity in renal transplant recipients. Ther
31. Salvadori M, Holzer H, de Mattos A, et al. Enteric-coated myco- Drug Monit 2005;27(4):435–41.
phenolate sodium is therapeutically equivalent to mycopheno- 56. Sanderson J, Ansari A, Marinaki T, et al. Thiopurine methyltrans-
late mofetil in de novo renal transplant patients. Am J Transplant ferase: should it be measured before commencing thiopurine drug
2004;4(2):231–6. therapy? Ann Clin Biochem 2004;41(Pt 4):294–302.
32. Ensor CR, Trofe-Clark J, Gabardi S, et al. Generic maintenance 57. Thompson AJ, Newman WG, Elliott RA, et al. The cost-effectiveness
immunosuppression in solid organ transplant recipients. Pharmaco- of a pharmacogenetic test: a trial-based evaluation of TPMT geno-
therapy 2011;31(11):1111–29. typing for azathioprine. Value Health 2014;17(1):22–33.
33. Molnar AO, Fergusson D, Tsampalieros AK, et al. Generic immuno- 58. Lennard L, Murphy MF, Maddocks JL. Severe megaloblastic anaemia
suppression in solid organ transplantation: systematic review and associated with abnormal azathioprine metabolism. Br J Clin Phar-
meta-analysis. BMJ 2015;350:h3163. macol 1984;17(2):171–2.
34. van Gelder T, Hesselink DA. Mycophenolate revisited. Transpl Int 59. Coghill AE, Johnson LG, Berg D, et al. Immunosuppressive medica-
2015;28(5):508–15. tions and squamous cell skin carcinoma: nested case-control study
35. Vincenti F, Klintmalm G, Halloran PF. Open letter to the FDA: new within the skin cancer after organ transplant (SCOT) cohort. Am J
drug trials must be relevant. Am J Transplant 2008;8(4):733–4. Transplant 2016;16(2):565–73.
36. Giblett ER, Anderson JE, Cohen F, et al. Adenosine-deaminase defi- 60. Ducroux E, Martin C, Bouwes Bavinck JN, et al. Risk of aggressive
ciency in two patients with severely impaired cellular immunity. skin cancers after kidney retransplantation in patients with previous
Lancet 1972;2(7786):1067–9. posttransplant cutaneous squamous cell carcinomas: a retrospective
37. Carr SF, Papp E, Wu JC, et al. Characterization of human type I and study of 53 cases. Transplantation 2017;101(4):e133–41.
type II IMP dehydrogenases. J Biol Chem 1993;268(36):27286–90. 61. Jiyad Z, Olsen CM, Burke MT, et al. Azathioprine and risk of skin
38. Colic M, Stojic-Vukanic Z, Pavlovic B, et al. Mycophenolate mofetil cancer in organ transplant recipients: systematic review and meta-
inhibits differentiation, maturation and allostimulatory function analysis. Am J Transplant 2016;16(12):3490–503.
of human monocyte-derived dendritic cells. Clin Exp Immunol 62. Safaeian M, Robbins HA, Berndt SI, et al. Risk of colorectal cancer
2003;134(1):63–9. after solid organ transplantation in the United States. Am J Trans-
39. Grailer A, Nichols J, Hullett D, et al. Inhibition of human B cell plant 2016;16(3):960–7.
responses in vitro by RS-61443, cyclosporine A and DAB486 IL-2. 63. Hofbauer GF, Attard NR, Harwood CA, et al. Reversal of UVA skin
Transplant Proc 1991;23(1 Pt 1):314–5. photosensitivity and DNA damage in kidney transplant recipients by
40. Kimball JA, Pescovitz MD, Book BK, et al. Reduced human IgG anti- replacing azathioprine. Am J Transplant 2012;12(1):218–25.
ATGAM antibody formation in renal transplant recipients receiving 64. Bullingham R, Monroe S, Nicholls A, et al. Pharmacokinetics and
mycophenolate mofetil. Transplantation 1995;60(12):1379–83. bioavailability of mycophenolate mofetil in healthy subjects after
41. Figueroa J, Fuad SA, Kunjummen BD, et al. Suppression of synthe- single-dose oral and intravenous administration. J Clin Pharmacol
sis of natural antibodies by mycophenolate mofetil (RS-61443). 1996;36(4):315–24.
Its potential use in discordant xenografting. Transplantation 65. de Winter BC, Mathot RA, Sombogaard F, et al. Nonlinear relation-
1993;55(6):1371–4. ship between mycophenolate mofetil dose and mycophenolic acid
42. Smith KG, Isbel NM, Catton MG, et al. Suppression of the humoral exposure: implications for therapeutic drug monitoring. Clin J Am
immune response by mycophenolate mofetil. Nephrol Dial Trans- Soc Nephrol 2011;6(3):656–63.
plant 1998;13(1):160–4. 66. Sunder-Plassmann G, Reinke P, Rath T, et al. Comparative pharma-
43. Moon JI, Kim YS, Kim MS, et al. Effect of cyclosporine, mycophenolic cokinetic study of two mycophenolate mofetil formulations in stable
acid, and rapamycin on the proliferation of rat aortic vascular smooth kidney transplant recipients. Transpl Int 2012;25(6):680–6.
muscle cells: in vitro study. Transplant Proc 2000;32(7):2026–7. 67. Arns W, Breuer S, Choudhury S, et al. Enteric-coated mycopheno-
44. Hauser IA, Renders L, Radeke HH, et al. Mycophenolate mofetil late sodium delivers bioequivalent MPA exposure compared with
inhibits rat and human mesangial cell proliferation by guanosine mycophenolate mofetil. Clin Transplant 2005;19(2):199–206.
depletion. Nephrol Dial Transplant 1999;14(1):58–63. 68. Shaw LM, Korecka M, Venkataramanan R, et al. Mycophenolic acid
45. Badid C, Vincent M, McGregor B, et al. Mycophenolate mofetil pharmacodynamics and pharmacokinetics provide a basis for ratio-
reduces myofibroblast infiltration and collagen III deposition in rat nal monitoring strategies. Am J Transplant 2003;3(5):534–42.
remnant kidney. Kidney Int 2000;58(1):51–61. 69. Atcheson BA, Taylor PJ, Mudge DW, et al. Mycophenolic acid phar-
46. Klupp J, Dambrin C, Hibi K, et al. Treatment by mycophenolate macokinetics and related outcomes early after renal transplant. Br J
mofetil of advanced graft vascular disease in non-human pri- Clin Pharmacol 2005;59(3):271–80.
mate recipients of orthotopic aortic allografts. Am J Transplant 70. Bullingham RE, Nicholls AJ, Kamm BR. Clinical pharmacokinetics of
2003;3(7):817–29. mycophenolate mofetil. Clin Pharmacokinet 1998;34(6):429–55.
47. Azuma H, Binder J, Heemann U, et al. Effects of RS61443 on func- 71. Nowak I, Shaw LM. Mycophenolic acid binding to human serum
tional and morphological changes in chronically rejecting rat kidney albumin: characterization and relation to pharmacodynamics. Clin
allografts. Transplantation 1995;59(4):460–6. Chem 1995;41(7):1011–7.
48. Jolicoeur EM, Qi S, Xu D, et al. Combination therapy of mycopheno- 72. Lamba M, Tafti B, Melcher M, et al. Population pharmacokinetic
late mofetil and rapamycin in prevention of chronic renal allograft analysis of mycophenolic acid coadministered with either tasocitinib
rejection in the rat. Transplantation 2003;75(1):54–9. (CP-690,550) or tacrolimus in adult renal allograft recipients. Ther
49. Shihab FS, Bennett WM, Yi H, et al. Combination therapy with siroli- Drug Monit 2010;32(6):778–81.
mus and mycophenolate mofetil: effects on the kidney and on trans- 73. Holt DW. Monitoring mycophenolic acid. Ann Clin Biochem
forming growth factor-beta1. Transplantation 2004;77(5):683–6. 2002;39(Pt 3):173–83.
50. Dell’Oglio MP, Zaza G, Rossini M, et al. The anti-fibrotic effect 74. Morris RG. Immunosuppressant drug monitoring: is the laboratory
of mycophenolic acid-induced neutral endopeptidase. J Am Soc meeting clinical expectations? Ann Pharmacother 2005;39(1):
Nephrol 2010;21(12):2157–68. 119–27.
228 Kidney Transplantation: Principles and Practice

75. Le Meur Y, Büchler M, Thierry A, et al. Individualized mycopheno- 96. Flechner SM, Goldfarb D, Modlin C, et al. Kidney transplantation with-
late mofetil dosing based on drug exposure significantly improves out calcineurin inhibitor drugs: a prospective, randomized trial of siro-
patient outcomes after renal transplantation. Am J Transplant limus versus cyclosporine. Transplantation 2002;74(8):1070–6.
2007;7(11):2496–503. 97. van Gelder T, Klupp J, Barten MJ, et al. Comparison of the effects of
76. Kiberd BA, Lawen J, Fraser AD, et al. Early adequate mycophenolic tacrolimus and cyclosporine on the pharmacokinetics of mycophe-
acid exposure is associated with less rejection in kidney transplanta- nolic acid. Ther Drug Monit 2001;23(2):119–28.
tion. Am J Transplant 2004;4(7):1079–83. 98. Vincenti F, Tedesco Silva H, Busque S, et al. Randomized phase
77. van Hest RM, Mathot RA, Vulto AG, et al. Within-patient variability 2b trial of tofacitinib (CP-690,550) in de novo kidney transplant
of mycophenolic acid exposure: therapeutic drug monitoring from a patients: efficacy, renal function and safety at 1 year. Am J Trans-
clinical point of view. Ther Drug Monit 2006;28(1):31–4. plant 2012;12(9):2446–56.
78. Borrows R, Chusney G, Loucaidou M, et al. Mycophenolic acid 12-h 99. Cattaneo D, Perico N, Gaspari F, et al. Glucocorticoids interfere with
trough level monitoring in renal transplantation: association with mycophenolate mofetil bioavailability in kidney transplantation.
acute rejection and toxicity. Am J Transplant 2006;6(1):121–8. Kidney Int 2002;62(3):1060–7.
79. Gaston RS, Kaplan B, Shah T, et al. Fixed- or controlled-dose myco- 100. Halloran P, Mathew T, Tomlanovich S, et al. Mycophenolate mofetil
phenolate mofetil with standard- or reduced-dose calcineurin inhibi- in renal allograft recipients: a pooled efficacy analysis of three ran-
tors: the Opticept trial. Am J Transplant 2009;9(7):1607–19. domized, double-blind, clinical studies in prevention of rejection.
80. de Winter BC, van Gelder T, Glander P, et al. Population pharmaco- The International Mycophenolate Mofetil Renal Transplant Study
kinetics of mycophenolic acid: a comparison between enteric-coated Groups. Transplantation 1997;63(1):39–47.
mycophenolate sodium and mycophenolate mofetil in renal trans- 101. Kuypers DR, de Jonge H, Naesens M, et al. Current target ranges
plant recipients. Clin Pharmacokinet 2008;47(12):827–38. of mycophenolic acid exposure and drug-related adverse events: a
81. Hale MD, Nicholls AJ, Bullingham RE, et al. The pharmacokinetic- 5-year, open-label, prospective, clinical follow-up study in renal
pharmacodynamic relationship for mycophenolate mofetil in renal allograft recipients. Clin Ther 2008;30(4):673–83.
transplantation. Clin Pharmacol Ther 1998;64(6):672–83. 102. Bunnapradist S, Lentine KL, Burroughs TE, et al. Mycophenolate
82. van Gelder T, Hilbrands LB, Vanrenterghem Y, et al. A randomized mofetil dose reductions and discontinuations after gastrointestinal
double-blind, multicenter plasma concentration controlled study of complications are associated with renal transplant graft failure.
the safety and efficacy of oral mycophenolate mofetil for the preven- Transplantation 2006;82(1):102–7.
tion of acute rejection after kidney transplantation. Transplantation 103. European Mycophenolate Mofetil Cooperative Study Group. Myco-
1999;68(2):261–6. phenolate mofetil in renal transplantation: 3-year results from the
83. Jacobson P, Huang J, Rydholm N, et al. Higher mycophenolate dose placebo-controlled trial. Transplantation 1999;68(3):391–6.
requirements in children undergoing hematopoietic cell transplant 104. Silva HT, Yang HC, Abouljoud M, et al. One-year results with
(HCT). J Clin Pharmacol 2008;48(4):485–94. extended-release tacrolimus/MMF, tacrolimus/MMF and cyclospo-
84. Jain A, Venkataramanan R, Kwong T, et al. Pharmacokinetics of rine/MMF in de novo kidney transplant recipients. Am J Transplant
mycophenolic acid in liver transplant patients after intravenous 2007;7(3):595–608.
and oral administration of mycophenolate mofetil. Liver Transpl 105. Maes BD, Lemahieu W, Kuypers D, et al. Differential effect of diarrhea
2007;13(6):791–6. on FK506 versus cyclosporine A trough levels and resultant preven-
85. Weber LT, Hoecker B, Armstrong VW, et al. Long-term pharmaco- tion of allograft rejection in renal transplant recipients. Am J Trans-
kinetics of mycophenolic acid in pediatric renal transplant recipients plant 2002;2(10):989–92.
over 3 years posttransplant. Ther Drug Monit 2008;30(5):570–5. 106. Bunnapradist S, Neri L, Wong W, et al. Incidence and risk factors for
86. Hwang S, Song GW, Jung DH, et al. Intra-individual variability of diarrhea following kidney transplantation and association with graft
mycophenolic acid concentration according to renal function in loss and mortality. Am J Kidney Dis 2008;51(3):478–86.
liver transplant recipients receiving mycophenolate monotherapy. 107. Maes BD, Dalle I, Geboes K, et al. Erosive enterocolitis in mycophe-
Ann Hepatobiliary Pancreat Surg 2017;21(1):11–6. nolate mofetil-treated renal-transplant recipients with persistent
87. Pescovitz MD, Guasch A, Gaston R, et al. Equivalent pharmacokinet- afebrile diarrhea. Transplantation 2003;75(5):665–72.
ics of mycophenolate mofetil in African-American and Caucasian 108. Guerard A, Rabodonirina M, Cotte L, et al. Intestinal microsporidi-
male and female stable renal allograft recipients. Am J Transplant osis occurring in two renal transplant recipients treated with myco-
2003;3(12):1581–6. phenolate mofetil. Transplantation 1999;68(5):699–707.
88. Shaw LM, Korecka M, Aradhye S, et al. Mycophenolic acid area under 109. Shapiro R, Jordan ML, Scantlebury VP, et al. A prospective, random-
the curve values in African American and Caucasian renal transplant ized trial of tacrolimus/prednisone versus tacrolimus/prednisone/
patients are comparable. J Clin Pharmacol 2000;40(6):624–33. mycophenolate mofetil in renal transplant recipients. Transplanta-
89. Ogawa N, Nagashima N, Nakamura M, et al. Measurement of myco- tion 1999;67(3):411–5.
phenolate mofetil effect in transplant recipients. Transplantation 110. Papadimitriou JC, Drachenberg CB, Beskow CO, et al. Graft-versus-host
2001;72(3):422–7. disease-like features in mycophenolate mofetil-related colitis. Trans-
90. Budde K, Braun KP, Glander P, et al. Pharmacodynamic monitoring plant Proc 2001;33(3):2237–8.
of mycophenolate mofetil in stable renal allograft recipients. Trans- 111. Ducloux D, Ottignon Y, Semhoun-Ducloux S, et al. Mycophenolate
plant Proc 2002;34(5):1748–50. mofetil-induced villous atrophy. Transplantation 1998;66(8):1115–6.
91. Fukuda T, Goebel J, Thøgersen H, et al. Inosine monophosphate 112. Chan L, Mulgaonkar S, Walker R, et al. Patient-reported gastroin-
dehydrogenase (IMPDH) activity as a pharmacodynamic biomarker testinal symptom burden and health-related quality of life following
of mycophenolic acid effects in pediatric kidney transplant recipi- conversion from mycophenolate mofetil to enteric-coated mycophe-
ents. J Clin Pharmacol 2011;51(3):309–20. nolate sodium. Transplantation 2006;81(9):1290–7.
92. Quéméneur L, Flacher M, Gerland LM, et al. Mycophenolic 113. Jacobson PA, Schladt D, Oetting WS, et al. Genetic determinants of
acid inhibits IL-2-dependent T cell proliferation, but not IL- mycophenolate-related anemia and leukopenia after transplanta-
2-dependent survival and sensitization to apoptosis. J Immunol tion. Transplantation 2011;91(3):309–16.
2002;169(5):2747–55. 114. Bouamar R, Elens L, Shuker N, et al. Mycophenolic acid-related
93. Gregoor PJ, de Sévaux RG, Hené RJ, et al. Effect of cyclosporine on anemia and leucopenia in renal transplant recipients are
mycophenolic acid trough levels in kidney transplant recipients. related to genetic polymorphisms in CYP2C8. Transplantation
Transplantation 1999;68(10):1603–6. 2012;93(10):e39–40.
94. Grinyó JM, Ekberg H, Mamelok RD, et al. The pharmacokinetics of 115. van Besouw NM, van der Mast BJ, Smak Gregoor PJ, et al. Effect of
mycophenolate mofetil in renal transplant recipients receiving stan- mycophenolate mofetil on erythropoiesis in stable renal transplant
dard-dose or low-dose cyclosporine, low-dose tacrolimus or low-dose patients is correlated with mycophenolic acid trough levels. Nephrol
sirolimus: the Symphony pharmacokinetic substudy. Nephrol Dial Dial Transplant 1999;14(11):2710–3.
Transplant 2009;24(7):2269–76. 116. Kuypers DR, Vanrenterghem Y, Squifflet JP, et al. Twelve-month
95. Kobayashi M, Saitoh H, Tadano K, et al. Cyclosporin A, but not evaluation of the clinical pharmacokinetics of total and free myco-
tacrolimus, inhibits the biliary excretion of mycophenolic acid gluc- phenolic acid and its glucuronide metabolites in renal allograft recip-
uronide possibly mediated by multidrug resistance-associated pro- ients on low dose tacrolimus in combination with mycophenolate
tein 2 in rats. J Pharmacol Exp Ther 2004;309(3):1029–35. mofetil. Ther Drug Monit 2003;25(5):609–22.
15 • Azathioprine and Mycophenolates 229

117. van Gelder T, Silva HT, de Fijter JW, et al. Comparing mycophenolate 139. McKay DB, Josephson MA. Pregnancy after kidney transplantation.
mofetil regimens for de novo renal transplant recipients: the fixed- Clin J Am Soc Nephrol 2008;3(Suppl. 2):S117–25.
dose concentration-controlled trial. Transplantation 2008;86(8): 140. Deierhoi MH, Kauffman RS, Hudson SL, et al. Experience with myco-
1043–51. phenolate mofetil (RS61443) in renal transplantation at a single
118. Hurst FP, Altieri M, Nee R, et al. Poor outcomes in elderly kidney center. Ann Surg 1993;217(5):476–82; discussion 482–474.
transplant recipients receiving alemtuzumab induction. Am J 141. Sollinger HW, Belzer FO, Deierhoi MH, et al. RS-61443 (mycophe-
Nephrol 2011;34(6):534–41. nolate mofetil). A multicenter study for refractory kidney transplant
119. Banerjee R, Halil O, Bain BJ, et al. Neutrophil dysplasia caused by rejection. Ann Surg 1992;216(4):513–8; discussion 518–9.
mycophenolate mofetil. Transplantation 2000;70(11):1608–10. 142. Vincenti F, Monaco A, Grinyo J, et al. Multicenter randomized pro-
120. Hubner GI, Eismann R, Sziegoleit W. Relationship between myco- spective trial of steroid withdrawal in renal transplant recipients
phenolate mofetil side effects and mycophenolic acid plasma receiving basiliximab, cyclosporine microemulsion and mycopheno-
trough levels in renal transplant patients. Arzneimittelforschung late mofetil. Am J Transplant 2003;3(3):306–11.
2000;50(10):936–40. 143. Young M, Plosker GL. Mycophenolate mofetil: a pharmacoeconomic
121. Weber LT, Shipkova M, Armstrong VW, et al. The pharmacokinetic- review of its use in solid organ transplantation. Pharmacoeconomics
pharmacodynamic relationship for total and free mycophenolic 2002;20(10):675–713.
Acid in pediatric renal transplant recipients: a report of the german 144. Meier-Kriesche HU, Steffen BJ, Hochberg AM, et al. Mycophenolate
study group on mycophenolate mofetil therapy. J Am Soc Nephrol mofetil versus azathioprine therapy is associated with a significant
2002;13(3):759–68. protection against long-term renal allograft function deterioration.
122. van Gelder T, ter Meulen CG, Hené R, et al. Oral ulcers in kidney Transplantation 2003;75(8):1341–6.
transplant recipients treated with sirolimus and mycophenolate 145. Ojo AO, Meier-Kriesche HU, Hanson JA, et al. Mycophenolate mofetil
mofetil. Transplantation 2003;75(6):788–91. reduces late renal allograft loss independent of acute rejection.
123. Nashan B, Gaston R, Emery V, et al. Review of cytomegalovirus infec- Transplantation 2000;69(11):2405–9.
tion findings with mammalian target of rapamycin inhibitor-based 146. Wagner M, Earley AK, Webster AC, et al. Mycophenolic acid versus
immunosuppressive therapy in de novo renal transplant recipients. azathioprine as primary immunosuppression for kidney transplant
Transplantation 2012;93(11):1075–85. recipients. Cochrane Database Syst Rev 2015;(12):CD007746.
124. Kotton CN, Kumar D, Caliendo AM, et al. International consensus 147. Eckhoff DE, Young CJ, Gaston RS, et al. Racial disparities in
guidelines on the management of cytomegalovirus in solid organ renal allograft survival: a public health issue? J Am Coll Surg
transplantation. Transplantation 2010;89(7):779–95. 2007;204(5):894–902. discussion 902-3.
125. Hirsch HH, Brennan DC, Drachenberg CB, et al. Polyomavirus- 148. Irish W, Sherrill B, Brennan DC, et al. Three-year posttransplant
associated nephropathy in renal transplantation: interdisciplinary anal- graft survival in renal-transplant patients with graft function at 6
yses and recommendations. Transplantation 2005;79(10):1277–86. months receiving tacrolimus or cyclosporine microemulsion within
126. Hardinger KL, Koch MJ, Bohl DJ, et al. BK-virus and the impact of a triple-drug regimen. Transplantation 2003;76(12):1686–90.
pre-emptive immunosuppression reduction: 5-year results. Am J 149. Schweitzer EJ, Yoon S, Fink J, et al. Mycophenolate mofetil reduces
Transplant 2010;10(2):407–15. the risk of acute rejection less in African-American than in Cauca-
127. Neff RT, Hurst FP, Falta EM, et al. Progressive multifocal leukoen- sian kidney recipients. Transplantation 1998;65(2):242–8.
cephalopathy and use of mycophenolate mofetil after kidney trans- 150. Ekberg H, Tedesco-Silva H, Demirbas A, et al. Reduced exposure
plantation. Transplantation 2008;86(10):1474–8. to calcineurin inhibitors in renal transplantation. N Engl J Med
128. Schmedt N, Andersohn F, Garbe E. Signals of progressive multifo- 2007;357(25):2562–75.
cal leukoencephalopathy for immunosuppressants: a dispropor- 151. Remuzzi G, Lesti M, Gotti E, et al. Mycophenolate mofetil versus aza-
tionality analysis of spontaneous reports within the US Adverse thioprine for prevention of acute rejection in renal transplantation
Event Reporting System (AERS). Pharmacoepidemiol Drug Saf (MYSS): a randomised trial. Lancet 2004;364(9433):503–12.
2012;21(11):1216–20. 152. Remuzzi G, Cravedi P, Costantini M, et al. Mycophenolate mofetil
129. Rostaing L, Izopet J, Sandres K, et al. Changes in hepatitis C virus versus azathioprine for prevention of chronic allograft dysfunction
RNA viremia concentrations in long-term renal transplant patients in renal transplantation: the MYSS follow-up randomized, controlled
after introduction of mycophenolate mofetil. Transplantation clinical trial. J Am Soc Nephrol 2007;18(6):1973–85.
2000;69(5):991–4. 153. Tedesco Silva H, Cibrik D, Johnston T, et al. Everolimus plus reduced-
130. Maes BD, van Pelt JF, Peeters JC, et al. The effect of mycophenolate exposure CsA versus mycophenolic acid plus standard-exposure
mofetil on hepatitis B viral load in stable renal transplant recipients CsA in renal-transplant recipients. Am J Transplant 2010;10(6):
with chronic hepatitis B. Transplantation 2001;72(6):1165–6. 1401–13.
131. Luan FL, Schaubel DE, Zhang H, et al. Impact of immunosuppressive 154. Sommerer C, Glander P, Arns W, et al. Safety and efficacy of inten-
regimen on survival of kidney transplant recipients with hepatitis C. sified versus standard dosing regimens of enteric-coated mycophe-
Transplantation 2008;85(11):1601–6. nolate sodium in de novo renal transplant patients. Transplantation
132. Funch DP, Ko HH, Travasso J, et al. Posttransplant lymphopro- 2011;91(7):779–85.
liferative disorder among renal transplant patients in relation to 155. Neylan JF. Immunosuppressive therapy in high-risk transplant
the use of mycophenolate mofetil. Transplantation 2005;80(9): patients: dose-dependent efficacy of mycophenolate mofetil in African-
1174–80. American renal allograft recipients. U.S. Renal Transplant Mycophe-
133. Robson R, Cecka JM, Opelz G, et al. Prospective registry-based obser- nolate Mofetil Study Group. Transplantation 1997;64(9):1277–82.
vational cohort study of the long-term risk of malignancies in renal 156. Meier-Kriesche HU, Ojo AO, Leichtman AB, et al. Effect of mycophe-
transplant patients treated with mycophenolate mofetil. Am J Trans- nolate mofetil on long-term outcomes in African American renal
plant 2005;5(12):2954–60. transplant recipients. J Am Soc Nephrol 2000;11(12):2366–70.
134. Kasiske BL, Snyder JJ, Gilbertson DT, et al. Cancer after kidney trans- 157. Miller J, Mendez R, Pirsch JD, et al. Safety and efficacy of tacrolimus
plantation in the United States. Am J Transplant 2004;4(6):905–13. in combination with mycophenolate mofetil (MMF) in cadaveric
135. Kauffman HM, Cherikh WS, Cheng Y, et al. Maintenance immu- renal transplant recipients. FK506/MMF Dose-Ranging Kidney
nosuppression with target-of-rapamycin inhibitors is associated Transplant Study Group. Transplantation 2000;69(5):875–80.
with a reduced incidence of de novo malignancies. Transplantation 158. Nankivell BJ, Borrows RJ, Fung CL, et al. The natural history of chronic
2005;80(7):883–9. allograft nephropathy. N Engl J Med 2003;349(24):2326–33.
136. Stallone G, Schena A, Infante B, et al. Sirolimus for Kaposi’s sar- 159. Nankivell BJ, Wavamunno MD, Borrows RJ, et al. Mycophenolate
coma in renal-transplant recipients. N Engl J Med 2005;352(13): mofetil is associated with altered expression of chronic renal trans-
1317–23. plant histology. Am J Transplant 2007;7(2):366–76.
137. Downs SM. Induction of meiotic maturation in vivo in the mouse by 160. Meier-Kriesche HU, Steffen BJ, Hochberg AM, et al. Long-term use
IMP dehydrogenase inhibitors: effects on the developmental capacity of mycophenolate mofetil is associated with a reduction in the inci-
of ova. Mol Reprod Dev 1994;38(3):293–302. dence and risk of late rejection. Am J Transplant 2003;3(1):68–73.
138. Sifontis NM, Coscia LA, Constantinescu S, et al. Pregnancy outcomes 161. Glicklich D, Gupta B, Schurter-Frey G, et al. Chronic renal allograft
in solid organ transplant recipients with exposure to mycophenolate rejection: no response to mycophenolate mofetil. Transplantation
mofetil or sirolimus. Transplantation 2006;82(12):1698–702. 1998;66(3):398–9.
230 Kidney Transplantation: Principles and Practice

162. Gonzalez Molina M, Seron D, Garcia del Moral R, et al. Mycopheno- 182. Ekberg H, Grinyó J, Nashan B, et al. Cyclosporine sparing with myco-
late mofetil reduces deterioration of renal function in patients with phenolate mofetil, daclizumab and corticosteroids in renal allograft
chronic allograft nephropathy. a follow-up study by the Spanish recipients: the Caesar Study. Am J Transplant 2007;7(3):560–70.
Cooperative Study Group of Chronic Allograft Nephropathy. Trans- 183. Ekberg H, Bernasconi C, Tedesco-Silva H, et al. Calcineurin inhibitor
plantation 2004;77(2):215–20. minimization in the Symphony study: observational results 3 years
163. Gaston RS, Cecka JM, Kasiske BL, et al. Evidence for antibody-mediated after transplantation. Am J Transplant 2009;9(8):1876–85.
injury as a major determinant of late kidney allograft failure. Transplan- 184. Suwelack B, Gerhardt U, Hohage H. Withdrawal of cyclosporine or
tation 2010;90(1):68–74. tacrolimus after addition of mycophenolate mofetil in patients with
164. Sellarés J, de Freitas DG, Mengel M, et al. Understanding the causes chronic allograft nephropathy. Am J Transplant 2004;4(4):655–
of kidney transplant failure: the dominant role of antibody-mediated 62.
rejection and nonadherence. Am J Transplant 2012;12(2):388–99. 185. Schnuelle P, van der Heide JH, Tegzess A, et al. Open randomized
165. Johnson C, Ahsan N, Gonwa T, et al. Randomized trial of tacrolimus trial comparing early withdrawal of either cyclosporine or mycophe-
(Prograf) in combination with azathioprine or mycophenolate mofetil nolate mofetil in stable renal transplant recipients initially treated
versus cyclosporine (Neoral) with mycophenolate mofetil after cadav- with a triple drug regimen. J Am Soc Nephrol 2002;13(2):536–43.
eric kidney transplantation. Transplantation 2000;69(5):834–41. 186. Hricik DE, Formica RN, Nickerson P, et al. Adverse outcomes of
166. Bunnapradist S, Daswani A, Takemoto SK. Graft survival following tacrolimus withdrawal in immune-quiescent kidney transplant
living-donor renal transplantation: a comparison of tacrolimus and recipients. J Am Soc Nephrol 2015;26(12):3114–22.
cyclosporine microemulsion with mycophenolate mofetil and ste- 187. Tran HT, Acharya MK, McKay DB, et al. Avoidance of cyclospo-
roids. Transplantation 2003;76(1):10–5. rine in renal transplantation: effects of daclizumab, mycophenolate
167. Ciancio G, Burke GW, Gaynor JJ, et al. A randomized long-term trial mofetil, and steroids. J Am Soc Nephrol 2000;11(10):1903–9.
of tacrolimus and sirolimus versus tacrolimus and mycophenolate 188. Theodorakis J, Schneeberger H, Illner WD, et al. Nephrotoxicity-free,
mofetil versus cyclosporine (NEORAL) and sirolimus in renal trans- mycophenolate mofetil-based induction/maintenance immunosup-
plantation: drug interactions and rejection at one year. Transplanta- pression in elderly recipients of renal allografts from elderly cadav-
tion 2004;77(2):244–51. eric donors. Transplant Proc 2000;32(Suppl. 1A):9S–11S.
168. Mendez R, Gonwa T, Yang HC, et al. A prospective, randomized 189. Grinyó JM, Gil-Vernet S, Cruzado JM, et al. Calcineurin inhibitor-free
trial of tacrolimus in combination with sirolimus or mycophenolate immunosuppression based on antithymocyte globulin and myco-
mofetil in kidney transplantation: results at 1 year. Transplantation phenolate mofetil in cadaveric kidney transplantation: results after
2005;80(3):303–9. 5 years. Transpl Int 2003;16(11):820–7.
169. Augustine JJ, Chang PC, Knauss TC, et al. Improved renal function 190. Glotz D, Charpentier B, Abramovicz D, et al. Thymoglobulin induc-
after conversion from tacrolimus/sirolimus to tacrolimus/myco- tion and sirolimus versus tacrolimus in kidney transplant recipients
phenolate mofetil in kidney transplant recipients. Transplantation receiving mycophenolate mofetil and steroids. Transplantation
2006;81(7):1004–9. 2010;89(12):1511–7.
170. Kreis H, Cisterne JM, Land W, et al. Sirolimus in association with myco- 191. Ahsan N, Hricik D, Matas A, et al. Prednisone withdrawal in kidney
phenolate mofetil induction for the prevention of acute graft rejection transplant recipients on cyclosporine and mycophenolate mofetil—
in renal allograft recipients. Transplantation 2000;69(7):1252–60. a prospective randomized study. Steroid Withdrawal Study Group.
171. Schena FP, Pascoe MD, Alberu J, et al. Conversion from calcineu- Transplantation 1999;68(12):1865–74.
rin inhibitors to sirolimus maintenance therapy in renal allograft 192. Vincenti F, Schena FP, Paraskevas S, et al. A randomized, multi-
recipients: 24-month efficacy and safety results from the CONVERT center study of steroid avoidance, early steroid withdrawal or stan-
trial. Transplantation 2009;87(2):233–42. dard steroid therapy in kidney transplant recipients. Am J Transplant
172. Mjörnstedt L, Sørensen SS, von Zur Mühlen B, et al. Improved renal 2008;8(2):307–16.
function after early conversion from a calcineurin inhibitor to evero- 193. Squifflet JP, Vanrenterghem Y, van Hooff JP, et al. Safe with-
limus: a randomized trial in kidney transplantation. Am J Transplant drawal of corticosteroids or mycophenolate mofetil: results of a
2012;12(10):2744–53. large, prospective, multicenter, randomized study. Transplant Proc
173. Budde K, Lehner F, Sommerer C, et al. Conversion from cyclosporine 2002;34(5):1584–6.
to everolimus at 4.5 months posttransplant: 3-year results from the 194. Vanrenterghem Y, Lebranchu Y, Hené R, et al. Double-blind com-
randomized ZEUS study. Am J Transplant 2012;12(6):1528–40. parison of two corticosteroid regimens plus mycophenolate mofetil
174. Weir MR, Mulgaonkar S, Chan L, et al. Mycophenolate mofetil- and cyclosporine for prevention of acute renal allograft rejection.
based immunosuppression with sirolimus in renal transplanta- Transplantation 2000;70(9):1352–9.
tion: a randomized, controlled Spare-the-Nephron trial. Kidney Int 195. Rostaing L, Cantarovich D, Mourad G, et al. Corticosteroid-free
2011;79(8):897–907. immunosuppression with tacrolimus, mycophenolate mofetil, and
175. Vincenti F, Larsen C, Durrbach A, et al. Costimulation blockade with daclizumab induction in renal transplantation. Transplantation
belatacept in renal transplantation. N Engl J Med 2005;353(8):770–81. 2005;79(7):807–14.
176. Larson TS, Dean PG, Stegall MD, et al. Complete avoidance of calci- 196. Woodle ES, First MR, Pirsch J, et al. A prospective, randomized,
neurin inhibitors in renal transplantation: a randomized trial compar- double-blind, placebo-controlled multicenter trial comparing early
ing sirolimus and tacrolimus. Am J Transplant 2006;6(3):514–22. (7 day) corticosteroid cessation versus long-term, low-dose cortico-
177. Vincenti F, Larsen CP, Alberu J, et al. Three-year outcomes from BEN- steroid therapy. Ann Surg 2008;248(4):564–77.
EFIT, a randomized, active-controlled, parallel-group study in adult 197. Hanaway MJ, Woodle ES, Mulgaonkar S, et al. Alemtuzumab
kidney transplant recipients. Am J Transplant 2012;12(1):210–7. induction in renal transplantation. N Engl J Med 2011;364(20):
178. Vincenti F. Belatacept and long-term outcomes in kidney transplan- 1909–19.
tation. N Engl J Med 2016;374(26):2600–1. 198. Bennett WM. Immunosuppression with mycophenolic acid: one size
179. Adams AB, Goldstein J, Garrett C, et al. Belatacept combined with does not fit all. J Am Soc Nephrol 2003;14(9):2414–6.
transient calcineurin inhibitor therapy prevents rejection and pro- 199. Kuypers DR, Claes K, Evenepoel P, et al. Clinical efficacy and toxicity
motes improved long-term renal allograft function. Am J Transplant profile of tacrolimus and mycophenolic acid in relation to combined
2017;17(11):2922–36. Available online at: https://doi.org/10.1111/ long-term pharmacokinetics in de novo renal allograft recipients.
ajt.14353. Clin Pharmacol Ther 2004;75(5):434–47.
180. Grinyó J, Charpentier B, Pestana JM, et al. An integrated safety pro- 200. Le Meur Y, Borrows R, Pescovitz MD, et al. Therapeutic drug moni-
file analysis of belatacept in kidney transplant recipients. Transplan- toring of mycophenolates in kidney transplantation: report of
tation 2010;90(12):1521–7. The Transplantation Society consensus meeting. Transplant Rev
181. Pascual M, Curtis J, Delmonico FL, et al. A prospective, random- 2011;25(2):58–64.
ized clinical trial of cyclosporine reduction in stable patients 201. van Gelder T, Tedesco Silva H, de Fijter JW, et al. Renal transplant
greater than 12 months after renal transplantation. Transplanta- patients at high risk of acute rejection benefit from adequate expo-
tion 2003;75(9):1501–5. sure to mycophenolic acid. Transplantation 2010;89(5):595–9.
16 Steroids
SIMON R. KNIGHT

CHAPTER OUTLINE Introduction Pancreatitis


Mechanism of Action Skin Changes
Steroid Resistance Peptic Ulceration
Dosage Acute Abdomen
Treatment of Acute Rejection Adrenal Insufficiency
Side Effects Steroid Withdrawal and Avoidance
Cushingoid Facies Early Experience
Wound Healing Contemporary Immunosuppression
Growth Retardation Effect of Induction Agent
Diabetes Graft and Patient Survival
Hyperlipidemia Sensitized Recipients
Bone Disease Steroid Withdrawal in Children
Obesity Complete Steroid Avoidance
Hypertension Conclusions
Psychiatric Disturbance
Cataracts

Introduction Mechanism of Action


When the first renal transplants were being performed Steroids are administered as prednisone or prednisolone.
in the early 1960s the immunosuppressive properties of These agents are absorbed rapidly from the gut, and peak
corticosteroids were already noted, and steroids were suc- plasma concentrations occur 1 to 3 hours after adminis-
cessfully used to reverse episodes of acute rejection.1 In his tration. The mechanism of action of steroids is extremely
1964 book, Starzl described the use of steroids as a “pre- complex and is still not understood fully.6–8 Steroids are
treatment” alongside azathioprine as prophylaxis against antiinflammatory as well as being immunosuppressive. It
rejection, based on the premise that rejection was almost was first noted by Billingham et al.9 that cortisone would
inevitable with azathioprine alone.2 This work was soon produce a modest prolongation of the life of skin allografts
followed by groups from Virginia, London, and Cleveland, in the rabbit. In the treatment of acute rejection, it is prob-
with the use of steroids in combination with azathioprine ably the antiinflammatory activity that produces the imme-
from the time of transplantation and increased doses dur- diate response, whereas when used prophylactically it is the
ing episodes of acute rejection.3–5 This combination of aza- immunosuppressive activity that is predominant. A small
thioprine and steroids became the mainstay of transplant randomized trial comparing prednisolone with a nonsteroi-
immunosuppression for almost 20 years. dal antiinflammatory agent (ibuprofen) showed a higher
From the beginning of this so-called azathioprine era, rate of rejection in the patients receiving the nonsteroidal
arbitrarily large doses of steroids were given from the time agent, confirming that the antiinflammatory effect of ste-
of transplantation with a gradual reduction over 6 to 12 roids is not its major role in renal transplantation.10
months to maintenance levels. The high doses of steroids Steroids are metabolized in the liver, where prednisone is
used with azathioprine were responsible for most of the mor- converted to prednisolone. Although it has been estimated
bidity of transplantation (discussed later). It was not until that the bioavailability of prednisone is approximately 80%
the 1970s that a series of randomized trials as well as obser- of that achieved by prednisolone, no evidence exists in prac-
vational studies slowly led to the realization that low-dose tice that there is a difference in outcome between predni-
steroids were as effective as high-dose steroids in preventing sone (used most commonly in the US) or prednisolone (used
rejection and that there was a major reduction in steroid most commonly in Europe).11,12 The half-life of steroids is
complications of transplantation with low-dose regimens. short—about 60 minutes for prednisone and 200 minutes
With the introduction of newer, more potent induction for prednisolone. These half-lives are increased substan-
and maintenance agents, there has been a great deal of tially in the presence of hepatic dysfunction and are shorter
interest over recent years in further reducing steroid doses in the presence of drugs such as phenytoin and rifampicin
and either withdrawing them from maintenance immuno- that induce hepatic enzymes. There is no evidence that
suppressive regimens or avoiding them altogether. these interactions have produced significant problems in
231
232 Kidney Transplantation: Principles and Practice

clinical practice. It has also been shown that the clearance downregulation by glucocorticoid exposure, negative inhi-
of prednisolone is slower in patients on cyclosporine com- bition by the beta-isoform of the glucocorticoid receptor, or
pared with patients on azathioprine.13 A later study sug- inhibition of the alpha isoform of the receptor by the pro-
gested, however, that cyclosporine did not influence the inflammatory transcription factor NK-κB in inflammatory
metabolism of methylprednisolone, but the authors noted conditions.29
a considerable variation of the metabolism of methylpred-
nisolone among patients.14 The time- and dose-dependent
induction of UDP-glucuronosyltransferase activity by ste- Dosage
roids may increase the clearance of mycophenolic acid,
reducing exposure to mycophenolate. Cattaneo et al. have When used prophylactically with azathioprine, steroids
demonstrated that as steroids are tapered over the postoper- were used initially in high doses (e.g., 100 mg/day), reduc-
ative period, the mycophenolic acid area-under-the-curve ing to a maintenance dose of 20 mg/day over 6 to 9 months.
increases.15 The pharmacokinetics of prednisolone during McGeown and coworkers were the first to report consis-
sirolimus therapy have also been studied, with some evi- tently excellent graft survival with a low incidence of ste-
dence for a minor interaction between sirolimus and pred- roid-related complications using a lower prednisolone dose
nisolone in some patients.16 of 20 mg/day given orally as a single morning dose, with a
Steroids do have a significant effect in vitro on T cell pro- further reduction occurring at 6 months to a baseline main-
liferation, blocking interleukin-2 production.17 A variety tenance dose of 10 mg/day.30 Because most of the Belfast
of other actions may augment their immunosuppressive patients had had bilateral nephrectomies and all had had
activity (e.g., preventing the induction of interleukin-1 and more than 100 blood transfusions before transplantation, it
interleukin-6 genes in macrophages).18,19 Its antiinflam- was not clear whether the excellent results were related to
matory activity is perhaps mediated by the inhibition of the low dosage of steroids or to a transfusion effect, which
migration of monocytes to areas of inflammation,7 and this was recognized widely as an important factor in improving
same antiinflammatory activity has a marked deleterious graft outcome in the azathioprine era.
effect on wound healing. Trials of low-dose steroids versus high-dose steroids were
carried out in Oxford, then in many other centers, all of
which showed not only that low-dose steroids were as effec-
Steroid Resistance tive as high-dose steroids in preventing rejection but also
that there was a significant reduction in steroid-related
The sensitivity of individuals to steroid therapy is highly complications in patients receiving low-dose steroids.31–39
variable. A study in healthy volunteers demonstrated a The results of these trials led quickly to the wide adoption of
wide interindividual variation in the inhibition of lympho- low-dose steroid regimens with azathioprine. The results of
cyte proliferation by steroids.20 Steroid resistance is fre- a large multicenter trial reported by D’Apice et al.,40 dem-
quently seen in patients with inflammatory conditions and onstrated that low-dose steroids were only equally effective
has been shown to correlate well with in vitro measure- as high-dose steroids in preventing rejection if therapeutic
ments of lymphocyte steroid sensitivity in patients suffering doses of azathioprine were used (i.e., >2 mg/kg/day).
with rheumatoid arthritis,21 ulcerative colitis,22 asthma,23 With the introduction of cyclosporine, steroids remained
and systemic lupus erythmatosus.24 in use with or without azathioprine. In general, low-dose
In vitro studies of lymphocyte steroid sensitivity have steroid protocols were continued, although there was a ten-
demonstrated a higher incidence of resistance in patients dency, particularly in North America, to go back toward
with chronic renal failure than in healthy volunteers higher steroid dosage regimens in the first few weeks post-
(52.9% vs. 3.8%).25 In renal transplant recipients, Lang- transplantation. This was a relatively transient practice
hoff and colleagues have demonstrated that pretransplant and now, with modern immunosuppressive protocols, not
in vitro measurements of lymphocyte sensitivity are predic- only are low-dose steroids the norm but indeed discontinua-
tive of graft survival at 1 year in patients coadministered tion of steroids is becoming increasingly possible (see later).
azathioprine, but less so in those receiving cyclosporine.26 Whether steroids should be given as a single daily dose
These results have been confirmed in vivo, with significantly in the morning or in divided doses has not been resolved.
higher sensitivity to methylprednisolone seen in those Because of the short half-life of prednisone and predniso-
patients with graft function at 6 months compared with lone, divided doses may be more rational, but it could be
patients with graft failure.27 This difference in sensitivity is argued that a single morning daily dose would be more
smaller in cyclosporine-treated patients than those receiv- appropriate, taking into account the diurnal rhythm of glu-
ing azathioprine, suggesting that this effect may in part be cocorticoid metabolism.41,42 There is no clinical evidence
offset by the use of newer, more potent immunosuppres- that one or the other protocol is more effective or less likely
sant agents. Dialysis patients demonstrating steroid resis- to produce side effects.
tance have an increased risk of acute rejection and chronic For many years, maintenance doses of prednisone or
allograft nephropathy posttransplant.28 Interestingly, prednisolone of 10 mg/day were standard therapy in asso-
reduced lymphocyte prednisolone sensitivity correlates ciation with azathioprine. In patients with long-surviv-
with impaired sensitivity to cyclosporine and tacrolimus, ing grafts with good function, steroid dosages have been
which may play a role in the high risk of allograft rejection reduced to 5 or 6 mg/day. It is unlikely, however, that
in these patients.25 many patients who are on long-term azathioprine and ste-
A number of potential mechanisms for this resis- roids would be able to have their steroid dosage reduced to
tance to steroids have been suggested, including receptor much less than 5 mg/day. Attempts in the past to withdraw
16 • Steroids 233

steroids have often led to the onset of rejection when doses trial, Stromstrad et al.54 failed to show any therapeutic ben-
of less than 5 mg/day are reached. This is important to note efit of a 30 mg/kg bolus over a 3 mg/kg bolus, and similarly
as there are many long surviving patients still on azathio- Lui et al.55 failed to show any benefit of a bolus of 15 mg/kg
prine and steroids. It should also be remembered that when of body weight over a bolus of 3 mg/kg.
patients have been on steroids for many years, their adre-
nocortical function may not recover from the long-standing
suppression as the steroid dose is reduced (see later). Side Effects
Alternate-day steroid therapy for maintenance has also
been used widely, especially in children, in an attempt to The side effects of continuous steroid therapy are numer-
reduce side effects, particularly growth retardation.43–48 In ous (Table 16.1). High-dose steroids were responsible for
children, alternate-day therapy may be associated with a most complications of renal transplantation in the azathi-
greater incidence of rejection, but this is probably not the oprine era, which have reduced markedly with the more
case in adults. A small randomized trial of alternate-day recent widespread use of low-dose steroids. In a study of
therapy failed to show any benefit over daily steroids, how- the cost of steroid side effects over 10 years in a cohort of
ever.47 Alternate-day therapy may lead to greater problems 50 patients, the additional cost per patient attributable to a
with respect to compliance, in contrast to a daily regimen steroid complication was assessed at $5300 (US dollars).56
of steroids. Cost analysis in a randomized controlled trial of early ste-
It has been and still is common practice to administer a roid withdrawal from Egypt has shown a 2.9-fold increase
bolus of methylprednisolone prophylactically during the in management costs for steroid-related morbidities in the
transplant operation with the aim of increasing immuno- steroid maintenance arm.57
suppression and perhaps preventing delayed graft function,
but a randomized prospective trial of bolus methylpredniso-
CUSHINGOID FACIES
lone versus placebo at the time of surgery did not show any
benefit of the high perioperative intravenous dose of meth- Cushingoid facies used to be the hallmark of a renal trans-
ylprednisolone.49 Nevertheless it remains standard practice plant patient: a moon face, buffalo hump, acne, obese torso,
whatever the immunosuppressive regimen is to be. and thin, easily bruised skin, all representing the cumulative
effect of high-dose steroids. With lower-dose steroids, Cushin-
goid facies is seen much less often, although most patients
Treatment of Acute Rejection show modest changes in their facies in the early months post-
transplantation. Most patients on low-dose steroids, which is
Steroids in high doses are the first approach to the treatment the normal practice now with cyclosporine or tacrolimus,
of an acute rejection episode. Early experience involved have relatively minimal facial changes related to steroids.
either increasing the oral dosage of steroids to high levels
(e.g., 200 mg/day for 3 days), with a rapid reduction over
WOUND HEALING
10 days to the dosage levels of steroids being given before
the acute rejection episode, or boluses of intravenous meth- The antiinflammatory activity of steroids leads to poor
ylprednisolone (e.g., 0.5–1 g/day for 3–5 days). Probably wound healing. In the days of high-dose steroids, this was
both approaches are equally effective. In an early random- a major problem, influencing the healing not only of the
ized prospective trial in Oxford, however, high intravenous incision, but also of the ureterovesical reconstruction. With
doses were as effective as high oral doses in reversing rejec- low-dose steroids, poor wound healing is no longer a major
tion, but there was a definite suggestion that steroid-related problem.
complications were lower in those who received intrave-
nous therapy.50 In a randomized study in children, a high
GROWTH RETARDATION
intravenous dose of methylprednisolone (600 mg/m2 daily
for 3 days) was no more effective than low oral doses of Growth retardation is of particular concern in children after
prednisolone, reversing rejection in 70% as opposed to 72% renal transplantation. With modern lower doses of steroids
of episodes.51 growth retardation is less of a problem.58 As already dis-
The most common form of high-dose intravenous ther- cussed, the use of alternate-day steroids may reduce growth
apy to treat acute rejection has been 1 g of methylpred- retardation, and further steroid reduction and withdrawal
nisolone given intravenously as a single bolus daily for 3 have also been successful in allowing catch-up growth in
days. The intravenous bolus should be administered slowly some studies (see later).
over 5 minutes because the sudden injection of the bolus
can lead to cardiac arrhythmias.52 It is probable that 1 g of
methylprednisolone is a much greater dose than required; TABLE 16.1 Side Effects of Steroids After Renal
in Oxford for many years now we have used 0.5 g of meth- Transplantation
ylprednisolone daily intravenously for 3 days, whereas the Cushingoid facies Hypertension
Stockholm unit has used 0.25 g daily intravenously for Wound healing Psychiatric disturbance
3 days. The lower intravenous doses do not appear to be Growth retardation Cataracts
associated with any greater incidence of steroid-resistant Diabetes Pancreatitis
rejection, as originally suggested by a prospective trial of Hyperlipidemia Skin changes
Bone disease Peptic ulceration
high-dose versus low-dose intravenous steroids to treat Obesity
rejection.53 Similarly in a small double-blind, randomized
234 Kidney Transplantation: Principles and Practice

DIABETES
problem in the calcineurin inhibitor (CNI) era as cyclospo-
Glycosuria and insulin-dependent and non–insulin-dependent rine and tacrolimus both lead to an increased incidence,
diabetes are common after transplantation. The occurrence of although this may be less of a problem with tacrolimus.60
diabetes is related, in part, to steroid usage59 but it has become Withdrawal of steroids leads to improvements in the lipid
more common with the concomitant use of cyclosporine and profile (see later).
tacrolimus, both of which can induce diabetes independently
of steroids. In the presence of these two agents, the use of ste-
BONE DISEASE
roids augments the potential for diabetes, and often patients
who become diabetic on cyclosporine or tacrolimus have a Bone disease is a common and major problem posttrans-
regression of the diabetes when steroid therapy is discontinued. plantation, especially in the postmenopausal woman.61–66
In the days of high-dose steroid therapy posttransplanta-
HYPERLIPIDEMIA tion, avascular necrosis of bones, particularly of the head
of the femur, was common, occurring with an incidence of
Hypercholesterolemia and hypertriglyceridemia are associ- approximately 10% to 15% within 2 years of transplanta-
ated with steroid use. Hyperlipidemia has become a greater tion (Fig. 16.1). All the evidence suggests that this incidence

A B

C
Fig. 16.1 The progression of avascular necrosis of the head of the femur. (A) Normal radiograph on first complaint of pain 1-year posttransplanta-
tion, (B) 5 months later, and (C) 20 months later. At this time, a hip replacement was performed.
16 • Steroids 235

was due to a cumulative effect of steroid dosage. As low- CATARACTS


dose steroid protocols were introduced, the incidence of
avascular necrosis decreased dramatically. However, the Steroid-related cataracts are common after renal transplan-
cumulative dose of steroids received by a patient on a high- tation, occurring in approximately 25% of patients.70
dose steroid regimen, as opposed to a low-dose regimen, is
not that much higher after 6 months. PANCREATITIS
Osteoporosis is associated with steroid therapy. In a ran-
domized study, Hollander and colleagues showed that ver- Acute pancreatitis occurs with a much greater incidence
tebral bone density was increased significantly in patients after renal transplantation than would be expected. Aza-
discontinuing steroids.67 Similar evidence was reported thioprine and steroids have been associated with acute
by Aroldi et al.68 in a randomized study of three different pancreatitis. The pancreatitis is probably related to overall
immunosuppressive protocols and vertebral bone den- immunosuppression and is often severe.71 The clinical fea-
sity. These investigators showed that lumbar bone density tures of acute pancreatitis can be masked to some extent by
decreased significantly in patients receiving cyclosporine steroids.
and steroids but increased significantly in patients receiv-
ing cyclosporine alone without steroids. Meta-analysis of SKIN CHANGES
studies in transplant recipients has demonstrated a signifi-
cant reduction in the risk of fractures and increase in bone Long-term steroids produce typical skin changes in renal
mineral density with the use of bisphosphonates or vitamin transplant patients, the skin being thin, atrophic, easily
D analogs, suggesting that the use of these agents should bruised, and susceptible to knocks. A syndrome known as
be considered, particularly in recipients on long-term transplant leg is associated with long-term steroid usage;
steroids.69 this occurs, for example, when a patient bumps into a chair
Many patients coming to renal transplantation have or a table (a trivial injury), and a flap of skin is stripped or
a degree of secondary hyperparathyroidism, and bone elevated from the lower leg.
changes related to the hyperparathyroidism are enhanced
by steroid therapy. Much more aggressive approaches PEPTIC ULCERATION
to parathyroidectomy in patients with renal failure are
being taken by most units now before transplantation. Although it is debatable whether steroids lead to develop-
In patients after transplantation with raised parathor- ment of peptic ulceration, most units use prophylactic H2
mone levels, early parathyroidectomy also should be antagonists or proton pump inhibitors in the early months
considered. posttransplantation, when steroid doses are at their high-
est. The advent of low-dose steroid therapy has been associ-
ated with a dramatic diminution in the incidence of peptic
OBESITY
ulceration after transplantation.
Steroid therapy leads to a marked increase in appetite,
and without any dietary restrictions after transplanta- ACUTE ABDOMEN
tion, all patients tend to gain weight, which is in addition
to a weight increase resulting from salt and water reten- In all renal transplant patients who present with an acute
tion. Many patients become obese (body mass index >30), abdomen, steroids may mask the symptoms noted by the
and this adds to the risks of poor survival. Every attempt patient. If this fact is not remembered, diagnosis of diver-
should be made to advise patients from the time of trans- ticulitis or a perforated peptic ulcer may be delayed with
plantation to restrict calorie intake carefully because once disastrous results.
patients have gained weight in the presence of steroid
therapy, it is extremely difficult for them to reduce their ADRENAL INSUFFICIENCY
weight.
As in other conditions in which long-term corticosteroids
are used, there is a risk of adrenal insufficiency with ces-
HYPERTENSION
sation of therapy or during periods of stress, trauma, sur-
Hypertension after transplantation is common and is gery, or acute illness. A recent meta-analysis suggests that
related in part to steroids as well as CNI use. In steroid with- the absolute risk of adrenal insufficiency in renal trans-
drawal protocols, hypertension improves once steroids are plant recipients is 56.2% (95% confidence interval [CI]
discontinued (see later). 42.9%–68.6%).72 Although higher doses and longer-term
use increase the risk, the effect is not entirely predictable,
and the risk of insufficiency should always be considered in
PSYCHIATRIC DISTURBANCE
patients taking maintenance steroids.
Psychiatric disturbance is evident in patients on steroids in
two ways. In the early days posttransplantation, particu-
larly with the need for high-dose steroids to treat rejection,
Steroid Withdrawal and
significant psychiatric mood changes may be observed. Avoidance
Later, when steroids are being withdrawn or reduced to
low doses, psychiatric mood changes, especially depression, The side effects of steroids as outlined previously have
may occur. generated a great deal of interest in withdrawal or even
236 Kidney Transplantation: Principles and Practice

complete avoidance of these agents after renal transplanta- from month 6.79 Randomization and blinding were main-
tion. One of the major causes of death with a functioning tained for the duration of the 5-year follow-up. Although
graft after renal transplantation is cardiovascular disease, the study demonstrated an excess of mild, steroid-sensitive
so the effect of long-term steroid use on cardiovascular risk acute rejection in the withdrawal arm, 5-year patient and
factors such as hypertension, hyperlipidemia, and post- graft survival were unaffected. Benefits were seen in cardio-
transplant diabetes is likely to be important.73 Any advan- vascular risk factors such as triglycerides, insulin require-
tage seen in terms of cardiovascular risk must be balanced ments, and weight gain, leading the authors to conclude
against the reduction in immunosuppression and potential that early steroid withdrawal is safe in the longer term in
detriment to graft function, and such concerns have led to patients treated with a tacrolimus and MMF regimen.
a large number of clinical trials investigating the risk and It was first noted by Pescovitz and colleagues that siroli-
benefit of steroid withdrawal protocols. mus may aid in the withdrawal of steroids from a calcineu-
rin-based regimen.80 Further observational studies appear
to support this, and the use of sirolimus may also allow
EARLY EXPERIENCE
steroid withdrawal alongside reduction in the exposure to
In early patients treated with steroids and azathioprine, CNIs.81–84
attempts to withdraw steroids resulted in almost inevi- The use of newer immunosuppressive agents such as
table acute rejection when the dose fell below 5 to 6 mg/ MMF or sirolimus may also allow the safe withdrawal of ste-
day.74,75 The introduction of cyclosporine led to renewed roids earlier than previously seen with cyclosporine-based
interest in such protocols, and spawned a number of clini- regimens. Kumar and colleagues have reported a 3-year
cal trials investigating steroid withdrawal at varying times analysis of a large trial of 300 patients receiving basiliximab
posttransplant. An early meta-analysis of seven studies in induction, a CNI, and MMF or sirolimus, in which patients
cyclosporine-treated patients by Hricik and colleagues dem- were randomized to have steroids withdrawn on day 2 or to
onstrated an increased risk of acute rejection, but with no continue steroids.85 There was no difference in graft func-
detriment to patient or graft survival.76 Five of the seven tion, patient and graft survival, biopsy proven acute rejec-
included studies did not utilize triple therapy with aza- tion, or chronic allograft nephropathy. The incidence of
thioprine (cyclosporine alone) and in four studies steroids new-onset diabetes was lower in the steroid-free group.
were avoided completely from the time of transplantation. A study by Gelens and colleagues attempted to combine
Furthermore, only one study reported follow-up beyond 2 early steroid withdrawal with a CNI-free maintenance
years posttransplant. This was the Canadian Multicentre regimen.86 Patients were randomized to receive tacrolimus
Cyclosporine trial, which demonstrated superior long-term and sirolimus; tacrolimus and MMF; or daclizumab induc-
graft survival in patients who continued taking steroids.77 tion, sirolimus, and MMF. Steroids were withdrawn after
The results of this meta-analysis led to concerns about 2 days in all patients. The trial was halted after an interim
the early withdrawal of steroids from cyclosporine-based analysis demonstrated an unacceptably high incidence of
regimens, leading to further randomized controlled tri- acute rejection in the CNI-free group. Thus it would appear
als investigating late withdrawal in patients with stable that even with modern immunosuppressant agents and
renal function at 1 year posttransplant.67,78 In the trial antibody induction, it is not possible to combine the com-
from Oxford, patients on triple therapy (cyclosporine, aza- plete withdrawal of CNIs with steroid withdrawal. Recent
thioprine, and steroids) randomized to stop steroids dem- attempts to combine belatacept-based immunosuppression
onstrated a 10% deterioration in renal function at 1 year, with steroid avoidance have resulted in unacceptably high
which then stabilized with no increase in graft loss.78 Both rates of acute rejection, even in the context of depleting
of these studies demonstrated beneficial effects in cardiovas- antibody induction.87 Until further evidence is generated,
cular risk factors such as hypertension and blood lipids. steroid avoidance or withdrawal cannot be recommended
in combination with belatacept-based, CNI-free regimens.
A number of meta-analyses of steroid withdrawal pro-
CONTEMPORARY IMMUNOSUPPRESSION
tocols have been published to attempt to draw conclusions
After the introduction of tacrolimus and mycophenolate from the large body of randomized trial evidence in this
mofetil (MMF) to maintenance immunosuppressive regi- area. Pascual and colleagues performed a meta-analysis
mens, a large number of randomized trials with various of six such trials, four with MMF and cyclosporine and two
combinations of induction and maintenance agents, as with MMF and tacrolimus.88 Although the risk of acute
well as varying times of steroid withdrawal, have been pub- rejection was increased just over twofold when steroids
lished in the literature. In general the trend has been back were withdrawn, there was no significant difference in the
toward earlier tapering and withdrawal of steroids, with incidence of graft failure. Unfortunately, this meta-analysis
withdrawal as early as 1 week posttransplant in a num- did not differentiate the relative steroid-sparing potential of
ber of studies. One of the major concerns, given the excess cyclosporine and tacrolimus. A similar meta-analysis from
acute rejection seen in the early steroid withdrawal trials Tan and colleagues also demonstrated an increase in the
with cyclosporine, is the effect of the reduction of immuno- risk of acute rejection, balanced by a reduction in the risk of
suppression on long-term graft and patient outcomes. Per- opportunistic and urinary tract infections in a small subset
haps the most impressive study to date addressing this issue of the included trials.89
is the Astellas Steroid Withdrawal study from Woodle and A comprehensive meta-analysis from our own group
colleagues, in which patients receiving tacrolimus, MMF, included all steroid withdrawal and avoidance studies
and steroids were randomized at day 7 to withdraw steroids over a 27-year period, incorporating 34 studies and 5637
or continue a long-term maintenance dose of 5 mg/day patients.90 Overall analysis confirmed an increase in acute
16 • Steroids 237

rejection with steroid avoidance or withdrawal (Fig. 16.2; of hypertension (RR 0.90, P < 0.0001), new onset diabe-
relative risk [RR] 1.56, P < 0.0001), although no difference tes (RR 0.64, P = 0.0006), and hypercholesterolemia (RR
in steroid-resistant acute rejection was found, suggesting 0.76, P < 0.0001) all reduced. Interestingly, no interac-
that these are mainly steroid-sensitive mild rejection epi- tion was found between the use of induction immunosup-
sodes. Despite the increased rate of rejection and a small pression, maintenance immunosuppression, or the time of
(clinically insignificant) increase in serum creatinine, steroid withdrawal on the ability to withdraw steroids. Post
there was no difference in graft or patient survival. Signifi- hoc analysis has, however, demonstrated a dose effect, with
cant benefits in cardiovascular risk factors were identified the cardiovascular benefit seen reduced when withdrawing
with steroid avoidance or withdrawal, with the incidence lower doses of steroids.91

Study/subcategory Avoidance/withdrawal (n/N) Maintenance (n/N) RR [95% Cl]


01. Avoidance
Spanish Monotherapy Study Group 1994 30/41 18/44 1.79 [1.20,2.67]
Stiller 1983 25/33 24/36 1.14 [0.84,1.54]
Vincenti 2008 35/111 8/54 2.13 [1.06,4.27]

Subgroup total 185 134 1.52 [1.03,2.23]

02. Induction
De Vecchi 1986 19/25 13/26 1.52 [0.98,2.37]
Laftavi 2005 4/32 3/28 1.17 [0.29,4.77]
Ponticelli 1997 80/115 58/117 1.40 [1.13,1.75]
Schulak 1990 26/32 19/35 1.50 [1.06,2.12]
Kumar 2005 7/45 4/32 1.24 [0.40,3.90]
Montagnino 2005 21/65 12/68 1.83 [0.98,3.41]
Vincenti 2003 8/40 7/43 1.23 [0.49,3.08]
Vitko 2005 46/151 12/147 3.73 [2.06,6.75]
Woodle 2008 34/191 21/195 1.65 [1.00,2.74]
Vincenti 2008 30/115 8/54 1.76 [0.87,3.58]
Nematalla 2007 8/50 8/50 1.00 [0.41,2.46]

Subgroup total 861 795 1.57 [1.33,1.85]

03. Early withdrawal


Boletis 2001 0/34 0/32 NA [ NA, NA]
Aswad 1998 2/6 1/6 2.00 [0.24,16.61]
Gulanikar 1991 10/41 3/44 3.58 [1.06,12.09]
Cristinelli 1986 16/35 3/31 4.72 [1.52,14.69]
Park 1994 42/141 48/153 0.95 [0.67,1.34]
Pisani 2001 0/9 0/10 NA [NA, NA]
Sola 2002 9/46 5/46 1.80 [0.65,4.96]
Ahsan 1999 26/134 7/132 3.66 [1.65,8.14]
lsoniemi 1990 4/29 0/32 9.92 [0.56,176.45]
Vanrenterghem 2005 42/252 57/245 0.72 [0.50,1.02]
Smak Gregoor 2002 3/76 1/73 2.88 [0.31,27.08]
Vanrenterghem 2000 63/252 36/248 1.72 [1.19,2.49]

Subgroup total 1055 1052 1.82 [1.15,2.89]

04. Late withdrawal


Kacar 2004 1/31 6/30 0.16 [0.02,1.26]
Matl 2000 3/46 2/42 1.37 [0.24,7.80]
Farmer 2006 1/44 2/48 0.55 [0.05,5.81]
Pelletier 2006 3/59 3/59 1.00 [0.21,4.75]
Hollander 1997 9/42 1/42 9.00 [1.19,67.93]
Ratcliffe 1996 3/49 1/51 3.12 [0.34,29.01]

Subgroup total 271 272 1.23 [0.43,3.54]

Overall total 2372 2253 1.56 [1.31,1.87]

0.1 1.0 10.0 100.0


Relative Risk
Fig. 16.2 Forest plot to show the relative risk of acute rejection with steroid avoidance or withdrawal at various times after transplantation.
Blue boxes show treatment effect for individual studies. Red diamonds show summary treatment effect for each subgroup and overall analysis derived
by random effects analysis. Horizontal lines show 95% confidence intervals. CI, confidence interval; N, total number of patients in study arm; n, number
of patients with acute rejection; RR, relative risk. Subgroups are (1) complete steroid avoidance, (2) induction steroids (≤7 days), (3) early steroid with-
drawal (8 days to 12 months), (4) late steroid withdrawal (>12 months). (From Knight SR, Morris PJ. Steroid avoidance or withdrawal after renal transplanta-
tion increases the risk of acute rejection but decreases cardiovascular risk: A meta-analysis. Transplantation 2010;89(1):1–14.)
238 Kidney Transplantation: Principles and Practice

Analysis of data from a recent randomized controlled a subsequent analysis examining outcome only in patients
trial has shed further light on the mechanisms for increased with satisfactory renal function at 1 year found the same
acute rejection risk. Steroid avoidance and withdrawal are results. A more recent prospective study from the same
both associated with an increased expression of Th1 tran- group has confirmed a benefit in both graft and patient
script expression and decreased natural killer cell suppres- survival in renal and cardiac transplant patients in whom
sion compared with maintenance steroid regimens.92 steroids were withdrawn more than 6 months after trans-
plantation, with no increase in incidence of acute rejec-
tion (Fig. 16.3).95 Benefits were also seen in cardiovascular
EFFECT OF INDUCTION AGENT
parameters in the withdrawal group, with significantly
It would seem reasonable to hypothesize that early with- fewer patients demonstrating elevated cholesterol levels.
drawal, or even complete avoidance, of steroids would Although the number of patients and length of follow-up in
be more successful in the face of higher potency induc- this study is impressive (7 years), a major criticism is a lack
tion agents. There are surprisingly few data in support of of a randomized design, with the control cohort being retro-
this theory, with subgroup analyses from the large meta- spectively matched patients from the CTS registry.
analyses failing to demonstrate a difference between mono- Further analysis of CTS data has suggested a dose-
clonal and polyclonal antibody preparations. The recently response effect, with steroid dose at 1 year correlating with
completed HARMONY study from Germany directly com- risk of death with a functioning graft (primarily due to car-
pared rabbit ATG with basiliximab as part of a rapid steroid- diovascular and infective causes).96 This effect is seen even
withdrawal protocol, demonstrating that both induction in those patients with excellent graft function in whom ste-
regimens were safe and efficacious in a low-risk popula- roids would appear to be unnecessary.
tion.93 However, a Cochrane review of polyclonal and
monoclonal antibody induction agents has provided some SENSITIZED RECIPIENTS
evidence that alemtuzumab induction may be preferable to
ATG in the context of early steroid withdrawal, with a rela- Many of the randomized controlled trials presented previ-
tive risk for rejection of 0.57 (95% CI 0.35–0.93) at 1 year. ously were undertaken in immunologically low-risk patient
cohorts. Many units have greater reservations about minimiz-
ing immunosuppression in higher-risk patient groups. These
GRAFT AND PATIENT SURVIVAL
fears appear to be justified by an analysis of Scientific registry
Whereas none of the randomized controlled trial data sup- of transplant recipients (SRTR) data from the United States.
port an effect on graft or patient survival with steroid with- Sureshkumar and colleagues analyzed data from 42,851
drawal, registry data from the Collaborative Transplant transplants over an 8-year period, splitting the cohort into
Study (CTS)94 has suggested that patients with a function- groups by peak-reactive antibody (PRA) level.97 In the low-risk
ing graft at 1 year who had steroids withdrawn had better recipients (PRA < 30%), steroid maintenance was associated
graft and patient survival thereafter than patients remain- with higher all-cause graft loss and patient death, in keeping
ing on steroids. The increased risk of acute rejection with with the effects on death with a functioning graft seen in the
steroid withdrawal was not seen in the registry data. The CTS registry analysis earlier. In highly sensitized recipients
initial criticism of this study was that patients still on ste- (PRA > 60%), maintenance steroid use resulted in improved
roids at 1 year represented patients with poorer function as death-censored graft survival, suggesting that steroid minimi-
a result of rejection episodes or delayed graft function, but zation should be undertaken with care in this group.

Grafts surviving Patients surviving Functional surviving


100 100 100

90 90 90
% Surviving after study entry

80 80 80

70 70 70

60 60 60

50 50 50

0 0 0
0 1 2 3 4 5 6 7 0 1 2 3 4 5 6 7 0 1 2 3 4 5 6 7
Years Years Years

Study patients n  1015 Study patients n  1015 Study patients n  1015


A Controls n  3045 B Controls n  3045 C Controls n  3045
Fig. 16.3 Seven-year graft (A), patient (B), and functional graft (C) survival in renal transplant recipients after steroid withdrawal (study patients) or
steroid continuation (matched controls). (From Opelz G, Dohler B, Laux G. Long-term prospective study of steroid withdrawal in kidney and heart transplant
recipients. Am J Transplant 2005;5(4 pt 1):720–8.)
16 • Steroids 239

STEROID WITHDRAWAL IN CHILDREN and treatment of acute rejection. Particularly in high doses,
they are associated with a plethora of adverse effects and
Given the detrimental effects of long-term corticosteroids on the overall trend is now toward reduction in steroid expo-
growth, there has been some interest in steroid withdrawal sure with withdrawal or avoidance.
in pediatric transplant recipients. Benfield and colleagues Steroid withdrawal is associated with an increased
performed a double-blind, randomized trial in children 6 risk of acute rejection, but the evidence seems to suggest
months posttransplant to either continue maintenance that this excess rejection is mild and treatable, and does
steroids or withdraw gradually by 12 months.98 Baseline not appear to be detrimental to longer-term graft sur-
immunosuppression was basiliximab induction, CNI, and vival and function in low-risk patient populations. With-
sirolimus. The study was stopped by the drug safety moni- drawal has significant benefits in terms of cardiovascular
toring board after enrolment of 274 patients because of a risk, bone complications, and growth, and registry data
very high incidence of posttransplant lymphoprolifera- suggest that this may translate to a reduction in the risk
tive disorder (6.9%), probably relating to the very heavy of death with a functioning graft from cardiovascular or
baseline immunosuppression. The authors concluded that infective causes.
although the immunosuppressive regimen used allowed Although the role of complete steroid avoidance is uncer-
for the safe withdrawal of steroids, it could not be recom- tain, early withdrawal in low-risk renal transplant recipi-
mended on safety grounds. ents on a modern immunosuppressive regimen appears
Another study reporting withdrawal of steroids beyond 1 to be safe, with significant benefits to the patient. Caution
year posttransplant from a more conventional regimen has should be exercised when considering steroid minimization
demonstrated no excess in acute rejection at 2 years.99 There in recipients of high immunologic risk (PRA > 60%) or in
was an improvement in growth and reduction in cardio- conjunction with belatacept or CNI minimization.
vascular risk factors, with no detriment to graft survival or
function. Similar benefits have also been demonstrated in the References
multicenter TWIST study, in which steroids were withdrawn 1. Goodwin WE, Mims MM, Kaufman JJ. Human renal transplanta-
at 4 days and replaced with daclizumab at induction.100 tion III. Technical problems encountered in six cases of kidney
These findings are supported by a recent meta-analy- homotransplantation. Trans Am Assoc Genitourin Surg 1962;54:
sis.101 Zhang and colleagues identified five randomized 116–25.
2. Starzl TE. Pretreatment with prednisolone. In: Experience in renal
trials, concluding that steroid avoidance or withdrawal is transplantation. Philadelphia: W.B. Saunders Company; 1964.
associated with significant improvement in growth with no 3. Hume DM, Lee HM, Williams GM, et al. Comparative results of
increase in the incidence of acute rejection. It should, how- cadaver and related donor renal homografts in man, and immuno-
ever, be noted that the majority of these studies were under- logic implications of the outcome of second and paired transplants.
taken in low-risk Caucasian populations. Ann Surg 1966;164(3):352–97.
4. Mowbray JF, Cohen SL, Doak PB, et al. Human cadaveric renal trans-
plantation: report of twenty cases. BMJ 1965;2(5475):1387–94.
COMPLETE STEROID AVOIDANCE 5. Straffon RA, Hewitt CKW, Stewart BH, Nakamoto S, Kolff WJ. Clini-
cal experience with the use of 79 kidneys from cadavers for trans-
A number of the studies described here include data from plantation. Surg Gynecol Obstet 1966;123(3):483–92.
6. Cupps TR, Fauci AS. Corticosteroid-mediated immunoregulation in
patients in whom steroids are avoided completely from the man. Immunol Rev 1982;65:133–55.
time of transplantation. Meta-analysis has shown no clear 7. Fauci AS. Mechanisms of the immunosuppressive and anti-
difference between subgroups of patients avoiding steroids inflammatory effects of glucocorticosteroids. J Immunopharmacol
compared with those continuing for a period after trans- 1978;1(1):1–25.
plantation, although the number of studies is small, and 8. Rhen T, Cidlowski JA. Antiinflammatory action of glucocorti-
coids—new mechanisms for old drugs. N Engl J Med 2005;353(16):
thus the statistical power is limited. Individual studies have 1711–23.
attempted to address the timing of steroid withdrawal, most 9. Billingham RE, Krohn PL, Medawar PB. Effect of cortisone on sur-
notably the three-arm FREEDOM study.102 In this study, vival of skin homografts in rabbits. BMJ 1951;1(4716):1157–63.
patients received basiliximab induction, mycophenolate 10. Kreis H, Chkoff N, Droz D, et al. Nonsteroid antiinflammatory agents
as a substitute treatment for steroids in ATGAM-treated cadaver kid-
sodium and cyclosporine, and were randomized to receive ney recipients. Transplantation 1984;37(2):139–45.
no steroids, steroids for 7 days, or continue steroids. Inci- 11. Burleson RL, Marbarger PD, Jermanovich N, Brennan AM, Scruggs
dence of biopsy-proven acute rejection was higher in the BF. A prospective study of methylprednisolone and prednisone as
avoidance group compared with the withdrawal group immunosuppressive agents in clinical renal transplantation. Trans-
(31.5% vs. 26.1%), but no differences in graft survival or plant Proc 1981;13(1 Pt 1):339–43.
12. Gambertoglio JG, Frey FJ, Holford NH, et al. Prednisone and pred-
renal function were seen. The composite end-point at 12 nisolone bioavailability in renal transplant patients. Kidney Int
months of biopsy-proven acute rejection, graft loss, or death 1982;21(4):621–6.
was 36.0% in the steroid-free group, 29.6% with steroid 13. Ost L. Impairment of prednisolone metabolism by cyclosporine treat-
withdrawal, and 19.3% with standard steroids, leading the ment in renal graft recipients. Transplantation 1987;44(4):533–5.
14. Tornatore KM, Walshe JJ, Reed KA, Holdsworth MT, Venuto RC.
authors to conclude that early withdrawal may be prefer- Comparative methylprednisolone pharmacokinetics in renal trans-
able to complete avoidance. plant patients receiving double- or triple-drug immunosuppression.
Ann Pharmacother 1993;27(5):545–9.
15. Cattaneo D, Perico N, Gaspari F, Gotti E, Remuzzi G. Glucocorticoids
Conclusions interfere with mycophenolate mofetil bioavailability in kidney trans-
plantation. Kidney Int 2002;62(3):1060–7.
16. Jusko WJ, Ferron GM, Mis SM, Kahan BD, Zimmerman JJ. Pharmaco-
Corticosteroids remain an important part of the transplant kinetics of prednisolone during administration of sirolimus in patients
immunosuppressive armory for induction, maintenance, with renal transplants. J Clin Pharmacol 1996;36(12):1100–6.
240 Kidney Transplantation: Principles and Practice

17. Northrop JP, Crabtree GR, Mattila PS. Negative regulation of inter- 40. d’Apice AJ, Becker GJ, Kincaid-Smith P, et al. A prospective random-
leukin 2 transcription by the glucocorticoid receptor. J Exp Med ized trial of low-dose versus high-dose steroids in cadaveric renal
1992;175(5):1235–45. transplantation. Transplantation 1984;37(4):373–7.
18. Knudsen PJ, Dinarello CA, Strom TB. Glucocorticoids inhibit tran- 41. Grant SD, Forsham PH, DiRaimondo VC. Suppression of
scriptional and post-transcriptional expression of interleukin 1 in 17-­hydroxycorticosteroids in plasma and urine by single and divided
U937 cells. J Immunol 1987;139(12):4129–34. doses of triamcinolone. N Engl J Med 1965;273(21):1115–8.
19. Zanker B, Walz G, Wieder KJ, Strom TB. Evidence that glucocortico- 42. Nichols T, Nugent CA, Tyler FH. Diurnal variation in supression
steroids block expression of the human interleukin-6 gene by acces- of adrenal function by glucocorticoids. J Clin Endocrinol Metab
sory cells. Transplantation 1990;49(1):183–5. 1965;25:343–9.
20. Hearing SD, Norman M, Smyth C, Foy C, Dayan CM. Wide variation 43. Breitenfield RV, Hebert LA, Lemann Jr J, et al. Stability of renal trans-
in lymphocyte steroid sensitivity among healthy human volunteers. plant function with alternate-day corticosteroid therapy. JAMA
J Clin Endocrinol Metab 1999;84(11):4149–54. 1980;244(2):151–6.
21. Kirkham BW, Corkill MM, Davison SC, Panayi GS. Response to 44. Diethelm AG, Sterling WA, Hartley MW, Morgan JM. Alternate-day
glucocorticoid treatment in rheumatoid arthritis: in vitro cell prednisone therapy in recipients of renal allografts: risk and benefits.
mediated immune assay predicts in vivo responses. J Rheumatol Arch Surg 1976;111(8):867–70.
1991;18(6):821–5. 45. Dumler F, Levin NW, Szego G, Vulpetti AT, Preuss LE. Long-term
22. Hearing SD, Norman M, Probert CS, Haslam N, Dayan CM. Predict- alternate day steroid therapy in renal transplantation: a controlled
ing therapeutic outcome in severe ulcerative colitis by measuring study. Transplantation 1982;34(2):78–82.
in vitro steroid sensitivity of proliferating peripheral blood lympho- 46. Leb DE. Alternate day prednisone treatment may increase kidney
cytes. Gut 1999;45(3):382–8. transplant rejection. Proc Clin Dial Transplant Forum 1979;9:
23. Corrigan CJ, Brown PH, Barnes NC, Tsai JJ, Frew AJ, Kay AB. Glu- 136–9.
cocorticoid resistance in chronic asthma. Peripheral blood T lym- 47. McDonald FD, Horensten ML, Mayor GB, Turcotte JG, Selez-
phocyte activation and comparison of the T lymphocyte inhibitory inka W, Schork MA. Effect of alternate-day steroids on renal
effects of glucocorticoids and cyclosporin A. Am Rev Respir Dis transplant function: a controlled study. Nephron 1976;17(6):
1991;144(5):1026–32. 415–29.
24. Seki M, Ushiyama C, Seta N, et al. Apoptosis of lymphocytes induced 48. Potter DE, Holliday MA, Wilson CJ, Salvatierra Jr O, Belzer FO.
by glucocorticoids and relationship to therapeutic efficacy in ­Alternate-day steroids in children after renal transplantation. Trans-
patients with systemic lupus erythematosus. Arthritis & Rheuma- plant Proc 1975;7(1):79–82.
tism. 1998;41(5):823–30. 49. Kauffman HM, Sampson D, Fox PS, Stawicki AT. High dose (bolus)
25. Kang XX, Hirano T, Oka K, Sakurai E, Tamaki T, Kozaki M. Role intravenous methylprednisolone at the time of kidney homotrans-
of altered prednisolone-specific lymphocyte sensitivity in chronic plantation. Ann Surg 1977;186(5):631–4.
renal failure as a pharmacodynamic marker of acute allograft 50. Gray D, Shepherd H, Daar A, Oliver DO, Morris PJ. Oral versus intra-
rejection after kidney transplantation. European J Clin Pharmacol venous high-dose steroid treatment of renal allograft rejection: the
1991;41(5):417–23. big shot or not? Lancet 1978;1(8056):117–8.
26. Langhoff E, Ladefoged J, Jakobsen BK, et al. Recipient lymphocyte 51. Orta-Sibu N, Chantler C, Bewick M, Haycock G. Comparison of
sensitivity to methylprednisolone affects cadaver kidney graft sur- high-dose intravenous methylprednisolone with low-dose oral pred-
vival. Lancet 1986;1(8493):1296–7. nisolone in acute renal allograft rejection in children. BMJ Clinical
27. Langhoff E, Ladefoged J. The impact of high lymphocyte sensitiv- Research Ed 1982;285(6337):258–60.
ity to glucocorticoids on kidney graft survival in patients treated 52. Thompson JF, Chalmers DH, Wood RF, Kirkham SR, Morris PJ. Sud-
with azathioprine and cyclosporine. Transplantation 1987;43(3): den death following high-dose intravenous methylprednisolone.
380–4. Transplantation 1983;36(5):594–6.
28. De Antonio SR, Saber LT, Chriguer RS, de Castro M. Glucocorticoid 53. Kauffman Jr HM, Stromstad SA, Sampson D, Stawicki AT. Random-
resistance in dialysis patients may impair the kidney allograft out- ized steroid therapy of human kidney transplant rejection. Trans-
come. Nephrol Dial Transplant 2008;23(4):1422–8. plant Proc 1979;11(1):36–8.
29. Schaaf MJM, Cidlowski JA. Molecular mechanisms of glucocorticoid 54. Stromstad SA, Kauffman HM, Sampson D, Stawicki AT. Randomized
action and resistance. J Steroid Biochem Mol Biol 2002;83(1-5): steroid therapy of human kidney transplant rejection. Surg Forum
37–48. 1978;29:376–7.
30. McGeown MG, Kennedy JA, Loughridge WG, et al. One hundred kid- 55. Lui SF, Sweny P, Scoble JE, Varghese Z, Moorhead JF, Fernando ON.
ney transplants in the Belfast city hospital. Lancet 1977;2(8039): Low-dose vs high-dose intravenous methylprednisolone therapy
648–51. for acute renal allograft rejection in patients receiving cyclosporin
31. Buckels JA, Mackintosh P, Barnes AD. Controlled trial of low versus therapy. Nephrol Dial Transplant 1989;4(5):387–9.
high dose oral steroid therapy in 100 cadaveric renal transplants. 56. Veenstra DL, Best JH, Hornberger J, Sullivan SD, Hricik DE. Incidence
Proc Eur Dial Transplant Assoc 1981;18:394–9. and long-term cost of steroid-related side effects after renal trans-
32. Chan L, French ME, Beare J, Oliver DO, Morris PJ. Prospective trial of plantation. Am J Kidney Dis 1999;33(5):829–39.
high-dose versus low-dose prednisolone in renal transplant patients. 57. Gheith OA, Nematalla AH, Bakr MA, Refaie A, Shokeir AA, Ghoneim
Transplant Proc 1980;12(2):323–6. MA. Steroid avoidance reduces the cost of morbidities after live-
33. Chan L, French ME, Oliver DO, Morris PJ. High- and low-dose pred- donor renal allotransplants: a prospective, randomized, controlled
nisolone. Transplant Proc 1981;13(1 Pt 1):336–8. study. Exp Clin Transplant 2011;9(2):121–7.
34. De Vecchi A, Rivolta E, Tarantino A, et al. Controlled trial of two dif- 58. Rizzoni G, Broyer M, Guest G, Fine R, Holliday MA. Growth retarda-
ferent methylprednisolone doses in cadaveric renal transplantation. tion in children with chronic renal disease: scope of the problem. Am
Nephron 1985;41(3):262–6. J Kidney Dis 1986;7(4):256–61.
35. Hricik DE, Almawi WY, Strom TB. Trends in the use of glucocor- 59. Isoniemi H. Renal allograft immunosuppression V: glucose intoler-
ticoids in renal transplantation. Transplantation 1994;57(7): ance occurring in different immunosuppressive treatments. Clin
979–89. Transplant 1991;5(3):268–72.
36. Isoniemi H, Ahonen J, Eklund B, et al. Renal allograft immunosup- 60. Jensik SC. Tacrolimus (FK 506) in kidney transplantation: three-
pression. II. A randomized trial of withdrawal of one drug in triple year survival results of the US multicenter, randomized, compara-
drug immunosuppression. Transplant Int 1990;3(3):121–7. tive trial. FK 506 Kidney Transplant Study Group. Transplant Proc
37. Morris PJ, Chan L, French ME, Ting A. Low dose oral prednisolone in 1998;30(4):1216–8.
renal transplantation. Lancet 1982;1(8271):525–7. 61. Almond MK, Kwan JT, Evans K, Cunningham J. Loss of regional
38. Papadakis J, Brown CB, Cameron JS, et al. High versus “low” dose bone mineral density in the first 12 months following renal trans-
corticosteroids in recipients of cadaveric kidneys: prospective con- plantation. Nephron 1994;66(1):52–7.
trolled trial. BMJ Clinical Research Ed 1983;286(6371):1097–100. 62. Grotz WH, Mundinger FA, Gugel B, Exner V, Kirste G, Schollmeyer
39. Stabile C, Vincenti F, Garovoy M, et al. Is a “low” dose of prednisone PJ. Bone fracture and osteodensitometry with dual energy X-ray
better than a “high” dose at the time of renal transplantation? Braz J absorptiometry in kidney transplant recipients. Transplantation
Med Biol Res 1986;19(3):355–66. 1994;58(8):912–5.
16 • Steroids 241

63. Grotz WH, Mundinger FA, Gugel B, Exner VM, Kirste G, Schollmeyer 83. Mahalati K, Kahan BD. A pilot study of steroid withdrawal from kid-
PJ. Bone mineral density after kidney transplantation: a cross- ney transplant recipients on sirolimus-cyclosporine a combination
sectional study in 190 graft recipients up to 20 years after transplan- therapy. Transplant Proc 2001;33(7-8):3232–3.
tation. Transplantation 1995;59(7):982–6. 84. Mahalati K, Kahan BD. Sirolimus permits steroid withdrawal from a
64. Horber FF, Casez JP, Steiger U, Czerniak A, Montandon A, Jaeger P. cyclosporine regimen. Transplant Proc 2001;33(1-2):1270.
Changes in bone mass early after kidney transplantation. J Bone 85. Kumar MS, Heifets M, Moritz MJ, et al. Safety and efficacy of steroid
Miner Res 1994;9(1):1–9. withdrawal two days after kidney transplantation: analysis of results
65. Julian BA, Laskow DA, Dubovsky J, Dubovsky EV, Curtis JJ, Quarles at three years. Transplantation 2006;81(6):832–9.
LD. Rapid loss of vertebral mineral density after renal transplanta- 86. Gelens MA, Christiaans MH, van Heurn EL, van den Berg-Loonen
tion. N Engl J Med 1991;325(8):544–50. EP, Peutz-Kootstra CJ, van Hooff JP. High rejection rate during calci-
66. Wolpaw T, Deal CL, Fleming-Brooks S, Bartucci MR, Schulak JA, neurin inhibitor-free and early steroid withdrawal immunosuppres-
Hricik DE. Factors influencing vertebral bone density after renal sion in renal transplantation. Transplantation 2006;82(9):1221–3.
transplantation. Transplantation 1994;58(11):1186–9. 87. Newell KA, Mehta AK, Larsen CP, et al. Lessons learned: early termi-
67. Hollander AA, Hene RJ, Hermans J, van Es LA, van der Woude FJ. nation of a randomized trial of calcineurin inhibitor and corticosteroid
Late prednisone withdrawal in cyclosporine-treated kidney trans- avoidance using belatacept. Am J Transplant 2017;17(10):2712–9.
plant patients: a randomized study. J Am Soc Nephrol 1997;8(2): Available online at: https://doi.org/10.1111/ajt.14377.
294–301. 88. Pascual J, Quereda C, Zamora J, Hernandez D. Steroid withdrawal in
68. Aroldi A, Tarantino A, Montagnino G, Cesana B, Cocucci C, Pon- renal transplant patients on triple therapy with a calcineurin inhibi-
ticelli C. Effects of three immunosuppressive regimens on vertebral tor and mycophenolate mofetil: a meta-analysis of randomized, con-
bone density in renal transplant recipients: a prospective study. trolled trials. Transplantation 2004;78(10):1548–56.
Transplantation 1997;63(3):380–6. 89. Tan JY, Zhao N, Wu TX, et al. Steroid withdrawal increases risk of
69. Stein EM, Ortiz D, Jin Z, McMahon DJ, Shane E. Prevention of frac- acute rejection but reduces infection: a meta-analysis of 1681 cases
tures after solid organ transplantation: a meta-analysis. J Clin Endo- in renal transplantation. Transplant Proc 2006;38(7):2054–6.
crinol Metab 2011;96(11):3457–65. 90. Knight SR, Morris PJ. Steroid avoidance or withdrawal after renal
70. Shun-Shin GA, Ratcliffe P, Bron AJ, Brown NP, Sparrow JM. The lens transplantation increases the risk of acute rejection but decreases
after renal transplantation. Br J Ophthalmol 1990;74(5):267–71. cardiovascular risk: a meta-analysis. Transplantation 2010;89(1):
71. Slakey DP, Johnson CP, Cziperle DJ, et al. Management of severe 1–14.
pancreatitis in renal transplant recipients. Ann Surg 1997;225(2): 91. Knight SR, Morris PJ. Interaction between maintenance steroid dose
217–22. and the risk/benefit of steroid avoidance and withdrawal regimens
72. Broersen LH, Pereira AM, Jorgensen JO, Dekkers OM. Adrenal insuf- following renal transplantation. Transplantation 2011;92(11):e63–
ficiency in corticosteroids use: systematic review and meta-analysis. 4.
J Clin Endocrinol Metab 2015;100(6):2171–80. 92. Hruba P, Tycova I, Krepsova E, et al. Steroid free immunosuppres-
73. Ojo AO, Hanson JA, Wolfe RA, Leichtman AB, Agodoa LY, Port FK. sion is associated with enhanced Th1 transcripts in kidney trans-
Long-term survival in renal transplant recipients with graft func- plantation. Transpl Immunol 2017;42:18–23.
tion. Kidney Int 2000;57(1):307–13. 93. Thomusch O, Wiesener M, Opgenoorth M, et al. Rabbit-ATG or
74. Anderton JL, Fananapazir L, Eccleston M. Minimum steriod require- basiliximab induction for rapid steroid withdrawal after renal trans-
ments in renal transplant patients monitored by urinary fibrin deg- plantation (Harmony): an open-label, multicentre, randomised con-
radation products and complement. Proc Eur Dial Transplant Assoc trolled trial. Lancet 2016;388(10063):3006–16.
1977;14:342–50. 94. Opelz G. Influence of treatment with cyclosporine, azathioprine and
75. Naik RB, Chakraborty J, English J, Marks V, Slapak M, Lee HA. Seri- steroids on chronic allograft failure. The Collaborative Transplant
ous renal transplant rejection and adrenal hypofunction after grad- Study. Kidney Int Suppl 1995;52:S89–92.
ual withdrawal of prednisolone two years after transplantation. BMJ 95. Opelz G, Dohler B, Laux G. Long-term prospective study of steroid
1980;280(6228):1337–40. withdrawal in kidney and heart transplant recipients. Am J Trans-
76. Hricik DE, O’Toole MA, Schulak JA, Herson J. Steroid-free immuno- plant 2005;5(4 Pt 1):720–8.
suppression in cyclosporine-treated renal transplant recipients: a 96. Opelz G, Dohler B. Association between steroid dosage and death
meta-analysis. J Am Soc Nephrol 1993;4(6):1300–5. with a functioning graft after kidney transplantation. Am J Trans-
77. Sinclair NR. Low-dose steroid therapy in cyclosporine-treated plant 2013;13(8):2096–105.
renal transplant recipients with well-functioning grafts. The Cana- 97. Sureshkumar KK, Marcus RJ, Chopra B. Role of steroid mainte-
dian Multicentre Transplant Study Group. CMAJ 1992;147(5): nance in sensitized kidney transplant recipients. World J Transplant
645–57. 2015;5(3):102–9.
78. Ratcliffe PJ, Dudley CR, Higgins RM, Firth JD, Smith B, Morris PJ. 98. Benfield MR, Bartosh S, Ikle D, et al. A randomized double-blind, pla-
Randomised controlled trial of steroid withdrawal in renal trans- cebo controlled trial of steroid withdrawal after pediatric renal trans-
plant recipients receiving triple immunosuppression. Lancet plantation. Am J Transplant 2010;10(1):81–8.
1996;348(9028):643–8. 99. Hocker B, Weber LT, Feneberg R, et al. Improved growth and car-
79. Woodle ES, First MR, Pirsch J, Shihab F, Gaber AO, Van Veldhuisen diovascular risk after late steroid withdrawal: 2-year results of a
P. A prospective, randomized, double-blind, placebo-controlled prospective, randomised trial in paediatric renal transplantation.
multicenter trial comparing early (7 day) corticosteroid cessa- Nephrol Dial Transplant 2009;25(2):617–24.
tion versus long-term, low-dose corticosteroid therapy. Ann Surg 100. Grenda R, Watson A, Trompeter R, et al. A randomized trial to assess
2008;248(4):564–77. the impact of early steroid withdrawal on growth in pediatric renal
80. Pescovitz MD, Kahan BD, Julian BA, Neylan J, Chan G. Sirolimus transplantation: the twist study. Am J Transplant 2010;10(4):
(SRL) permits early steroid withdrawal from a triple therapy renal 828–36.
prophylaxis regimen. ASTP Abstracts 1997;62:261. 101. Zhang H, Zheng Y, Liu L, et al. Steroid avoidance or withdrawal regi-
81. Kahan BD, Podbielski J, Schoenberg L. Use of sirolimus to facilitate mens in paediatric kidney transplantation: a meta-analysis of ran-
steroid withdrawal from a cyclosporine regimen. Transplant Proc domised controlled trials. PLoS ONE 2016;11(3):e0146523.
2006;38(9):2842–6. 102. Vincenti F, Schena FP, Paraskevas S, Hauser IA, Walker RG, Grinyo
82. Citterio F, Sparacino V, Altieri P, et al. Addition of sirolimus to cyclo- J. A randomized, multicenter study of steroid avoidance, early steroid
sporine in long-term kidney transplant recipients to withdraw ste- withdrawal or standard steroid therapy in kidney transplant recipi-
roid. Transplant Proc 2005;37(2):827–9. ents. Am J Transplant 2008;8(2):307–16.
17 Calcineurin Inhibitors
MATTHEW L. HOLZNER, VIKRAM WADHERA, AMIT BASU, SANDER
FLORMAN, and RON SHAPIRO

CHAPTER OUTLINE Cyclosporine Early Corticosteroid Withdrawal Regimens


Tacrolimus Corticosteroid-Free Immunosuppression
Mechanism of Action Regimens
Pharmacokinetic Properties Comparison of Corticosteroid-Sparing
Pharmacogenetics Regimens Using Tacrolimus-Based and
Cyclosporine-Based Immunosuppression
Absorption and Distribution
Calcineurin Inhibitor Avoidance and Low-
Metabolism and Elimination Dose Tacrolimus Regimens
Special Patient Populations Other Calcineurin Inhibitors and
Clinical Studies in Kidney Transplantation Formulations
Rescue Therapy in Adults Pediatric Renal Transplantation
Antibody-Mediated Rejection Clinical Studies in Kidney–Pancreas
Maintenance Immunosuppression Transplantation
Comparison of Tacrolimus-Based and Simultaneous Pancreas–Kidney
Cyclosporine-Based Regimens Transplantation
Comparison of Calcineurin Inhibitor/ Steroid Withdrawal and Steroid-Free
Azathioprine and Tacrolimus/ Protocols
Mycophenolate Mofetil Regimens Pancreas after Kidney Transplantation
Comparison of Tacrolimus/Mycophenolate Side Effects and Tolerability of Calcineurin
Mofetil and Calcineurin Inhibitor/ Inhibitors
Sirolimus Regimens Cardiovascular Adverse Effects
Comparison of Tacrolimus-Based Dual Posttransplant Diabetes Mellitus
Versus Triple Immunosuppression
Therapy Malignancies
Induction Therapies With Calcineurin Other Side Effects
Inhibitor Regimens Special Patient Populations
Role of Calcineurin Inhibitors and Conclusion
Corticosteroids in the Development of
Hypertension and Hyperglycemia

Although the improved outcomes over the past 60+ years never made it into clinical trials, as the company saw little
in kidney transplantation have been related to a number of point in developing it for transplantation, citing a small
factors, the effect of immunosuppression has perhaps been market and limited financial return. Calne, then professor
the most significant. The advent of the calcineurin inhibi- of surgery at the University of Cambridge, succeeded in con-
tors (CNIs), first cyclosporine and then tacrolimus, repre- vincing Sandoz to develop the agent for transplantation.5
sented a huge advance for the field of transplantation in It was first used clinically in England in the late 1970s by
general and for kidney transplantation in particular. It is Calne and his associates in Cambridge. Initially, it was used
fair to describe transplant outcomes in terms of being in the with other drugs, such as prednisolone or Asta 036.5122
pre-CNI and post-CNI eras. This chapter will discuss the two (cytimum, an analog of cyclophosphamide).6,7 Cyclospo-
CNIs in clinical use, cyclosporine and tacrolimus. rine revolutionized the field of transplantation, improving
outcomes in renal transplantation, making it possible for
routine liver and heart transplantation to be performed,
Cyclosporine and allowing the first clinical trials of pancreas and lung
transplants.
Cyclosporine was first isolated from two strains of imper-
fect fungi (Cylindrocarpon lucidum Booth and Trichoderma
polysporum Rifai) from soil samples by the Department of Tacrolimus
Microbiology at Sandoz (Basel, Switzerland) as an antifun-
gal agent of limited activity.1 Borel and colleagues demon- Tacrolimus (FK506, Prograf) was isolated in 1984 from
strated its potent immunosuppressive activity in a variety of the fermentation broth of Streptomyces tsukubaensis, a soil
in vitro and in vivo experiments.2–4 Interestingly, it nearly organism found at the foot of Mount Tsukuba near Tokyo.
242
17 • Calcineurin Inhibitors 243

This compound was developed by researchers at the Chiba discharge.17 This transition was based on the outcomes of
University of Japan. In the first clinical (rescue) trial, tacroli- a number of prospective, randomized trials demonstrating
mus was administered to patients who were taking standard the superior efficacy of various tacrolimus-based regimens
immunosuppressive therapy but who faced retransplanta- compared with cyclosporine-based regimens.
tion because of ongoing organ rejection, or who had unde-
sirable drug toxicities.8 The initial clinical trial of tacrolimus
as a primary immunosuppressive agent for the prophylaxis Mechanism of Action
of rejection in liver transplant recipients began in the spring
of 1990 at the University of Pittsburgh.9 This work eventu- Calcineurin inhibitors exert their immunosuppressive
ally led to multicenter randomized trials in liver and kidney effects by reducing interleukin-2 (IL-2) production and
transplantation.10,11 Patients treated with tacrolimus had IL-2 receptor expression, leading to a reduction in T cell
significantly fewer and less severe episodes of acute rejec- activation. Tacrolimus inhibits T lymphocyte activation by
tion than did patients given cyclosporine therapy. The binding to FKBP-12, an intracellular protein. A complex is
phase III trials leading to US Food and Drug Administration then formed of tacrolimus–FKBP-12, calcium, calmodulin,
(FDA) approval of tacrolimus (in 1994) were conducted and calcineurin, which inhibits the phosphatase activity of
first in liver rather than in kidney transplant recipients, in calcineurin. This complex prevents the dephosphorylation
contrast to other immunosuppressive agents. Subsequent and subsequent translocation of the nuclear factor of acti-
clinical trials in kidney transplantation led to FDA approval vated T cells (NF-AT), a nuclear component that initiates
for kidney transplantation in 1997. Tacrolimus has also gene transcription for the formation of IL-2 (Fig. 17.1). As
shown efficacy as a rescue agent and as a primary mainte- a result, T lymphocyte activation is inhibited.8 The mech-
nance immunosuppressive agent in heart, lung, pancreas, anism of action of cyclosporine is similar, except that the
and small-bowel transplantation,12–16 and was approved binding protein is cyclophilin. Tacrolimus is 10 to 100
for heart transplantation in 2006. times more potent than cyclosporine in its immunosuppres-
Cyclosporine and tacrolimus are the current mainstream sive effects.18
maintenance immunosuppressive medications used, with a
shift toward progressively higher utilization of tacrolimus-
based regimens over the past 15 years. In 2013 more than Pharmacokinetic Properties
90% of all new adult and 96% of all new pediatric kidney
transplant recipients in the US were receiving tacroli- The pharmacokinetic characteristics of CNIs show high
mus as maintenance immunosuppressive therapy before interindividual and intraindividual variability (i.e., different

Antigen

Helper T-cell

Tacrolimus
Ca2+ Calcineurin
FKBP-12

NF-ATc NF-ATc/n

P P P
IL-2 transcription

Immunosuppression

Fig. 17.1 Mechanism of action of tacrolimus. A complex is formed of tacrolimus–FKBP-12, calcium, calmodulin, and calcineurin, which inhibits the
phosphatase activity of calcineurin. This prevents the dephosphorylation and subsequent translocation of nuclear factor of activated T cells (NF-AT),
a nuclear component that initiates gene transcription for the formation of interleukin-2 (IL-2). C, cytoplasm; n, nucleus, P, phosphate. (From Fung JJ.
Tacrolimus and transplantation: a decade in review. Transplantation 2004;77:S41.)
244 Kidney Transplantation: Principles and Practice

patients have different pharmacokinetic characteristics, Metabolism and Elimination


and the same patient may have different pharmacokinetic
characteristics at different time points after transplanta- Cyclosporine is metabolized almost entirely in the liver,
tion), and the drugs have a narrow therapeutic index; mostly through the CYP-450 system. Most of the drug is
therefore therapeutic drug monitoring is necessary to opti- excreted in the bile, with only trace amounts being excreted
mize treatment. Because 90% of the agents is partitioned in in the urine. Tacrolimus is metabolized extensively in the
the cellular components of blood, whole-blood concentra- liver as well and, to a much lesser extent, in the intestinal
tions correlate better with drug exposure (area under the mucosa, with metabolism mediated at both sites by CYP3A4
curve [AUC]) than do plasma concentrations.19 isoenzymes.18,22 Tacrolimus is converted by hydroxylation
and demethylation to at least 15 metabolites, with the main
metabolite being 13-O-dimethyl-tacrolimus. The mean
Pharmacogenetics clearance after intravenous administration of tacrolimus
is as follows: 0.040 L/h/kg in healthy volunteers, 0.083
The absorption, bioavailability, and elimination of these L/h/kg in adult kidney transplant patients, 0.053 L/h/kg in
drugs are primarily controlled by efflux pumps and enzymes adult liver transplant patients, and 0.051 L/h/kg in adult
of the cytochrome P (CYP) 450 family. DNA variants of heart transplant patients. When administered orally, fecal
the genes encoding these proteins contribute to the inter- elimination accounts for 92.6 ± 3.07% and urinary elimi-
individual heterogeneity of the metabolism of CNIs. Cyclo- nation accounts for 2.3 ± 1.1% of the administered dose in
sporine and tacrolimus are metabolized by CYP3A4 and healthy volunteers.23
CYP3A5, and several single-nucleotide polymorphisms The main drugs that interact with CNIs when admin-
in the two genes have been associated with differences in istered simultaneously are either inducers or inhibitors of
drug clearance. Homozygotes for a common DNA variant CYP3A4. CYP3A4 inhibitors potentially increase whole-
that affects gene splicing (CYP3A5*3) may require a lower blood concentrations, whereas CYP3A4 inducers decrease
dose to remain within the target blood concentration. The CNI concentrations (Table 17.1).
Tactique study randomly assigned 236 patients to either
receive tacrolimus at a fixed dosage or dosage according
to their CYP3A5 genotype. Dosing tacrolimus according Special Patient Populations
to CYP3A5 genotype had no effect on long-term clinical
outcomes, including rates of acute rejection and graft sur- Three percent of patients require higher dosages (>0.4
vival.20 A prospective randomized trial evaluated tacroli- mg/kg/d) to reach therapeutic tacrolimus concentrations.
mus dosing in patients heterozygous for CYP3A4*22, an This is a reflection of the low bioavailability and, to a lesser
allelic variant that is associated with decreased cytochrome extent, the high clearance of the drug.22 In a nonblinded
activity. Carriers of the CYP3A4*22 allele had significantly parallel-group study, the bioavailability of tacrolimus was
altered tacrolimus metabolism and reached higher plasma significantly (P = 0.01) lower in African American (11.9%)
concentrations compared with the control group.21 and Latin American (14.4%) patients than in white patients
(18.8%).24 A retrospective study in renal transplant recipi-
ents showed that African American recipients required
Absorption and Distribution higher dosages of tacrolimus on a milligram-per-kilogram
basis.22
The gastrointestinal absorption of CNIs is highly dependent Children typically require higher tacrolimus dosages on
on the presence of food, bile acids, and motility. They are a milligram-per-kilogram basis than adult patients, most
rapidly but incompletely absorbed in the gastrointestinal likely reflecting the higher mean total body clearance and
tract, and peak concentrations in whole blood are attained
1 to 2 hours after oral administration.22 Tacrolimus has low
oral bioavailability (average 25%; range 4%–93%).22 The
mean oral bioavailability of tacrolimus is comparable in TABLE 17.1 Drug Interactions Associated With
Calcineurin Inhibitors
adult (25%) and pediatric (31%) transplant recipients. The
rate and extent of absorption of tacrolimus are reduced in DRUGS INCREASING TACROLIMUS CONCENTRATION
the presence of food, with the peak concentration in whole (CYTOCHROME P-450 3A4 INHIBITORS)
blood compared with the fasting state decreased by approxi- Calcium channel blockers: diltiazem, nicardipine, nifedipine, vera-
mately 50% to 75%, and the AUC decreased by 25% to 40% pamil
Imidazole antifungal agents: clotrimazole, fluconazole, itraconazole,
when the drug is taken after a meal.18 Tacrolimus is highly ketoconazole
bound to erythrocytes, in a concentration-dependent man- Macrolide antibiotics: clarithromycin, erythromycin
ner, with reduced ratios at higher drug concentrations Prokinetic agents: cisapride, metoclopramide
related to binding saturation. Plasma protein binding may Other drugs: bromocriptine, cimetidine, corticosteroids, danazol,
be 99%, with most of the drug bound to α1-acid glycopro- protease inhibitors
Grapefruit juice
tein and albumin. Tacrolimus is widely distributed in most
tissues, including lungs, spleen, heart, kidney, pancreas, DRUGS DECREASING TACROLIMUS CONCENTRATION
(CYTOCHROME P-450 3A4 INDUCERS)
brain, muscle, and liver; tacrolimus crosses the placenta,
with umbilical cord plasma concentrations one-third of Anticonvulsants: carbamazepine, phenobarbital, phenytoin
Rifabutin/rifampicin, isoniazid
those in maternal plasma.18,22 Tacrolimus also is present in St. John’s wort
breast milk, but at extremely low levels (<2.5 ng/mL).
17 • Calcineurin Inhibitors 245

volume of distribution in children. Clinically relevant differ- cyclosporine-induced hypertrichosis (n = 116) showed
ences do not exist between adults and children, however, marked improvement. The mean serum low-density lipo-
in terms of the time taken to reach maximal blood concen- protein (LDL) level decreased from 138 to 120 mg/dL, and
trations (2.1 hours in children vs. 2 hours in adults), bio- the high-density lipoprotein levels remained unchanged
availability (31% vs. 25%), and mean terminal elimination in patients with cyclosporine-induced hyperlipidemia (n =
half-life (11.5 hours vs. 12 hours).25 The mean clearance of 78). Finally, hypertension markedly or completely resolved
tacrolimus in patients with renal dysfunction was similar in 25% of patients (n = 75).31 A randomized study in which
to that in normal volunteers; tacrolimus pharmacokinet- patients were either converted to tacrolimus (n = 27) or
ics after a single intravenous administration was similar remained on cyclosporine-based immunosuppression (n
in seven patients not receiving dialysis and five receiving = 30) demonstrated a significant reduction in cholesterol
dialysis.23 from 255 to 206 mg/dL in the patients randomized to
The mean clearance of tacrolimus in patients with mild tacrolimus.32
hepatic dysfunction (mean Pugh score of 6.2) was not sub-
stantially different from that in normal volunteers after a
ANTIBODY-MEDIATED REJECTION
single intravenous and oral dose. The mean clearance was
substantially lower in patients with severe hepatic dys- Antibody-mediated rejection often occurs within the first
function (mean Pugh score >10), regardless of the route of 2 weeks after transplantation and is associated with oli-
administration.23 guria, graft tenderness, fever, leukocytosis, and circulat-
ing donor-specific antibodies. Before the introduction of
tacrolimus, combinations of bolus corticosteroids, plasma-
Clinical Studies in Kidney pheresis, and antilymphocyte antibody preparations were
used to treat acute humoral rejection, with inconsistent
Transplantation and unsatisfactory response rates. Tacrolimus-based regi-
mens were developed for acute humoral rejection in renal
RESCUE THERAPY IN ADULTS
transplant recipients, based on clinical experiences with
The efficacy of tacrolimus in kidney transplantation was tacrolimus in treating liver and heart transplants with
first shown in recipients with refractory rejection. Refrac- acute humoral rejection.33–35 Experimental evidence also
tory rejection episodes in cyclosporine-treated patients supported the potential of tacrolimus in limiting antibody
were reversed by replacing cyclosporine with tacrolimus responses.36,37
as the maintenance immunosuppressive agent. In contrast The studies of tacrolimus in acute humoral rejection pre-
to antilymphocyte antibody preparations (e.g., OKT3 and ceded the development of plasmapheresis and intravenous
polyclonal antibody preparations) that induce long-term immunoglobulin regimens in the management of humoral
suppression of T cell responses, the immunosuppressive rejection and highly sensitized patients (see Chapter 20),38–40
effects of tacrolimus could be titrated on a daily basis by fol- and more recent work with bortezomib and eculizumab. Thus
lowing drug levels.26 conversion to tacrolimus for antibody-mediated rejection is
An early large experience with tacrolimus in treating not currently a primary treatment modality, although it may
refractory acute renal allograft rejection in 77 patients accompany antibody-specific therapy.
receiving cyclosporine-based immunosuppressive therapy
was reported from Pittsburgh.27 Several conclusions were MAINTENANCE IMMUNOSUPPRESSION
drawn from this study, as follows: (1) tacrolimus provided
effective therapy for acute renal allograft rejection; (2) The outcomes of kidney transplantation have improved
tacrolimus often provided effective therapy for vascular with the advent of CNIs as part of highly effective immuno-
rejection in kidney transplants; and (3) the success of tacro- suppressive regimens. Several studies have addressed short-
limus therapy for refractory acute renal allograft rejection term outcomes of immunosuppression, including rates of
was related to the severity and duration of rejection. acute rejection and patient and graft survival. Studies also
The 5-year follow-up of the Pittsburgh experience have addressed medium- and long-term outcomes with
showed good long-term renal allograft function in patients CNI-based immunosuppression, including 10-year patient
undergoing tacrolimus rescue therapy.28,29 A total of 169 and graft survival, renal function, cardiovascular events,
patients were converted from cyclosporine to tacrolimus for and posttransplant diabetes mellitus (PTDM).
refractory rejection, with a 74% success rate and a mean
serum creatinine value of 2.3 ± 1.1 mg/dL (202 μmol/L). COMPARISON OF TACROLIMUS-BASED AND
A prospective randomized multicenter comparative trial CYCLOSPORINE-BASED REGIMENS
confirmed the efficacy of tacrolimus-based rescue therapy
in patients with acute renal transplant rejection.30 Rescue The phase III US multicenter clinical trial compared the effi-
therapy with tacrolimus-based regimens reduced the inci- cacy and safety of tacrolimus with that of the original formula-
dence of recurrent acute rejection to 8.8% versus 34.1% (P tion of cyclosporine.11 At 1 year posttransplantation, 30.7%
= 0.002) in patients who remained on cyclosporine-based of tacrolimus-treated patients had experienced acute rejec-
immunosuppression. tion compared with 46.4% of cyclosporine-treated patients
In a large European study on tacrolimus conversion (P = 0.001). The incidence of moderate to severe rejection
for cyclosporine-induced toxicities, 73% of patients with was 10.8% in the tacrolimus-treated group compared with
cyclosporine-induced gingival hyperplasia (n = 32) showed 26.5% in the cyclosporine-treated group. The intent-to-treat
significant resolution of hyperplasia, and recipients with analysis showed no significant differences in 5-year patient
246 Kidney Transplantation: Principles and Practice

or graft survival between the tacrolimus-treated and the arm and 11.8% in the tacrolimus/azathioprine arm; P =
cyclosporine-treated patients. When crossover because of 0.05). There were no significant differences among the three
rejection was counted as graft failure, a statistically signifi- groups in patient or graft survival at 1, 2, and 3 years. A
cant increase in graft survival was found in the tacrolimus 2012 single-institution, prospective, open-labeled, random-
group at 5 years (63.8% vs. 53.8%; P = 0.014). The patients ized controlled trial comparing cyclosporine/azathioprine
treated with tacrolimus had a lower incidence of hirsutism (n = 146) versus tacrolimus/MMF (n = 143) in 289 kidney
and gingival hyperplasia, but a higher incidence of alopecia, transplant recipients treated with antithymocyte globulin
than patients treated with cyclosporine. (ATG) and prednisone showed equal patient and graft sur-
Racial differences were also evaluated for acute rejection vivals at 1 year; however, the tacrolimus/MMF group had
in the US phase III multicenter clinical trial.41 Among Afri- better estimated glomerular filtration rates (GFR; cyclospo-
can Americans, 23.2% of patients in the tacrolimus-treated rine/azathioprine 48 ± 1 vs. tacrolimus/MMF 53 ± 1 mL/
group developed acute rejection compared with 47.9% of min/1.73 m2, P = 0.007), and a trend toward lower rates
patients in the cyclosporine-treated group (P = 0.012). of biopsy-proven acute rejection (BPAR), cyclosporine/aza-
When crossover because of rejection was counted as graft thioprine (14.4%) versus 11 (7.7%) with tacrolimus/MMF
failure, there was a significant increase in the 5-year graft (P = 0.07).47
survival in African American patients in the tacrolimus-
treated group (65.4% vs. 42.6%; P = 0.013) compared with COMPARISON OF TACROLIMUS/
the cyclosporine-treated group.42 MYCOPHENOLATE MOFETIL AND CALCINEURIN
The US multicenter study that compared the efficacy and
INHIBITOR/SIROLIMUS REGIMENS
tolerability of tacrolimus versus cyclosporine also revealed
that significantly fewer kidney transplant recipients required A randomized study comparing the combination of sirolimus
antihypertensive treatment in the tacrolimus-treated group or MMF with tacrolimus-based immunosuppression showed
compared with the cyclosporine-treated group.43 The pro- no significant differences in the incidence of biopsy-confirmed
jected graft half-life evaluated by the European Multicenter acute rejection (BCAR; 13% tacrolimus/sirolimus (n = 185)
Renal Transplant Study also favored tacrolimus over cyclo- versus 11.4% tacrolimus/MMF (n = 176; P = 0.64).48 Graft
sporine (15.8 vs. 10.8 years).44 survival and patient survival were not significantly different
A multicenter trial evaluated the effect of tacrolimus between the groups at 6 months after transplantation. The
as secondary intervention in patients being treated with combination of tacrolimus and MMF was superior to tacroli-
cyclosporine for 3 or more months after transplantation mus and sirolimus in terms of improved renal function and a
who had one of the following risk factors for chronic renal lower risk of hypertension and hyperlipidemia.48 The incidence
allograft failure: serum creatinine 2 mg/dL or greater for of acute rejection was significantly higher in the tacrolimus/
men and 1.7 mg/dL or greater for women, or a greater than sirolimus arm (30% vs. 2% for cyclosporine/sirolimus and
30% increase in the nadir posttransplant serum creatinine 12% for tacrolimus/MMF) at 36 months in a randomized trial
level. The trial randomly assigned 197 patients to convert comparing these three regimens in renal transplantation.49
to tacrolimus or remain on cyclosporine.45 At 24 months, There was not, however, a significant difference in actuarial
56.8% of the patients in the tacrolimus-treated group and graft survival at 8 years posttransplant among the groups.
87.5% in the cyclosporine-treated group had a serum cre- These optimistic findings were countered by an analysis of
atinine level 2 mg/dL or greater (P = 0.002). Significantly 44,915 adult renal transplants in the Scientific Renal Trans-
fewer patients who were converted from cyclosporine to plant Registry from 2000 to 2004. A total of 3524 (7.8%)
tacrolimus experienced a cardiovascular event compared patients received a baseline immunosuppressive regimen of
with patients who continued treatment with cyclosporine tacrolimus/sirolimus, with an inferior overall survival (P <
(5.6% vs. 24.3%; P = 0.002). Median serum cholesterol 0.001) and death-censored graft survival (P < 0.001) com-
and LDL cholesterol levels were significantly lower in the pared with tacrolimus/MMF (n = 27,007). Six-month acute
tacrolimus-treated group compared with the cyclosporine- rejection rates were low and did not differ among groups.50
treated group. Therapeutic intervention with tacrolimus These data have to be interpreted in the context of the limi-
resulted in improved renal function, better lipid profiles, tations of any retrospective database analysis.
and fewer cardiovascular events in patients who were at A steroid-free randomized trial comparing sirolimus/
risk for developing chronic renal allograft failure.45 tacrolimus, sirolimus/mycophenolate, and tacrolimus/
mycophenolate showed higher than expected BCAR in the
COMPARISON OF CALCINEURIN INHIBITOR/ sirolimus/mycophenolate arm. The BCAR and graft survival
AZATHIOPRINE AND TACROLIMUS/ at 2 years for the sirolimus/tacrolimus group was 17.4% and
MYCOPHENOLATE MOFETIL REGIMENS 88.5% respectively, and for the tacrolimus/mycophenolate
group 12.3% and 95.4%. Among the side effects, the siroli-
A randomized prospective three-arm study compared the mus group had a higher incidence of delayed wound healing
effect of immunosuppressive protocols using tacrolimus/ and hyperlipidemia.51
azathioprine (n = 76), tacrolimus/mycophenolate mofetil
(MMF) (n = 72), and cyclosporine microemulsion/MMF (n COMPARISON OF TACROLIMUS-BASED DUAL
= 75).46 At 1 year, although there were no significant dif- VERSUS TRIPLE IMMUNOSUPPRESSION
ferences in overall rejection rates, there were significant
THERAPY
differences in the total number of patients who required anti-
lymphocyte antibody treatment (4.2% in the tacrolimus/ Dual immunosuppression therapy refers to the use of
MMF arm compared with 10.7% in the cyclosporine/MMF tacrolimus with a second agent, such as a corticosteroid.
17 • Calcineurin Inhibitors 247

Triple immunosuppression therapy refers to the use of risk patients, as defined by a prior failed renal transplant,
tacrolimus and a corticosteroid with a third agent, such as prior history of sensitization, or panel-reactive antibodies
azathioprine or MMF. >20%. Patients were randomized to receive single-dose
Dual therapy with tacrolimus-based immunosuppression alemtuzumab before graft reperfusion, with tacrolimus
provided similar efficacy to tacrolimus-based triple therapy monotherapy, or four doses of thymoglobulin with tacro-
for 36 months.52–57 At 12 months, patient survival rates in limus, mycophenolate, and steroids. One-year cumulative
the dual-therapy groups were ≥96% compared with ≥94% graft survival was 85.7% for the alemtuzumab group and
with triple therapy, with graft survival rates of ≥90% (dual- 87.5% for the thymoglobulin group.65 A similar study was
therapy groups) and ≥91% (triple-therapy groups). Three- done comparing the efficacy of alemtuzumab versus con-
year follow-up data are available from the Italian and ventional induction therapy (basiliximab or rabbit [rATG]).
Spanish trials, and graft survival was 87% in dual-therapy In this trial patients were stratified according to acute rejec-
and triple-therapy groups. A similar percentage of patients tion risk, with a high risk defined by a repeat transplant, a
experienced an acute rejection episode with dual-therapy peak or current value of panel-reactive antibodies of 20%
or triple-therapy tacrolimus-based immunosuppressive or more, or African American ethnicity. There were 139
regimens. Most of these episodes occurred in the first year high-risk patients, of whom 70 received alemtuzumab and
after transplantation, with a 10- to 15-fold reduction in 69 received rATG. The 335 low-risk patients were random-
the incidence of rejection over the next 2 years.58,55 In one ized to alemtuzumab (164 patients) or basiliximab (171
study, the addition of MMF to tacrolimus plus corticosteroid patients). All patients received tacrolimus and MMF and
therapy significantly (P = 0.007) reduced the incidence of underwent a 5-day glucocorticoid taper in a regimen of
rejection at 9 months.59 early steroid withdrawal. By the first year after transplanta-
In another prospective randomized trial the combination tion, BCAR was less frequent with alemtuzumab than with
of tacrolimus and prednisone was compared with tacroli- conventional therapy. The apparent superiority of alemtu-
mus, MMF, and prednisone in renal transplant recipients zumab with respect to early BCAR was restricted to patients
without induction therapy.60 The combination of tacro- at low risk for transplant rejection; among high-risk
limus, steroids, and MMF was associated with excellent patients, alemtuzumab and rATG had similar efficacy.66
patient and graft survival and a lower incidence of rejection In another study, alemtuzumab (n = 113) or rATG (n =
than the combination of tacrolimus and steroids. Currently, 109) induction was compared in 180 (81%) kidney trans-
most centers utilize a triple immunosuppression regimen, plants alone, 38 (17%) simultaneous pancreas–kidney
consistent with the 2009 Kidney Disease: Improving Global (SPK), and 4 (2%) pancreas after kidney (PAK) transplants.
Outcomes guidelines.61 Survival, initial length of stay, and maintenance immu-
nosuppression (including early steroid elimination) were
INDUCTION THERAPIES WITH CALCINEURIN similar between alemtuzumab and rATG groups, but BPAR
episodes occurred in 16 (14%) alemtuzumab patients com-
INHIBITOR REGIMENS
pared with 28 (26%) rATG patients (P = 0.02). Late BPAR
Margreiter et al.62 studied the efficacy of alemtuzumab (>12 months after transplant) occurred in one (8%) alem-
induction followed by tacrolimus monotherapy (n = 65) tuzumab patient and three (11%) rATG patients (P = not
compared with a control group of tacrolimus, MMF, and significant [NS]). Infections and malignancy were similar
steroids (n = 66). At 12 months the biopsy-proven rejection between the two induction arms. The results of this study
rate was 20% in the study group and 32% in the control showed that alemtuzumab and rATG induction therapies
group (P = 0.09). Patient survival at 1 year was 98% for were equally safe, but alemtuzumab was associated with
both groups. Graft survival was 96% for the study group less BPAR.67
versus 90% for the control group (P = 0.18). A similar
study compared alemtuzumab induction with tacrolimus ROLE OF CALCINEURIN INHIBITORS AND
monotherapy against daclizumab, tacrolimus, and myco- CORTICOSTEROIDS IN THE DEVELOPMENT OF
phenolate therapy. The alemtuzumab and tacrolimus arm HYPERTENSION AND HYPERGLYCEMIA
showed a survival with a functioning graft at 1 year of
97.6% versus 95.1% in the daclizumab arm and at 2 years Although both CNIs and steroids have been associated with
of 92.6% versus 95.1% in the daclizumab group.63 These posttransplant hypertension and hyperglycemia, steroid
results suggest that alemtuzumab induction together with dosing may have a major part in the development of these
tacrolimus monotherapy is at least as efficient as the dacli- complications after transplantation (see Chapter 16). In
zumab, tacrolimus, and mycophenolate or a tacrolimus, one study, patients were evaluated 4 months after kidney
mycophenolate, and steroids regimen. The use of alemtu- transplantation; twice as many patients treated with tacro-
zumab induction with tacrolimus monotherapy was evalu- limus and high-dose prednisone developed hypertension
ated in 200 living donor kidney transplant recipients. The compared with patients treated with tacrolimus and low-
actuarial 3-year patient and graft survival in this study was dose prednisone (63% vs. 32%, P < 0.05).68
96.4% and 86.3%, respectively. The cumulative incidence Corticosteroids may promote the development of PTDM
of acute cellular rejection (ACR) at 3 years was 24%, about by inducing insulin resistance, decreasing insulin receptor
88.7% of the ACR episodes were Banff 1, and of those, 82% number and affinity, impairing endogenous glucose pro-
were steroid-sensitive. The mean serum creatinine (mg/ duction, and impairing glucose uptake by muscle.69
dL) and GFR (mL/min/1.73 m2) at 3 years were 1.5 ± 0.7 A study was done to assess the relative role of tacrolimus
and 54.9 ± 20.9, respectively.64 The alemtuzumab/tacro- and corticosteroids in the development of glucose metabolic
limus treatment was also evaluated in high immunologic disorders.70 Corticosteroid withdrawal in patients receiving
248 Kidney Transplantation: Principles and Practice

tacrolimus-based immunosuppression led to a 22% decrease roids.74 Anti-CD25 monoclonal antibody was administered
in fasting C-peptide levels (P = 0.0009). Fasting insulin to 25 patients at higher immunologic risk. After a median
levels and the insulin-to-glucose ratio also decreased but follow-up of 51 months (range 36–62 months), patient
not significantly (P = NS). Steroid withdrawal also led to survival was 97%, and graft survival was 91%. The inci-
a reduction in lipid levels. Tacrolimus trough level reduc- dence of acute rejection at 12 months was 19%. Only three
tion from 9.5 to 6.4 ng/mL resulted in a 36% increase in further episodes of rejection occurred beyond 12 months.
pancreatic beta-cell secretion (P = 0.04), and insulin secre- Graft function was stable during the study, with a mean
tion increased by a similar rate. Hemoglobin A1c improved estimated creatinine clearance of 57 mL/min at the end of
from 5.9% to 5.3% (P = 0.002), although lipid levels did follow-up. This steroid avoidance regimen was associated
not change after trough level reduction.70 Corticosteroid with excellent medium-term patient and graft outcomes
withdrawal resulted in a decrease in insulin resistance and and a low incidence of side effects. Most 10-year outcomes
a reduction in lipid levels; reduction of tacrolimus trough were described in a protocol incorporating discontinuation
levels also improved glucose metabolism. of steroids at postoperative day 7. The 10-year graft survival
A randomized multicenter double-blinded trial was per- was 61% for living donor transplant and 51% for deceased
formed to determine whether steroid avoidance reduced donor transplants, comparable to 10-year Scientific Regis-
the risk of new onset diabetes posttransplant in the setting try of Transplant Recipients national data.75 The Univer-
of CNI therapy. Patients were randomized 1:1 to either ste- sity of Minnesota’s prospective, randomized trial evaluated
roid withdrawal (n = 142) or continuation (n = 135) on outcomes of three different maintenance immunosup-
posttransplant day 8 in addition to an immunosuppressive pression protocols after early steroid withdrawal (6 days
regimen of tacrolimus and MMF.71 Low-dose prednisone (5 posttransplantation) in 440 kidney transplant recipients.
mg/d) was utilized in the steroid continuation arm starting Patients were randomized to cyclosporine/MMF (n = 151),
6 months posttransplant. At 5 years posttransplant, 36.3% high-dose tacrolimus/low-dose sirolimus (n = 149), or low-
of steroid continuation patients and 35.9% of steroid with- dose tacrolimus/high-dose sirolimus (n = 140). All patients
drawal patients had developed diabetes (P = 0.98). The received thymoglobulin induction. The 10-year graft and
percentage of patients requiring insulin therapy was lower patient survival did not differ between the three treatment
in the steroid withdrawal group compared with the steroid arms. There were no differences in 10-year rates of BPAR.
continuation group (3.7% vs. 11.6%, P = 0.049). However, The rate of new-onset diabetes was highest in the high-dose
through 5 years posttransplant the proportion of new onset tacrolimus group at 10 years (19%).76 The efficacy of early
diabetes patients requiring treatment was similar between steroid withdrawal compared with chronic steroid mainte-
the withdrawal and continuation group. This study sug- nance has also been shown to be a reasonable approach in
gests that steroid withdrawal has limited effect in prevent- repeat kidney transplant recipients, with results showing
ing new onset diabetes posttransplant. similar graft and patient survivals at 1 and 5 years.77
The effect on tacrolimus exposure in the setting of early
EARLY CORTICOSTEROID WITHDRAWAL steroid withdrawal was evaluated in a prospective, ran-
domized, multicenter study that included 397 patients.
REGIMENS
Patients were assigned on posttransplant day 8 to receive
The safety of early corticosteroid withdrawal (see Chapter either steroid withdrawal (n = 191) or continuation (n =
14) was evaluated by a prospective randomized multicenter 195). Significantly higher tacrolimus trough levels were
double-blind study of early (7 days posttransplantation) observed in the steroid withdrawal group compared with
corticosteroid cessation versus long-term maintenance the steroid continuation group at 2 weeks posttransplant,
with corticosteroids along with tacrolimus, MMF, and although this interaction was not seen in the African Amer-
antibody induction in primary renal transplant patients.72 ican subgroup.78
Patient and graft survivals at 1 year were 98% and 96%, Another randomized multicenter study assessed which
respectively. BPAR occurred in 9.8% of patients, and 4% induction agent was most effective in allowing rapid ste-
were treated empirically for rejection. One-year analysis roid withdrawal (day 8 posttransplant). In total, 615 low-
suggested that early withdrawal of corticosteroids was safe, immunologic-risk patients were randomly assigned (1:1:1)
resulting in excellent patient and graft survival, low acute to basiliximab/steroids (n = 215), basiliximab/steroid with-
rejection rates, and no graft loss to rejection72,73 Five-year drawal (n = 197), or thymoglobulin/steroid withdrawal
results confirmed that early corticosteroid withdrawal pro- (n = 203), in addition to a maintenance immunosuppres-
vides a similar long-term renal allograft survival and func- sion protocol of tacrolimus and MMF. At 1 year, BPAR rates
tion; however, early corticosteroid withdrawal is associated were similar at 9.9% for the thymoglobulin arm, 11.2% for
with a higher incidence of mild, Banff 1A, steroid-sensitive the basiliximab/steroids arm, and 10.6% for the basilix-
episodes of rejection. Steroid withdrawal provided improve- imab/steroid withdrawal arm.79 Patient and graft survival
ments in cardiovascular risk factors (triglyceride, diabetes, were comparable and excellent in all treatment groups at
weight gain).73 A randomized trial comparing cyclospo- 12 months.
rine versus tacrolimus as long-term immunosuppression
regimens after early steroid withdrawal 5 days after renal CORTICOSTEROID-FREE IMMUNOSUPPRESSION
transplantation showed that tacrolimus was more effective REGIMENS
in enabling patients to remain on a steroid-free regimen at
3 years (88% vs. 65%; P < 0.001).59 A 6-month open-label multicenter parallel-group study
In another study, 101 patients underwent renal trans­ included 538 renal patients randomly assigned (1:1) to
plantation with tacrolimus, MMF, and 7 days of corticoste- a daclizumab/tacrolimus/MMF regimen (n = 260) or a
17 • Calcineurin Inhibitors 249

tacrolimus/MMF/corticosteroid regimen (n = 278).15 The rates to be unacceptably high among African American
incidence of BPAR was 16.5% in both treatment groups; recipients83 A study examining corticosteroid withdrawal
the incidence of biopsy-proven corticosteroid-resistant in 52 stable renal transplant recipients treated with tacro-
acute rejection was 4.3% and 5% in the tacrolimus/MMF/ limus and MMF showed a 98% patient survival and 92.3%
corticosteroids and daclizumab/tacrolimus/ MMF groups (P graft survival.84 The tacrolimus-based regimen was thought
= NS). The median serum creatinine level at 6 months and to promote compliance by facilitating steroid withdrawal
overall safety profile were similar with both regimens. and reducing cosmetic complications (see Chapter 16). A
A single-center, nonrandomized, retrospective sequential prospective randomized study comparing 5-year outcomes
study was used to evaluate outcomes in kidney transplant in kidney recipients maintained on four different CNIs based
recipients given either alemtuzumab (Campath) (n = 123) immunosuppression protocols (cyclosporine/MMF, cyclo-
or basiliximab (n = 155) in combination with a prednisone- sporine/sirolimus, tacrolimus/MMF, tacrolimus/sirolimus)
free maintenance protocol using tacrolimus and MMF.80 with basiliximab induction without long-term steroid ther-
There was no significant difference in the 3-year graft and apy showed acceptable patient and graft survival at 5 years
patient survival rates between the two groups. A lower rate in all four groups.85
of early (<3 months) rejection was observed in the alem-
tuzumab (4.1%) versus the basiliximab (11.6%) group, but CALCINEURIN INHIBITOR AVOIDANCE AND
rejection rates for both groups were equivalent at 1 year. LOW-DOSE TACROLIMUS REGIMENS
Patient and graft survival and rejection rates were nearly
identical between whites and African Americans receiving Transitioning to an inhibitor of mammalian target of
alemtuzumab. The quality of renal function and the inci- rapamycin (mTOR)-based immunosuppressive regimen is a
dence of infectious complications were similar between the strategy to avoid nephrotoxicity caused by CNIs. The ELE-
alemtuzumab and basiliximab groups. VATE trial, a multicenter study, compared de novo adult
Two corticosteroid-free, tacrolimus-based regimens were kidney recipients randomized at 10 to 14 weeks to con-
compared with standard triple therapy in a 6-month phase vert to everolimus (n = 359) or to remain on CNI therapy
III open-label parallel-group multicenter study.81 A total (n = 356; 231 tacrolimus; 125 cyclosporine) in addition to
of 451 patients were randomly assigned (1:1:1) to receive MMF and steroids.86 At 12 months, the primary endpoint
tacrolimus/MMF/corticosteroids, tacrolimus/ MMF with- of GFR was similar between the two groups and biopsy-
out induction, or tacrolimus monotherapy with basiliximab proven rejection was more frequent in everolimus-treated
induction. The incidences of BPAR were 8.2% (triple ther- patients compared with tacrolimus-treated patients (9.7%
apy), 30.5% (tacrolimus/MMF), and 26.1% (basiliximab/ vs. 2.6%, P < 0.001). In a similar study, the ZEUS trial, 300
tacrolimus) (P = 0.001). The incidences of corticosteroid- renal transplant recipients were randomized to continue
resistant acute rejection were similar among the groups (P receiving cyclosporine or to convert to everolimus at 4.5
= NS). Graft and patient survival rates were similar among months posttransplant.87 At 5 years, mean eGFR was 66.2
the groups. Overall safety profiles were similar. Differences mL/min/1.73 m2 with everolimus and 60.9 mL/min/1.73
were noted for anemia (24.5% vs. 12.6% vs. 14.5%), diar- m2 with cyclosporine (P < 0.001), whereas rates of biopsy-
rhea (12.9% vs. 17.9% vs. 5.9%), and leukopenia (7.5% proven rejection were higher with everolimus (13.6%
vs. 18.5% vs. 5.9%) for the triple-therapy, tacrolimus/ vs. 7.5%, P = 0.095). A meta-analysis of 29 randomized
MMF, and basiliximab/tacrolimus groups, respectively. controlled trials investigating conversion of CNI to mTOR
Both corticosteroid-free regimens were equally effective in inhibitor determined that patients converted to mTOR ther-
preventing acute rejection, with the basiliximab/tacroli- apy had a higher GFR and rate of acute rejection at 1 year.88
mus regimen offering some safety benefits.81 A prospective A prospective, randomized trial in renal transplantation
randomized trial was done comparing tacrolimus/sirolimus compared sirolimus/MMF/prednisone (n = 81) with tacro-
with tacrolimus/mycophenolate in renal transplant recipi- limus/MMF/prednisone (n = 84). The mean follow-up was
ents using a prednisone-free regimen with more than 8.5 33 months. There was no difference in patient survival,
years of follow-up. All patients received anti-IL-2 receptor graft survival, or the incidence of clinical acute rejection
antagonist (basiliximab). The tacrolimus/MMF group had between the two groups. There was also no difference in
overall better renal allograft survival (91% vs. 70%, P = the mean GFR measured by iothalamate clearance between
0.02); 13 patients (35.1%) in the tacrolimus/sirolimus the tacrolimus and sirolimus groups at 1 or 2 years.89 At
group and 8 patients (17.8%) in the tacrolimus/MMF group 1 year, chronicity using the Banff schema showed no dif-
experienced biopsy-proven ACR (P = 0.07). By 3 months ference in interstitial, tubular, or glomerular changes, but
posttransplant, estimated GFR was significantly lower in fewer chronic vascular changes in the sirolimus group.
the tacrolimus/sirolimus group compared with the tacro- This study suggests that many of the promises of CNI-free
limus/MMF group (47.7 vs. 59.6 mL/min/1.73 m2; P = immunosuppression have perhaps not been achieved with
0.0002), and this trend persisted throughout the follow-up short-term follow-up. The question of improved safety and
period.82 efficacy in the longer term with CNI-free immunosuppres-
sion has to be subjected to longer-term follow-up of the
COMPARISON OF CORTICOSTEROID-SPARING aforementioned study and similar studies.90 Many previous
REGIMENS USING TACROLIMUS-BASED AND studies of complete CNI avoidance have used cyclosporine
as the comparator drug. The 15-year follow-up data of a
CYCLOSPORINE-BASED IMMUNOSUPPRESSION
multicenter randomized trial of CNI avoidance used cyclo-
Studies of corticosteroid-sparing protocols in patients sporine in the CNI arm. Renal transplant recipients initially
treated with cyclosporine and MMF showed acute rejection treated with MMF/cyclosporine/steroids were randomized
250 Kidney Transplantation: Principles and Practice

at 6 months posttransplant to MMF/steroids (n = 63), MMF/ months posttransplant without de novo donor-specific anti-
cyclosporine (n = 76), or MMF/cyclosporine/steroids (n = bodies (DSAs) or prior episode of acute rejection were ran-
73). Fifteen years after conversion to a CNI-free regimen, domized to tacrolimus withdrawal (n = 14) or continuation
there was no difference in patient or graft survival between (n = 14) in addition to induction with thymoglobulin, MMF,
the groups.91 Patients treated with CNI withdrawal expe- and steroids.97 The study was prematurely stopped because
rienced higher rates of acute rejection, and early rejection of high rates of rejection (4 of 14) and de novo DSAs (5 of
after CNI withdrawal was associated with decreased graft 14) in the tacrolimus withdrawal arm. In a similar study,
survival. There was no long-term benefit of CNI withdrawal low-immunologic-risk patients at least 4 years posttrans-
on the development of comorbidities such as diabetes mel- plant were randomized to tacrolimus weaning (n = 5) or
litus, malignancy, or cardiovascular disease. continuation (n = 5).98 All five patients in the withdrawal
A meta-analysis of 27 randomized controlled trials with group had to be restarted on tacrolimus because of the
4105 renal transplant recipients examined both CNI avoid- development of acute rejection or de novo DSAs, whereas
ance and withdrawal trials.92 The analysis concluded CNI all five patients in the control group remained stable. These
avoidance/ withdrawal protocols have better graft function two trials suggest that even in highly selective stable kidney
at 1- and 2-year follow-up, however CNI avoidance proto- recipients, tacrolimus withdrawal is to be avoided.
cols have rate rates of acute rejection at 1 year, although
the rates are similar to CNI groups at 2 years. Overall 1- and OTHER CALCINEURIN INHIBITORS AND
2-year patient and graft survival were similar.89 A meta- FORMULATIONS
analysis of 19 randomized controlled trials with analysis
of 3312 renal transplant recipients with median follow- One of the major risk factors for graft failure is noncom-
up of 12 months showed that CNI-sparing strategies with pliance with medication regimens. Advagraf (Tac-OD), a
adjunctive mycophenolate can achieve comparable short- once-daily formulation, was introduced by Astellas in 2008
term graft function.93 to potentially improve medication compliance. In phase I
A lower tacrolimus exposure regimen was evaluated by and II trials, Advagraf showed equivalent pharmacokinetic
Ekberg et al.94 In this trial, 1645 renal transplant recipients parameters (C0 and AUC0–24) compared with the twice-
were randomly assigned to receive standard-dose cyclospo- daily Prograf formulation (Tac-BID) in healthy controls, de
rine, MMF, and corticosteroids, or daclizumab induction, novo kidney transplant recipients, and stable kidney trans-
MMF, and corticosteroids in combination with low-dose plant recipients after conversion. In a phase III open-label
cyclosporine (target trough 50–100 ng/mL), low-dose randomized study, 4-year safety and efficacy profiles were
tacrolimus (target trough 3–7 ng/mL), or low-dose siroli- similar among patients receiving Advagraf (Tac-OD) and
mus (target trough 3–7 ng/mL). At 12 months the mean Tac-BID.99 The OSAKA trial randomized patients to Tac-
calculated GFR was higher in patients receiving low-dose BID 0.2 mg/kg per day (n = 237), Tac-OD 0.2 mg/kg per
tacrolimus (65.4 mL/min) than in the other three groups. day (n = 263), Tac-OD 0.3 mg/kg per day (n = 246) all with
The rate of BPAR was lower in patients receiving low-dose MMF and steroids, or Tac-OD 0.2mg/kg per day (n = 230)
tacrolimus (12.3%) than in those receiving standard-dose with MMF, basiliximab, and perioperative steroids.100 The
cyclosporine (25.8%), low-dose cyclosporine (24.0%), or study showed comparable outcomes between Tac-BID and
low-dose sirolimus (37.2%). One-year allograft survival dif- Tac-OD 0.2 mg/kg for its primary, composite endpoint of
fered significantly among the four groups (P = 0.02) and efficacy failure (42.2% vs. 40.6%). Advagraf was approved
was highest in the low-dose tacrolimus group (94.2%), for use in the US in 2013. It is currently approved as main-
followed by the low-dose cyclosporine group (93.1%), the tenance immunosuppression with MMF and steroids, with
standard-dose cyclosporine group (89.3%), and the low- or without basiliximab induction.
dose sirolimus group (89.3%).94 An observational 2-year Envarsus XR (Tac-ER) is another once-daily formulation
follow-up to this study showed that the mycophenolate and introduced by Veloxis to help with posttransplant immu-
low-dose tacrolimus (GFR 68.6 ± 23.8 mL/min) arm con- nosuppression adherence. Its design allows for improved
tinued to have the highest GFR compared with standard- bioavailability by decreasing the drug’s particle size via
dose cyclosporine, low-dose cyclosporine, and low-dose MeltDose (Veloxis Pharmaceuticals) technology.101 It was
sirolimus (65.9 ± 26.2, 64.0 ± 23.1, and 65.3 ± 26.2 mL/ granted approval by the FDA in 2015 as prophylaxis for
min, respectively), but the difference was not significant (P rejection in patients converting from twice-daily tacrolimus
= 0.17). The mycophenolate and low-dose tacrolimus arm formulations. Envarsus’ noninferiority was demonstrated
also had the highest graft survival rate, but with reduced in the phase III MELT trial.102 Stable patients between 3
differences between groups over time, and the least acute months and 5 years posttransplant were randomized to
rejection rate.95 Another trial evaluating low-dose tacroli- either conversion from Tac-BID to Tac-ER (n = 163) or to
mus randomized renal transplant recipients to either low- remain on Tac-BID (n = 163). Throughout the study the
dose tacrolimus/everolimus or standard-dose tacrolimus/ mean daily dose of Tac-ER was significantly lower than the
MMF with steroids and induction therapy. The rate of BPAR preconversion dose (P = 0.0001). The primary endpoint
was significantly higher (19.1% vs. 11.2%, P < 0.05) in the of efficacy failure rate was 2.5% for both arms. Patients
low-dose tacrolimus/everolimus group, whereas mean GFR receiving Tac-ER experienced more drug discontinuation
(63.1 vs. 63.1 mL/min) and safety were comparable at 1 within 12 months (12% vs. 5%, P = 0.028), although the
year.96 incidence of adverse events was similar between the two
The effects of tacrolimus withdrawal were studied in a groups. Another phase III multicenter trial evaluated the
multicenter prospective randomized trial of immunologi- efficacy and safety of Envarsus in adult de novo kidney
cally quiescent kidney transplant recipients. Patients 6 transplant recipients by randomizing patients to either
17 • Calcineurin Inhibitors 251

Envarsus (Tac-ER; n = 268) or Tac-BID (n = 275). The corticosteroids. The primary endpoint was incidence and
overall incidence of treatment failure (death, graft failure, time to first acute rejection. At 1 year, tacrolimus therapy
biopsy-proven acute cellular rejection, or lost to follow- was associated with a significantly lower incidence of acute
up) at 12 months was 18.3% in the Envarsus group and rejection (36.9%) compared with cyclosporine (59.1%; P =
19.6% in the Tac-BID. This treatment difference was below 0.003). At 4 years, patient survival was similar, but graft
the 10% noninferiority margin. Overall, patients receiv- survival significantly favored tacrolimus over cyclosporine
ing Envarsus had a cumulative drug dose 14% lower com- (86% vs. 69%; P = 0.025). At 1, 2, 3, and 4 years, the mean
pared with Tac-BID, and target trough levels were achieved GFR was significantly better in the tacrolimus group than
faster.101 in the cyclosporine group. Three patients in each arm devel-
Voclosporin (VCS, ISA247) is a novel CNI in develop- oped posttransplant lymphoproliferative disorder, and the
ment for organ transplantation. PROMISE was a phase incidence of diabetes mellitus was similar in the two groups.
IIb 6-month multicenter randomized open-label study of Tacrolimus was significantly more effective than cyclospo-
three ascending concentration-controlled groups of VCS rine in preventing acute rejection in pediatric renal trans-
(low, medium, and high) versus tacrolimus in 334 de novo plant recipients. Renal function and graft survival also were
kidney transplant recipients. This 6-month study showed superior with tacrolimus.111 The addition of basiliximab
VCS to be as efficacious as tacrolimus in preventing acute to a tacrolimus-based regimen (tacrolimus/azathioprine/
rejection (VCS low 10.7%, medium 9.1%, and high 2.3% steroids) did not show any improved clinical efficacy at 6
vs. tacrolimus 5.8%) with similar renal function in the low- months in a pediatric patient population with similar BPAR
and medium-exposure groups.103,104 rates (20.4% without basiliximab vs. 19.2% with basil-
Sandimmune, the original oil-based formulation of cyclo- iximab) and GFR (79.4 without basiliximab vs. 77.6 mL/
sporine (Sandimmune; Novartis, Basel, Switzerland), was min/1.73 m2 with basiliximab).112 According to the 2014
introduced in 1983. Although a significant advance in NAPRTCS, less than 3% of patients were being treated with
immunosuppressive therapy, this formulation had numer- cyclosporine 30 days posttransplant, whereas more than
ous problems. Absorption was slow and showed a great deal 90% of patients were being treated with tacrolimus.113
of intrapatient and interpatient variability, making dosing The effect of corticosteroids on the epiphyseal growth
difficult and increasing the risk of chronic rejection.105,106 plates is well recognized and results in irreversible growth
In 1995 Neoral (Sandimmune Neoral; Novartis, Basel, stunting. Experience with corticosteroid withdrawal under
Switzerland), a microemulsion formulation of cyclosporine, tacrolimus therapy in pediatric patients has been associated
was approved for use by the FDA. This formulation improved with favorable outcomes. Two-thirds of the pediatric kidney
bioavailability with more rapid absorption and less variabil- transplant recipients were withdrawn successfully from
ity in de novo and stable transplant patients.106,107 Since its corticosteroids, with a low incidence of graft dysfunction or
introduction, numerous randomized and nonrandomized acute rejection (23%).114 Many of these patients had remark-
studies have showed lower acute rejection rates, although able catch-up growth. The Tacrolimus and Withdrawal
long-term patient and graft survival compared with the of Steroids (TWIST) trial was a randomized study evaluat-
Sandimmune formulation are similar.108,109 ing the effect of early steroid withdrawal on posttransplant
growth. A total of 196 patients were randomized to either
early steroid withdrawal with tacrolimus/MMF/daclizumab
PEDIATRIC RENAL TRANSPLANTATION
or steroid continuation with tacrolimus/MMF.115 At 1 year,
See Chapter 37 for a more detailed discussion of pediatric the steroid withdrawal group (n = 54) grew better than the
renal transplantation. The efficacy of tacrolimus as an immu- steroid continuation (n = 59) cohort (0.25 difference in
nosuppressive agent in pediatric renal transplantation has adjusted mean height change, P = 0.001). Growth was sus-
been shown in single-center experiences and in multicenter tained at 2 years in the withdrawal group and significantly
trials. A retrospective cohort study of 986 pediatric renal better in prepubescent children (0.50 difference in adjusted
transplant recipients in the North American Pediatric Renal mean height change, P =. 0.004).
Transplant Cooperative Study (NAPRTCS) database (index Changes in kidney function, mixed lymphocyte culture,
renal transplant 1997 through 2000), who were treated cell-mediated lympholysis, cytotoxic antibodies, lympho-
with either cyclosporine/MMF/steroids (n = 766) or tacro- cyte populations, and cytokine response were studied in
limus/MMF/steroids (n = 220), was performed to examine 14 pediatric renal transplant recipients with chronic rejec-
differences in outcome between these two groups.110 In tion who were converted to tacrolimus from cyclosporine.
this analysis, tacrolimus and cyclosporine, in combination Serum creatinine levels decreased, creatinine clearance
with MMF and steroids, were associated with similar rejec- increased, and urinary protein excretion decreased after 6
tion rates and graft survival in pediatric renal transplant months, and these values were maintained after 2 years
recipients. Tacrolimus was associated with improved graft under tacrolimus treatment.58
function at 1 and 2 years after transplantation. A 6-month In one study eight pediatric renal transplant recipients
randomized prospective open parallel-group study with an (median age at transplant 2 years; range 1.2–12.9 years)
open extension phase was conducted in 18 centers from were converted to tacrolimus- and sirolimus-based immu-
nine European countries to compare the efficacy and safety nosuppression as rescue therapy. All patients had biopsy-
of tacrolimus with cyclosporine in pediatric renal trans- proven chronic allograft nephropathy. After the addition
plant recipients.111 The study randomly assigned (1:1) of sirolimus, the median dose required to keep tacroli-
196 pediatric patients (<18 years old) to receive either mus blood trough concentrations within the target range
tacrolimus (n = 103) or cyclosporine microemulsion (n = increased by 71.2% (range 21.9%–245.4%), and the dose-
93), administered concomitantly with azathioprine and normalized tacrolimus exposure (AUC) decreased to 67.1%.
252 Kidney Transplantation: Principles and Practice

Adding sirolimus to tacrolimus-based immunosuppression or gastrointestinal side effects.120 All rejection episodes
in young pediatric renal transplant recipients resulted in a responded to steroids.
significant decrease in tacrolimus exposure.116 Immunosuppression for SPK transplantation at North-
western University was divided into four eras over an 8.5-
year period.121 In era I (March 1993 to February 1997),
Clinical Studies in Kidney– three immunosuppression combinations were used: cyclo-
Pancreas Transplantation sporine/azathioprine/steroids (n = 28), cyclosporine/MMF/
steroids (n = 8), or tacrolimus/MMF/steroids (n = 10);
The increase in the number of pancreas transplants has been bladder drainage was used. In era II (July 1995 to Febru-
made possible by technical improvements and improved ary 1998), the combination of tacrolimus, MMF, and cor-
immunosuppressive regimens (see Chapter 36). Treatment ticosteroids was used, with bladder drainage. In era III,
with tacrolimus-based immunosuppression has been asso- combinations of tacrolimus (12-hour trough concentra-
ciated with lower rejection rates, higher graft survival rates, tions 10–12 ng/mL) and MMF (3 g/day) were used along
and less nephrotoxicity compared with treatment with with corticosteroids for maintenance immunosuppression;
cyclosporine.6,117 enteric drainage was used. In era IV, steroids were elimi-
nated within 6 days of transplantation, and tacrolimus was
SIMULTANEOUS PANCREAS–KIDNEY combined with either MMF (n = 20) or sirolimus (n = 38);
enteric drainage was used.
TRANSPLANTATION
In eras I and II, all recipients received induction therapy
In an analysis of 1194 pancreas transplantations per- with Atgam for 7 to 14 days after transplantation. In era III,
formed at the University of Minnesota, the results were for induction therapy, 17 patients were randomly assigned
divided into five time periods, or eras, based on the tech- to a noninduction therapy arm, and 37 patients were ran-
nique and immunosuppressive regimen used.118 In era II, domly assigned to an anti-IL-2 receptor monoclonal anti-
the immunosuppressive regimen consisted of Minnesota body (daclizumab, n = 35; basiliximab, n = 2). Induction
antilymphocytic globulin (MALG) or muromonab-CD3 therapy in era IV consisted of rATG, 1 mg/kg intraopera-
(OKT3) for induction and a combination of cyclosporine, tively and on postoperative days 1, 2, 4, 6, 8, 10, 12, and
azathioprine, and prednisone for maintenance. Duct man- 14. One-year actuarial patient survival rates in eras III and
agement in eras II and III was by bladder drainage. In era IV were 96.3% and 100%, respectively; 1-year actuarial
III, tacrolimus was used for pancreas transplantation as kidney survival rates in eras III and IV were 94.4% and
soon as it was approved by the FDA in 1994. Induction 97.7%, respectively, and the 1-year actuarial pancreas sur-
was with equine ATG (Atgam), and OKT3 was used for vival rates were 88.9% and 100%, respectively. The 1-year
treatment of rejection episodes. When MMF was approved rejection-free rate was 87.1% for era III and 96.6% for
a year later, it was added to the maintenance immunosup- era IV. Compared with era I, kidney function significantly
pressive regimen. In era IV, which began in March 1998, improved over the three eras. Rapid elimination of corti-
daclizumab, alone or in combination with the polyclonal costeroids was successful in all recipients in era IV, with
anti-T cell antibody (Atgam or ATG), was added to the higher patient and graft survival rates than in the previous
induction regimen. Enteric drainage was the principal exo- three eras. Rejection rates decreased further in era IV. The
crine drainage technique. In patients with primary SPK Northwestern group concluded that corticosteroids could
transplantation, pancreas and kidney graft survival rates be rapidly eliminated prospectively in all recipients without
were significantly higher in eras III and IV than in era II. In a decrease in graft survival rates or an increase in the rate
eras III and IV combined, 1-year patient, pancreas, and kid- of rejection.121
ney survival rates were 92%, 79%, and 88%, respectively; The incidence of cytomegalovirus and malignancies was
at 5 years, the corresponding figures were 88%, 73%, and not higher using tacrolimus/MMF compared with the cyclo-
81%, respectively.118 sporine/azathioprine regimen, with 5 years’ follow-up.
In a prospective, randomized trial, 42 SPK recipients A multicenter trial was done to assess the effect of anti-
received ATG and daclizumab induction, with tacrolimus body induction in SPK transplant recipients receiving
and steroids as baseline immunosuppression. Twenty-two tacrolimus, MMF, and corticosteroids.103 The trial ran-
patients were randomly assigned to receive MMF, and 20 domly assigned 174 SPK transplant recipients to induction
patients received sirolimus in addition to tacrolimus and (n = 87) or noninduction (n = 87), and the recipients were
steroids. Actuarial patient, kidney, and pancreas allograft followed for 3 years. Induction agents included T cell deplet-
survivals were 100%, 100%, and 95%, respectively, at 6 ing or IL-2 receptor antibodies. At 3 years, actual patient
months in the sirolimus group and 100%, 100%, and 100%, (94.3% and 89.7%) and pancreas (75.9% and 75.9%) sur-
respectively, in the MMF group. The incidence of acute vival rates were similar in the induction and noninduction
rejection was less than 10% and was limited to instances in groups. Actual kidney survival was significantly better
which recipient immunosuppression was reduced.119 in the induction group compared with the noninduction
A prospective study of combined tacrolimus, MMF, and group at 3 years (92% vs. 82%; P = 0.04).103
steroids without antibody induction was done in 17 SPK Another study evaluated the safety and efficacy of dacli-
transplant patients. Low-dose intravenous tacrolimus was zumab and ATG induction alongside triple therapy in SPK
used as induction therapy. Clinical and biopsy-proven transplantation. Thirty-nine patients were randomized
rejection occurred in four (23%) patients. Patients who to daclizumab (n = 20) or ATG (n = 20) induction and
developed rejection had low tacrolimus levels or had had dis- compared with patients on triple therapy alone.122 Over-
continuation of MMF because of leukopenia, gastroparesis, all patient, kidney, and pancreas graft survival were not
17 • Calcineurin Inhibitors 253

different between the induction and no induction arms at inclusion criteria: (1) low-maintenance steroid dose 0.075
1 year (97%/93%, 97%/96%, and 92%/96%, respectively). mg/kg/day; (2) MMF dose ≥750 mg orally twice a day; and
Both induction groups had significantly lower rates of rejec- (3) tacrolimus levels ≥8 ng/mL. Fifty-five patients (29 SPK,
tion at 6 months compared with triple therapy alone (P < 26 PAK) were randomly assigned to remain on steroids or
0.005). to steroid withdrawal after 4 to 8 weeks. The median fol-
The EuroSPK Study Group, which compared tacrolimus low-up was 27 months in the SPK category and 26 months
and cyclosporine in primary SPK transplantation, enrolled in the PAK category, and from randomization, 10 months
205 patients.123 After antilymphocyte globulin induction, in both categories. Steroid withdrawal 6 months after a
patients were randomly assigned to receive either tacroli- successful pancreas transplant was not associated with a
mus or cyclosporine microemulsion together with MMF decrease in patient or graft survival, and it was not associ-
and steroids. At 1 year after transplantation, patient and ated with an increase in the incidence of rejection or in the
kidney survival rates were excellent in both treatment rate of graft loss from rejection. There was a better quality of
groups. There was a significant difference in pancreas graft life and a reduction in serum cholesterol levels in the steroid
survival: 94.2% for tacrolimus and 73.9% for cyclospo- withdrawal group.128
rine (P = 0.00048). There were significantly fewer grade Rapid corticosteroid elimination was carried out in 40
2 and grade 3 rejections with tacrolimus-based therapy. SPK recipients from Northwestern University in Chicago.129
The EuroSPK group also presented data showing that 34 ATG was used for induction; maintenance immunosuppres-
patients were switched from cyclosporine to tacrolimus, but sion was with tacrolimus/MMF in 20 patients and tacroli-
only 6 patients receiving tacrolimus required conversion to mus/sirolimus in 20 patients. Patient and graft survival
alternative therapy during the course of the study.124 The rates and rejection rates were compared with historical
mean doses of MMF at 1 year also were lower in the tacro- controls (n = 86). One-year actuarial patient, kidney, and
limus group (1.36 g/day vs. 1.67 g/day; P = 0.007). Good pancreas survival rates in the rapid corticosteroid elimina-
results have been shown in smaller studies as well (17 SPK tion group were 100%, 100%, and 100%, respectively, and
transplantation, 5 PAK transplantation, and 6 pancreas in the historical control group rates were 97%, 93%, and
transplant alone) with the use of alemtuzumab 30 mg 97%, respectively. The 1-year rejection-free survival rate
induction, Prograf and mycophenolate immunosuppres- was 97% in the rapid corticosteroid elimination recipients
sion, with patient and graft survival of 100% and 100%, versus 80% in the historical controls. Serum creatinine lev-
respectively at 3 years.125 els remained stable in all groups at 6 and 12 months after
transplantation.129
STEROID WITHDRAWAL AND STEROID-FREE Steroid-free immunosuppression has been used with
excellent short-term results in low-risk pancreas–kidney
PROTOCOLS
transplantation recipients at the University of California at
Reduction of steroid use is extremely desirable in pancreas San Francisco.130 Forty patients underwent pancreas–kid-
transplantation because long-term steroid use is associated ney transplantation from November 2000 to July 2002.
with hypertension, hyperlipidemia, and glucose intoler- ATG induction was combined with MMF, tacrolimus, and
ance.51 Complete steroid withdrawal was achieved in 58 sirolimus for maintenance immunosuppression. Steroids
(47%) of 124 patients transplanted at the University of were used as pretreatment only, given with ATG and dis-
Pittsburgh, with a mean time to steroid withdrawal of 15.2 continued by the end of the first postoperative week. Patient,
± 8 months.126 Patient, pancreas, and kidney survival rates kidney, and pancreas survival rates were 95%, 92.5%, and
at 1 year were 100%, 100%, and 98%, respectively, (off 87.5%, respectively. Biopsy-proven pancreas rejection
steroids) versus 97%, 91%, and 96%, respectively, (on ste- rates at 1 and 3 months after transplantation were 2.5%,
roids; all P = NS). The cumulative risk of rejection was 74% and kidney rejection rates at 1 and 3 months were 2.5%.130
for patients off steroids versus 76% for patients on steroids
(these patients had not received antibody induction). The PANCREAS AFTER KIDNEY TRANSPLANTATION
mean glycosylated hemoglobin levels were 5.2 ± 0.9% (off
steroids) and 6.2 ± 2.1% (on steroids; P = 0.02). The Pitts- According to data from the International Pancreas Trans-
burgh group concluded that steroid withdrawal could be plant Registry, the current, nearly uniform use of tacroli-
achieved in pancreas transplant patients under tacrolimus- mus/MMF in PAK transplantation makes a comparison
based immunosuppression and was associated with excel- with other regimens difficult, although graft survival rates
lent patient and graft survival.126 have been significantly better than in the preceding era,
A single-center retrospective sequential study from when cyclosporine/azathioprine was used.131
Northwestern University of 59 SPK patients demonstrated In the overall analysis of tacrolimus/MMF-treated pri-
the efficacy of a steroid-free immunosuppression regimen mary PAK transplant recipients, graft survival rates did not
at 5 years with a kidney allograft survival of 90.7% and a differ significantly whether or not antibody induction was
pancreas allograft survival of 100% in the setting of thymo- given, although they tended to be numerically higher in PAK
globulin induction, tacrolimus/MMF regimen.127 recipients given depleting or nondepleting antibody than in
The Minnesota Group reported a prospective trial of ste- recipients not given antibody induction. In the PAK cate-
roid withdrawal in pancreas transplantation.128 Recipi- gory, the relative risk of pancreas graft failure was reduced
ents with functioning grafts ≥6 and 36 months after SPK by the use of tacrolimus/MMF for immunosuppression.131
transplantation or PAK transplantation were enrolled. All Between July 1, 1978, and April 30, 2002, 406 PAK
patients received triple therapy for maintenance immuno- transplants were performed at the University of Minne-
suppression using tacrolimus and MMF, with the following sota.132 Immunosuppression was divided into eras. In era
254 Kidney Transplantation: Principles and Practice

III, tacrolimus was used in combination with prednisone and vacuolization of smooth-muscle cells to thrombotic
and initially azathioprine. MMF replaced azathioprine microangiopathy.
when it was approved by the FDA. Polyclonal antibody A review of 21 patients with tacrolimus-associated
induction therapy with Atgam was used in 99%, and OKT3 thrombotic microangiopathy was published133; 17 of these
was used in 1% of patients; the median duration of antibody occurred in kidney transplant recipients, whereas two cases
therapy was 5 days. In era IV, tacrolimus, MMF, and pred- occurred in liver transplant recipients and one each in heart
nisone were the principal maintenance immunosuppres- and bone marrow transplant recipients. The mean time
sive agents. Daclizumab was used for induction either alone from transplantation to the onset of thrombotic microangi-
(21%) or in combination (79%) with a polyclonal antibody opathy was 9.3 ± 7.9 months. Clinical presentation varied
(Atgam or ATG). The median duration of antibody therapy from an absence of signs and symptoms of hemolysis to florid
was 3 days. Overall patient survival rates (deceased and liv- hemolytic anemia, thrombocytopenia, and azotemia. Renal
ing donor) at 1 and 3 years were 97% and 90%, respectively, biopsy specimens were obtained and showed acute thrombi
and at 1 year in era IV overall survival was 96%. Overall within the glomerular capillaries, arterioles, or both. There
pancreas graft survival rates (deceased and living donor) at are large circulating polymers of von Willebrand’s factor in
1 and 3 years in era III were 78% and 60%, respectively, thrombotic microangiopathy, which increase the tendency
and in era IV, at 1-year overall graft survival was 77%. Of for platelets to adhere to and aggregate on the subendothe-
technically successful transplants, pancreas graft loss rates lium, resulting in thrombi and fibrin deposition. Treatment
to rejection in era III at 1 and 3 years were 10% and 19%, consists of reducing the dose of tacrolimus and substitution
respectively; in era IV, at 1 year, it was 9%. PAK transplants with cyclosporine or sirolimus. Other treatment modali-
now can be performed almost as successfully as SPK trans- ties have included plasmapheresis, fresh frozen plasma
plants; the introduction of tacrolimus and MMF in the mid- exchange, and anticoagulation.133
1990s contributed to this development. Adverse events dictate the optimal dosage regimen of the
Using tacrolimus-based and MMF-based immunosuppres- drug. Decreasing the dosage of tacrolimus generally reduces
sion, only 20% of recipients experiencing rejection episodes its toxic effects, although some adverse effects are idiosyn-
ultimately lost their pancreas graft to irreversible rejection. cratic and do not respond to such measures.25 Tacrolimus
In eras III and IV, when tacrolimus was being used, there treatment is associated with a higher incidence of diarrhea,
no longer existed a difference in outcome between primary disturbances in glucose metabolism, and some types of neu-
transplants and second transplants. rotoxicity, but a lower incidence of hypertension and hyper-
cholesterolemia than with cyclosporine. Tacrolimus is only
SIDE EFFECTS AND TOLERABILITY OF rarely associated with the cyclosporine-specific adverse
effects of hirsutism, gum hyperplasia, and gingivitis, but it
CALCINEURIN INHIBITORS
may cause alopecia and pruritus.
The side effect profile of tacrolimus is somewhat similar to In a trial in renal transplant recipients, significantly
that of cyclosporine (Table 17.2). The physiologic effects, fewer (all P < 0.05) tacrolimus recipients (compared with
including reduction in renal blood flow and glomerular cyclosporine microemulsion recipients) experienced new-
filtration, are also similar between tacrolimus and cyclo- onset or worsening hypertension (15.7% vs. 23.2%), uri-
sporine. The pathologic manifestations of tacrolimus and nary tract disorders (4.9% vs. 9.2%), hypercholesterolemia
cyclosporine toxicity are similar in that they include tubu- (4.2% vs. 8.9%), hyperbilirubinemia (0.3% vs. 3.3%), gas-
lar vacuolization and arteriolar nodular hyalinosis that trointestinal hemorrhage (0.3% vs. 2.6%), cholestatic jaun-
are indistinguishable. Microvascular changes involving dice (0.3% vs. 2.6%), hirsutism (0% vs. 4.4%), and gum
arterioles or glomerular capillaries sometimes predomi- hyperplasia (0% vs. 4.1%).134 Tremor (12.2% vs. 4.1% of
nate, displaying a wide spectrum of severity from apoptosis patients), hypomagnesemia (6.6% vs. 1.5%), thrombosis
(4.5% vs. 1.5%, mainly affecting the dialysis access vessels),
and gastritis (3.1% vs. 0.4%) were significantly (all P <
TABLE 17.2 Adverse Effects Associated With Tacrolimus 0.05) more common in the tacrolimus group. Another less
Therapy common but serious side effect includes posterior reversible
Reduced renal blood flow, glomerular perfusion
encephalopathy syndrome with an incidence of 0.49% in a
Tubular and vascular toxicity recent report.135
Neurotoxicity Because tacrolimus and cyclosporine cause acute and
Headaches, tremors, seizures, peripheral neuropathy, paresthesias chronic nephrotoxicity, concomitant use of these two
Metabolic disturbances agents is contraindicated. Nephrotoxicity related to tacro-
Hyperkalemic, hyperchloremic acidosis
Hypomagnesemia limus treatment is dose-related and responds to dosage
Diabetes mellitus reduction.136 Mean or median serum creatinine levels in
Hyperuricemia renal transplant recipients were lower in tacrolimus-treated
Hypercholesterolemia patients, with 5 years’ follow-up, than in patients treated
Hypertension
Gastrointestinal disturbances
with cyclosporine microemulsion (or standard formula-
Diarrhea tion).42,137 Administration of other nephrotoxic agents
Anorexia, nausea, and vomiting simultaneously can exacerbate the adverse effects of tacro-
Epigastric cramping limus. Examples of such agents include aminoglycosides,
Cosmetic angiotensin-converting enzyme inhibitors, angiotensin
Alopecia
Reduced renal blood flow, glomerular perfusion receptor antagonists, amphotericin, and nonsteroidal anti-
inflammatory drugs.
17 • Calcineurin Inhibitors 255

CARDIOVASCULAR ADVERSE EFFECTS Recipient-related risk factors for PTDM include an under-
lying glucose metabolic disorder (e.g., family history of
Hyperlipidemia occurs commonly after transplantation and diabetes mellitus, older recipient age, nonwhite ethnicity,
is a risk factor for cardiovascular disease. Immunosuppres- sedentary lifestyle, higher body mass index) and hepatitis C
sion with tacrolimus-based regimens is associated with better virus positivity. Transplantation-related risk factors include
lipid profiles than cyclosporine-based regimens.4,91,138,139 acute rejection during first posttransplant year, high doses
Analysis of the US Renal Data System database showed of corticosteroids, and high tacrolimus trough levels.
that fewer tacrolimus (than cyclosporine) recipients had The risk of developing PTDM is highest in the first few
at least one new-onset hyperlipidemia code during the first months after transplantation, after which the incidence
year of treatment (11% vs. 16%; P = 0.0001); a multivari- increases more slowly. The European multicenter trial
ate analysis showed that the risk of new-onset hypertension found a 6-month PTDM incidence of 4.5% and an addi-
after transplantation was reduced by 35% under tacrolimus- tional incidence of 0.4% from 7 through 12 months.134
based immunosuppression.140 The 5-year follow-up results Many patients with PTDM can have reversal of diabetes
from a US randomized trial indicated that significantly fewer mellitus, with eventual discontinuation of insulin. In the
tacrolimus than cyclosporine recipients were receiving anti- European trial, the 1-year cumulative incidence of PTDM
hypertensive treatment (80.9% vs. 91.3%; P < 0.05).42 A with tacrolimus was 8.3%, whereas the prevalence at 1
study of 119 stable renal transplant patients on triple therapy year was 5.5%.10 In a US trial combining tacrolimus with
demonstrated decreased blood pressure with late CNI with- MMF and corticosteroids, the 1-year incidence was 6.5%,
drawal compared with MMF withdrawal.141 Follow-up data and the 1-year prevalence was 2.2%.67
from the ELEVATE trial, examining cardiovascular outcomes In the clinical setting, tacrolimus affects insulin secretion
at 2 years after patients were randomly converted from CNI but does not affect insulin resistance. In addition, PTDM
to everolimus, showed no clinically relevant effects of cardio- is probably not a separate entity but a consequence of an
vascular parameters after conversion to everolimus.142 underlying glucose metabolic disorder that is uncovered by
Concentric increases in left ventricular posterior wall immunosuppression.148 The effects of tacrolimus on insu-
and interventricular septum thickness can occur with lin release are reversible, and after the early posttransplant
tacrolimus immunosuppression in 0.1% of patients.143 This period, tacrolimus and cyclosporine are equivalent in terms
condition is reversible after dosage reduction or discontinu- of their effect on glucose metabolism.149
ation of the drug. Compared with cyclosporine-treated patients, the relative
risk for developing PTDM in tacrolimus-treated patients was
POSTTRANSPLANT DIABETES MELLITUS 1.53 (P < 0.001).150 Compared with cyclosporine-treated
patients, tacrolimus therapy was associated with a reduced
PTDM is a serious adverse effect of tacrolimus treatment; the risk of death (relative risk 0.65; P < 0.001) and graft fail-
complications of diabetes mellitus can result in decreased ure (relative risk 0.70; P < 0.001). Under tacrolimus-based
patient and graft survival.144 PTDM is defined as insulin use immunosuppression, the positive effects of lower blood
for more than 30 consecutive days in the absence of pre- pressure, less hypercholesterolemia, better renal function,
existing diabetes. The incidence of PTDM was significantly and lower fibrinogen151 offset the negative effect of PTDM.
higher in tacrolimus-treated patients than cyclosporine- Evidence from a meta-analysis suggests that targeting
treated patients (9.8% vs. 2.7%), according to a meta-analy- tacrolimus concentrations to less than 10 ng/mL minimizes
sis by Heisel et al.145 graft loss and reduces the risk of diabetes mellitus without
Disturbance in glucose metabolism, with rates as high as increasing the risk of acute rejection.152 A 2012 trial by
26% for a cyclosporine-treated group compared with 33.6% Hecking et al. suggests that treatment with basal insu-
in a tacrolimus group at 6 months, were reported in a recent lin in the immediate postoperative period may reduce the
study using a strict American Diabetes Association defini- incidence of new-onset diabetes mellitus after renal trans-
tion for new-onset diabetes after transplant and impaired plantation, presumably via insulin-mediated protection of
fasting glucose.146 The introduction of MMF and sirolimus B cells.153
and the use of combinations of these agents with tacrolimus A systematic review of nine clinical trials including
has led to a reduction in acute rejection rates and a reduc- 2323 patients examined the risk of metabolic complica-
tion in corticosteroid treatment for acute rejection episodes tions after conversion from CNI to mTOR inhibitor after
in the first year after transplantation; this has also resulted kidney transplantation.154 The analysis concluded that
in a reduced incidence of PTDM in recent years. converting from CNI to everolimus was associated with a
A study from Cleveland compared the outcomes of 56 nonsignificant risk for PTDM and a significant increase in
African American adult primary kidney transplant recipi- hypercholesterolemia.
ents treated with corticosteroids, sirolimus, and tacrolimus,
targeted to low trough blood levels, with 65 white patients
MALIGNANCIES
treated with steroids, MMF, and tacrolimus, targeted to
higher blood levels. There were no significant differences in The use of immunosuppressive agents increases the risk of
the actuarial 2-year patient, graft, and rejection-free graft malignancy, the most common being malignancies of the
survival rates between the two groups. PTDM occurred in skin and lymphoma. See Chapters 34 and 35 for a more
36% of the African American patients, however, despite detailed discussion of malignancies. All agents increase
similar doses of corticosteroids and lower trough levels these risks, and the risk is related to the intensity and
of tacrolimus, compared with 15% of white patients (P = duration of treatment. Epstein-Barr virus-related post-
0.024).147 transplant lymphoproliferative disorder is associated with
256 Kidney Transplantation: Principles and Practice

immunosuppressive treatment, with a lower risk in adults and pediatric kidney and in adult kidney–pancreas trans-
than in children. In the European Multicenter Renal Study, plantation. The key comparator for tacrolimus was cyclo-
the incidence of posttransplant lymphoproliferative dis- sporine. Tacrolimus-treated patients have been shown in
order at 1-year follow-up was 1% in the tacrolimus group multiple, earlier studies to have better graft function, fewer
and 0.7% in the cyclosporine microemulsion group.10 The episodes of acute rejection, and longer overall survival com-
incidence of posttransplant lymphoproliferative disorder in pared with cyclosporine-treated patients.11,159 Additionally,
pediatric renal transplant patients with tacrolimus immu- the cardiovascular effects of tacrolimus are more favorable
nosuppression was 0.96% based on an analysis of the than cyclosporine.43,44 Despite its side effects, the superior
NAPRTCS database.155 immunosuppressive efficacy of tacrolimus has led to its pref-
erential use in kidney and kidney–pancreas recipients. How-
ever, despite maximizing CNI-based immunosuppressive
OTHER SIDE EFFECTS
regimens, long-term renal allograft survival has remained
Tacrolimus-treated patients are more likely to have alo- static. The nephrotoxicity prevalent with chronic use of CNIs
pecia, tremor, headache, insomnia, dyspepsia, vomiting, has limited long-term outcomes of renal transplantation.
diarrhea, and hypomagnesemia than cyclosporine-treated
patients.152 Cyclosporine-treated patients are more likely to References
have constipation, hirsutism, and gingival hyperplasia. 1. Dreyfuss M, Härri E, Hofmann H, et al. Cyclosporin A and C: new
metabolites from Trichoderma polysporum. Eur J Appl Microbiol
Biotechnol 1976;3:125–33.
SPECIAL PATIENT POPULATIONS 2. Borel JF, Feurer C, Gubler HU, et al. Biological effects of cyclosporin
A: a new antilymphocytic agent. Agents Actions 1976;6:468–75.
In a large randomized multicenter trial involving pediat- 3. Borel JF, Feurer C, Magnée C, et al. Effects of the new anti-lymphocytic
ric renal transplant recipients (children and adolescents), peptide cyclosporin A in animals. Immunology 1977;32:1017–25.
the most common (3% of patients) adverse events associ- 4. Flechner SM, Kurian SM, Solez K, et al. De novo kidney transplanta-
tion without use of calcineurin inhibitors preserves renal structure
ated with tacrolimus-based primary immunosuppression and function at two years. Am J Transplant 2004;4:1776–85.
were hypertension, infections, hypomagnesemia, increased 5. Barker CF, Starzl TE. The arrival of cyclosporine. In: Hamilton D,
mean serum creatinine, diarrhea, PTDM, and tremor.156 editor. A history of organ tansplantation: ancient legends to mod-
Significantly more tacrolimus recipients experienced hypo- ern practice. Pittsburgh: University of Pittsburgh Press; 2012. p.
380–412.
magnesemia (P = 0.001) and diarrhea (P < 0.05) than 6. Calne RY, Rolles K, Thiru S, et al. Cyclosporin a initially as the only
cyclosporine recipients, and significantly fewer tacrolimus immunosuppressant in 34 recipients of cadaveric organs: 32 kid-
recipients experienced hypertrichosis (P = 0.005), flu syn- neys, 2 pancreases, and 2 livers. Lancet 1979;314:1033–6.
drome (P < 0.05), and gum hyperplasia (P < 0.05). 7. Calne RY, Rolles K, White DJG. Cyclosporin-A in clinical organ graft-
The risks of tacrolimus during pregnancy are similar to ing. Transplant Proc 1981;13:349–58.
8. Fung JJ. Tacrolimus and transplantation: a decade in review. Trans-
the risks associated with cyclosporine. Data from the US plantation 2004;77:S41.
National Transplantation Pregnancy Registry were used 9. Todo S, Fung J, Starzl T. Liver, kidney, and thoracic organ transplan-
to compare outcomes in 19 tacrolimus recipients (24 preg- tation under FK 506. Ann Surg 1990;212:295.
nancies) with outcomes in 56 cyclosporine microemulsion 10. Mayer AD, Dmitrewski J, Squifflet J-P, et al. Multicenter randomized
trial comparing tacrolimus (FK506) and cyclosporine in the preven-
recipients (71 pregnancies). Seventy-one percent of preg- tion of renal allograft rejection: a report of the European Tacrolimus
nancies resulted in live births in the tacrolimus group ver- Multicenter Renal Study Group. Transplantation 1997;64:436.
sus 80% of pregnancies in the cyclosporine microemulsion 11. Pirsch JD, Miller J, Deierhoi MH, et al. A comparison of tacrolimus
group; the mean gestational age was lower in the tacroli- (FK506) and cyclosporine for immunosuppression after cadaveric
mus group than in the cyclosporine group (32.9 vs. 35.8 renal transplantation. Transplantation 1997;63:977.
12. Jamieson NV. Adult small intestinal transplantation in Europe. Acta
weeks; P = 0.0035).157 There were no other statistically Gastroenterol Belg 1999;6:239.
significant differences in outcomes. 13. Kur F, Reichenspurner H, Meiser BM, et al. Tacrolimus (FK506) as
A single-center analysis was performed on 13 kidney primary immunosuppressant after lung transplantation. Thorac
transplant recipients and two SPK recipients who became Cardiovasc Surg 1999;47:174–8.
14. Meiser BM, Pfeiffer M, Schmidt D, et al. Combination therapy with
pregnant under tacrolimus-based immunosuppression.158 tacrolimus and mycophenolate mofetil following cardiac transplan-
The 13 mothers after kidney transplantation delivered tation: importance of mycophenolic acid therapeutic drug monitor-
19 infants, whereas the 2 mothers after SPK transplanta- ing. J Heart Lung Transplant 1999;18:143–9.
tion delivered 3 infants. All mothers survived the preg- 15. Rostaing L, Cantarovich D, Mourad G, et al. Corticosteroid-free
nancy, but one infant was stillborn. Forty-one percent of immunosuppression with tacrolimus, mycophenolate mofetil, and
daclizumab induction in renal transplantation. Transplantation.
the infants were either preterm or premature, and 27% of Transplantation 2005;79:807.
the infants were delivered by cesarean section. Toxemia of 16. Thompson J. Intestinal transplantation: experience in the United
pregnancy or preeclampsia was seen in 23% of these preg- States. Eur J Pediatr Surg 1999;9:271–3.
nancies. None of the mothers experienced rejection during 17. Matas AJ, Smith JM, Skeans MA, et al. OPTN/SRTR 2013 annual
data report: kidney. Am J Transplant 2015;15:1–34.
their pregnancy. 18. Plosker GL, Foster RH. Tacrolimus: a further update of its use in the
management of organ transplantation. Drugs 2000;59:323–89.
19. Christians U, Jacobsen W, Benet LZ, et al. Mechanisms of clinically
Conclusion relevant drug interactions associated with tacrolimus. Clin Pharma-
cokinet 2002;41:813–51.
20. Pallet N, Etienne I, Buchler M, et al. Long-term clinical impact of
The studies discussed in this chapter have confirmed the adaptation of initial tacrolimus dosing to CYP3A5 genotype. Am J
important role of CNIs, initially cyclosporine and then tacro- Transplant 2016;15.
limus, as the primary immunosuppressive agent in adult
17 • Calcineurin Inhibitors 257

21. Pallet N, Jannot AS, El Bahri M, et al. Kidney transplant recipients nolate mofetil versus cyclosporine (Neoral) with mycophenolate
carrying the CYP3A4*22 allelic variant have reduced tacrolimus mofetil after cadaveric kidney transplantation. Transplantation
clearance and often reach supratherapeutic tacrolimus concentra- 2000;69:834.
tions. Am J Transplant 2015;15:800–5. 47. Vacher-Coponat H, Moal V, Indreies M, et al. A randomized trial
22. Venkataramanan R, Swaminathan A, Prasad T, et al. Clinical phar- with steroids and antithymocyte globulins comparing cyclosporine/
macokinetics of tacrolimus. Clin Pharmacokinet 1995;29:404–30. azathioprine versus tacrolimus/mycophenolate mofetil (CATM2) in
23. Astellas Inc. Prograf prescribing information (US). https://www.aste renal transplantation. Transplantation 2012;93:437–43.
llas.us/docs/prograf.pdf. Accessed April 5, 2019. 48. Gonwa T, Mendez R, Yang HC, et al. Randomized trial of tacro-
24. Mancinelli LM, Frassetto L, Floren LC, et al. The pharmacokinetics limus in combination with sirolimus or mycophenolate mofetil
and metabolic disposition of tacrolimus: a comparison across ethnic in kidney transplantation: results at 6 months. Transplantation
groups. Clin Pharmacol Ther 2001;69:24–31. 2003;75:1213.
25. Scott LJ, McKeage K, Keam SJ, et al. Tacrolimus: a further update 49. Guerra G, Ciancio G, Gaynor JJ, et al. Randomized trial of immuno-
of its use in the management of organ transplantation. Drugs suppressive regimens in renal transplantation. J Am Soc Nephrol
2003;63:1247–97. 2011;22:1758–68.
26. Kulkarni S, Kopelan A. Tacrolimus therapy in renal transplantation. 50. Meier-Kriesche H-U, Schold JD, Srinivas TR, et al. Sirolimus in com-
In: Morris PJ, editor. Kidney transplantation: principles and practice. bination with tacrolimus is associated with worse renal allograft
Philadelphia: Saunders; 2001. p. 251–62. survival compared to mycophenolate mofetil combined with tacro-
27. Jordan ML, Shapiro R, Jensen CWB, et al. FK 506 conversion of renal limus. Am J Transplant 2005;5:2273–80.
allografts failing cyclosporine immunosuppression. Transplant Proc 51. Flechner SM, Glyda M, Cockfield S, et al. The ORION study: com-
1991;23:3078–81. parison of two sirolimus-based regimens versus tacrolimus and
28. Jordan ML, Naraghi R, Shapiro R, et al. Tacrolimus rescue therapy mycophenolate mofetil in renal allograft recipients. Am J Transplant
for renal allograft rejection-five-year experience. Transplantation 2011;11:1633–44.
1997;63:223–8. 52. Calconi G, Vianello A. One-year follow-up of a large European trial
29. Jordan ML, Naraghi RN, Shapiro R, et al. Five-year experience with comparing dual versus triple tacrolimus-based immunosuppres-
tacrolimus rescue for renal allograft rejection. Transplant Proc sive regimens following renal transplantation. Transplant Proc
1997;29:306. 2001;33:1021–4.
30. Dudley CRK. Conversion at first rejection: a prospective trial compar- 53. Chang RWS, Snowden S, Palmer A, et al. European randomised trial
ing cyclosporine microemulsion with tacrolimus in renal transplant of dual versus triple tacrolimus-based regimens for control of acute
recipients. Transplant Proc 2001;33:1034–5. rejection in renal allograft recipients. Transpl Int 2001;14:384–90.
31. Pohanka E, Margreiter R, Sparacino V. Switch to tacrolimus-based 54. Garcia I. Efficacy and safety of dual versus triple tacrolimus-based
therapy for cyclosporine-related side effects: a large, prospective therapy in kidney transplantation: two-year follow-up. Transplant
European study. Transplantation 2002;74(S):425 (abstract 2100). Proc 2002;34:1638–9.
32. Kohnle M, Zimmermann U, Lütkes P, et al. Conversion from cyclo- 55. Pascual J, Ortuño J. Simple tacrolimus-based immunosuppres-
sporine A to tacrolimus after kidney transplantation due to hyperlip- sive regimens following renal transplantation: a large multicenter
idemia. Transpl Int 2000;13:S345–8. comparison between double and triple therapy. Transplant Proc
33. Phelan D, Thompson C, Henschell J. Heart transplantation across pre- 2002;34:89–91.
formed class I antibody using FK506. Hum Immunol 1992;34:70. 56. Segoloni G, Bonomini V, Maresca MC, et al. Tacrolimus is highly
34. Woodle ES, Pedrizet G, Brunt E. FK506: inhibition of humoral mech- effective in both dual and triple therapy regimens following renal
anisms of hepatic allograft rejection. Transplantation 1992;54:377. transplantation. Spanish and Italian Tacrolimus Study Group.
35. Woodle ES, Pedrizet G, Brunt E. FK506: reversal of humorally-medi- Transpl Int 2000;13(Suppl. 1):S336–40.
ated rejection following ABO-incompatible liver transplantation. 57. Squifflet J-P, Bäckman L, Claesson K, et al. Dose optimization of
Transplant Proc 1991;23:2992. mycophenolate mofetil when administered with a low dose of tacro-
36. Walliser P, Benzie CR, Kay JE. Inhibition of murine B-lymphocyte limus in cadaveric renal transplant recipients. Transplantation
proliferation by the novel immunosuppressive drug FK-506. Immu- 2001;72:63.
nology 1989;68:434–5. 58. Ferraris JR, Tambutti ML, Cardoni RL, et al. Conversion from cyclo-
37. Wasik M, Stepień-Sopniewska B, Łagodziński Z, Gorski A. Effects of sporine A to tacrolimus in pediatric kidney transplant recipients
FK-506 and cyclosporine on human T and B lymphoproliferative with chronic rejection: changes in the immune responses. Trans-
responses. Immunopharmacology 1990;20:57–61. plantation 2004;77:532.
38. Wodle ES, Newell KA, Haas M. Reversal of accelerated renal allograft 59. Sandrini S, Aslam N, Tardanico R, et al. Tacrolimus versus cyclo-
rejection with FK506. Clin Transpl 1997;11:2251. sporine for early steroid withdrawal after renal transplantation.
39. Woodle ES, Spargo B, Ruebe M, et al. Treatment of acute glomerular J Nephrol 2012;25:43–9.
rejection with FK 506. Clin Transplant 1996;10:266–70. 60. Shapiro R, Jordan ML, Scantlebury VP, et al. A prospective, random-
40. Zachary AA, Montgomery RA, Ratner LE, et al. Specific and durable ized trial of tacrolimus/prednisone versus tacrolimus/prednisone/
elimination of antibody to donor HLA antigens in renal-transplant mycophenolate mofetil in renal transplant recipients. Transplanta-
patients. Transplantation 2003;76:1519. tion 1999;67:411.
41. Neylan JF. Racial differences in renal transplantation after immuno- 61. Kasiske BL, Zeier MG, Chapman JR, et al. KDIGO clinical practice
suppression with tacrolimus versus cyclosporine. Transplantation guideline for the care of kidney transplant recipients: a summary.
1998;65:515. Kidney Int 2010;77:299–311.
42. Vincenti F, Jensik SC, Filo RS, et al. A long-term comparison of 62. Margreiter R, Klempnauer J, Neuhaus P, et al. Alemtuzumab
tacrolimus (FK506) and cyclosporine in kidney transplantation: evi- (Campath-1H) and tacrolimus monotherapy after renal transplan-
dence for improved allograft survival at five years. Transplantation tation: results of a prospective randomized trial. Am J Transplant
2002;73:775. 2008;8:1480–5.
43. Jensik SC. Tacrolimus (FK 506) in kidney transplantation: three- 63. Chan K, Taube D, Roufosse C, et al. Kidney transplantation with
year survival results of the us multicenter, randomized, comparative minimized maintenance: alemtuzumab induction with tacrolimus
trial. Transplant Proc 1998;30:1216–8. monotherapy—an open label, randomized trial. Transplantation.
44. Mayer AD. Chronic rejection and graft half-life: five-year follow-up Transplantation 2011;92:774.
of the European tacrolimus multicenter renal study. Transplant Proc 64. Tan HP, Donaldson J, Basu A, et al. Two hundred living donor kid-
2002;34:1491–2. ney transplantations under alemtuzumab induction and tacrolimus
45. Waid T. Prograf as secondary intervention versus continuation of monotherapy: 3-year follow-up. Am J Transplant 2009;9:355–66.
cyclosporine in patients at risk for chronic renal allograft failure 65. Thomas PG, Woodside KJ, Lappin JA, et al. Alemtuzumab (Cam-
(CRAF) results in improved renal function, decreased CV risk, and path 1H) induction with tacrolimus monotherapy is safe for
no increased risk for diabetes. Am J Transplant 2003;3(Suppl.):436 high immunological risk renal transplantation. Transplantation
(abstract 1111). 2007;83:1509–12.
46. Johnson C, Ahsan N, Gonwa T, et al. Randomized trial of tacro- 66. Hanaway MJ, Woodle ES, Mulgaonkar S, et al. Alemtuzumab induc-
limus (Prograf) in combination with azathioprine or mychophe- tion in renal transplantation. N Engl J Med 2011;364:1909–19.
258 Kidney Transplantation: Principles and Practice

67. Farney AC, Doares W, Rogers J, et al. A randomized trial of alemtu- 88. Lim WH, Eris J, Kanellis J, et al. A systematic review of conversion
zumab versus antithymocyte globulin induction in renal and pan- from calcineurin inhibitor to mammalian target of rapamycin inhib-
creas transplantation. Transplantation 2009;88:810. itors for maintenance immunosuppression in kidney transplant
68. Donahoo WT, Kosmiski LA, Eckel RH. Drugs causing dyslipoprotein- recipients. Am J Transplant 2014;14:2106.
emia. Endocrinol Metab Clin North Am 1998;27:677–97. 89. Larson TS, Dean PG, Stegall MD, et al. Complete avoidance of calci-
69. Schwimmer J, Zand MS. Management of diabetes mellitus after solid neurin inhibitors in renal transplantation: a randomized trial com-
organ transplantation. Graft 2001;4:256–65. paring sirolimus and tacrolimus. Am J Transplant 2006;6:514–22.
70. van Duijnhoven EM, Christiaans MHL, Boots JMM, et al. Glucose 90. Meier-Kriesche H-U, Hricik DE. Are we ready to give up on calcineu-
metabolism in renal transplant recipients on tacrolimus: the effect of rin inhibitors? Am J Transplant 2006;6:445–6.
steroid withdrawal and tacrolimus trough level reduction. J Am Soc 91. Roodnat JI, Hilbrands LB, Hené RJ, et al. 15-year follow-up of a mul-
Nephrol 2002;13:221–7. ticenter, randomized, calcineurin inhibitor withdrawal study in kid-
71. Pirsch JD, Henning AK, First MR, et al. New-onset diabetes after ney transplantation. Transplantation 2014;98:47–53.
transplantation: results from a double-blind early corticosteroid 92. Yan HL, Zong HT, Cui YS, et al. Calcineurin inhibitor avoidance
withdrawal trial. Am J Transplant 2015;15:1982–90. and withdrawal for kidney transplantation: a systematic review
72. Woodle ES. A prospective, randomized, multicenter, double-blind and meta-analysis of randomized controlled trials. Transplant Proc
study of early corticosteroid cessation versus long-term mainte- 2014;46:1302–13.
nance of corticosteroid therapy with tacrolimus and mycopheno- 93. Moore J, Middleton L, Cockwell P, et al. Calcineurin inhibitor sparing
late mofetil in primary renal transplant recipients: one year report. with mycophenolate in kidney transplantation: a systematic review
Transplant Proc 2005;37:804–8. and meta-analysis. Transplantation 2009;87:591–605.
73. Woodle ES, First MR, Pirsch J, et al. A prospective, randomized, 94. Ekberg H, Tedesco-Silva H, Demirbas A, et al. Reduced exposure
double-blind, placebo-controlled multicenter trial comparing early to calcineurin inhibitors in renal transplantation. N Engl J Med
(7 day) corticosteroid cessation versus long-term, low-dose cortico- 2007;357:2562–75.
steroid therapy. Ann Surg 2008;248:564. 95. Ekberg H, Bernasconi C, Tedesco-Silva H, et al. Calcineurin inhibitor
74. Borrows R, Chan K, Loucaidou M, et al. Five years of steroid spar- minimization in the symphony study: observational results 3 years
ing in renal transplantation with tacrolimus and mycophenolate after transplantation. Am J Transplant 2009;9:1876–85.
mofetil. Transplantation 2006;81:125. 96. Qazi Y, Shaffer D, Kaplan B, et al. Efficacy and safety of everoli-
75. Rizzari MD, Suszynski TM, Gillingham KJ, et al. Ten-year outcome mus plus low-dose tacrolimus versus mycophenolate mofetil plus
after rapid discontinuation of prednisone in adult primary kidney standard-dose tacrolimus in de novo renal transplant recipients:
transplantation. Clin J Am Soc Nephrol 2012;7:494–503. 12-month data. Am J Transplant 2017;17:1358.
76. Suszynski TM, Gillingham KJ, Rizzari MD, et al. Prospective random- 97. Hricik DE, Formica RN, Nickerson P, et al. Adverse outcomes of
ized trial of maintenance immunosuppression with rapid discontinu- tacrolimus withdrawal in immune-quiescent kidney transplant
ation of prednisone in adult kidney transplantation. Am J Transplant recipients. J Am Soc Nephrol 2015;26:1–9.
2013;13:961–70. 98. Dugast E, Soulillou JP, Foucher Y, et al. Failure of calcineurin
77. Mujtaba MA, Taber TE, Goggins WC, et al. Early steroid with- inhibitor (tacrolimus) weaning randomized trial in long-term
drawal in repeat kidney transplantation. Clin J Am Soc Nephrol stable kidney transplant recipients. Am J Transplant 2016;16:
2011;6:404–11. 3255–61.
78. Shihab FS, Lee ST, Smith LD, et al. Effect of corticosteroid withdrawal 99. Silva HT, Yang HC, Meier-Kriesche H-U, et al. Long-term follow-up
on tacrolimus and mycophenolate mofetil exposure in a randomized of a phase III clinical trial comparing tacrolimus extended-release/
multicenter study. Am J Transplant 2013;13:474–84. MMF, tacrolimus/MMF, and cyclosporine/MMF in de novo kidney
79. Thomusch O, Wiesener M, Opgenoorth M, et al. Rabbit-ATG or transplant recipients. Transplantation 2014;97:636–41.
basiliximab induction for rapid steroid withdrawal after renal trans- 100. Albano L, Banas B, Klempnauer JL, et al. OSAKA trial: a randomized,
plantation (Harmony): an open-label, multicentre, randomised con- controlled trial comparing tacrolimus QD and BD in kidney trans-
trolled trial. Lancet 2016;388:3006–16. plantation. Transplantation 2013;96:897–903.
80. Kaufman DB, Leventhal JR, Axelrod D, et al. Alemtuzumab induc- 101. Budde K, Bunnapradist S, Grinyo JM, et al. Novel once-daily
tion and prednisone-free maintenance immunotherapy in kidney extended-release tacrolimus (LCPT) versus twice-daily tacrolimus
transplantation: comparison with basiliximab induction—long- in de novo kidney transplants: one-year results of phase III, double-
term results. Am J Transplant 2005;5:2539–48. blind, randomized trial. Am J Transplant 2014;14:2796–806.
81. Vítko S, Klinger M, Salmela K, et al. Two corticosteroid-free regi- 102. Bunnapradist S, Ciechanowski K, West-Thielke P, et al. Conversion
mens—tacrolimus monotherapy after basiliximab administration from twice-daily tacrolimus to once-daily extended release tacroli-
and tacrolimus/mycophenolate mofetil—in comparison with a mus (LCPT): the phase III randomized melt trial. Am J Transplant
standard triple regimen in renal transplantation: results of the Atlas 2013;13:760–9.
study. Transplantation 2005;80:1734. 103. Burke GWIII, Kaufman DB, Millis JM, et al. Prospective, randomized
82. Chhabra D, Skaro AI, Leventhal JR, et al. Long-term kidney allograft trial of the effect of antibody induction in simultaneous pancreas
function and survival in prednisone-free regimens: tacrolimus/ and kidney transplantation: three-year results. Transplantation
mycophenolate mofetil versus tacrolimus/sirolimus. Clin J Am Soc 2004;77:1269.
Nephrol 2012;7:504–12. 104. Busque S, Cantarovich M, Mulgaonkar S, et al. The Promise study:
83. Ahsan N, Hricik D, Matas AJ. Prednisone withdrawal in kid- a Phase 2b multicenter study of Voclosporin (ISA247) versus
ney transplant recipients on cyclosporine and mycopheno- tacrolimus in de novo kidney transplantation. Am J Transplant
late mofetil: a prospective randomized study. Transplantation 2011;11:2675–84.
1999;68:1865. 105. Kahan BD, Welsh M, Schoenberg L, et al. Variable oral absorption
84. Buell JF, Kulkarni S, Grewal HP, et al. Early corticosteroid cessa- of cyclosporine: a biopharmaceutical risk factor for chronic renal
tion at one week following kidney transplant under tacrolimus and allograft rejection. Transplantation 1996;62:599–606.
mycophenolate mofetil (MMF) immunosuppresion, three-year fol- 106. Kovarik J, Mueller E, Bree J van. Cyclosporine pharmacokinetics and
low-up. Transplantation 2000;69:S134 (abstract). variability from a microemulsion formulation: a multicenter investi-
85. Kumar MS, Irfan Saeed M, Ranganna K, et al. Comparison of four gation in kidney transplant patients. Transplantation 1994;58:663.
different immunosuppression protocols without long-term steroid 107. Kovarik JM, Mueller EA, Richard F, et al. Evidence for earlier stabili-
therapy in kidney recipients monitored by surveillance biopsy: five- zation of cyclosporine pharmacokinetics in de novo renal transplant
year outcomes. Transpl Immunol 2008;20:32–42. patients receiving a microemulsion formulation. Transplantation
86. de Fijter JW, Holdaas H, Øyen O, et al. Early conversion from cal- 1996;62:759–63.
cineurin inhibitor to everolimus-based therapy following kidney 108. Kaplan B, Schold JD, Meier-Kriesche HU. Long-term graft survival
transplantation: results of the randomized elevate trial. Am J Trans- with Neoral and tacrolimus: a paired kidney analysis. J Am Soc
plant 2017;17:1853. Nephrol 2003;14:2980–4.
87. Budde K, Lehner F, Sommerer C, et al. Five-year outcomes in kidney 109. Legendre C, Thervet E, Skhiri H, et al. Histologic features of chronic
transplant patients converted from cyclosporine to everolimus: the allograft nephropathy revealed by protocol biopsies in kidney trans-
randomized ZEUS study. Am J Transplant 2015;15:119. plant recipients. Transplantation 1998;65:1506.
17 • Calcineurin Inhibitors 259

110. Neu AM, Ho PL, Fine RN, et al. Tacrolimus vs. cyclosporine A as 132. Gruessner AC, Sutherland DER, Dunn DL, et al. Pancreas after kid-
primary immunosuppression in pediatric renal transplantation: a ney transplants in posturemic patients with type 1 diabetes mellitus.
NAPRTCS study. Pediatr Transplant 2003;7:217–22. J Am Soc Nephrol 2001;12:1–14.
111. Filler G, Webb NJA, Milford DV, et al. Four-year data after pediatric 133. Trimarchi HM, Truong LD, Brennan S, et al. FK506-associated
renal transplantation: a randomized trial of tacrolimus vs. cyclospo- thrombotic microangiopathy: report of two cases and review of the
rin microemulsion. Pediatr Transplant 2005;9:498–503. literature. Transplantation 1999;67:539.
112. Grenda R, Watson A, Vondrak K, et al. A prospective, random- 134. Klinger M, Vitko S, Karja S. Large prospective study evaluating
ized, multicenter trial of tacrolimus-based therapy with or without steroid-free immunosuppression with tacrolimus/basiliximab and
basiliximab in pediatric renal transplantation. Am J Transplant tacrolimus/MMF compared with tacrolimus/MMF/steroids in renal
2006;6:1666–72. transplantation. Nephrol Dial Transpl 2003;18:788.
113. Corporation Emmes. Collaborative Studies NAPRTCS 2014 Annual 135. Bartynski WS, Tan HP, Boardman JF, et al. Posterior reversible
Transplant Report. 2014. p. 102. encephalopathy syndrome after solid organ transplantation. Am J
114. Shapiro R, Scantlebury VP, Jordan ML, et al. Pediatric renal trans- Neuroradiol 2008;29:924.
plantation under tacrolimus-based immunosuppression. Transplan- 136. Shimizu T, Tanabe K, Tokumoto T, et al. Clinical and histological
tation 1999;67:299. analysis of acute tacrolimus (TAC) nephrotoxicity in renal allografts.
115. Webb NJA, Douglas SE, Rajai A, et al. Corticosteroid-free kidney Clin Transplant 1999;13(S):48–53.
transplantation improves growth: 2-year follow-up of the TWIST 137. Halloran PF, Ahsan N, Johnson CP. Three-year follow up of ran-
randomized controlled trial. Transplantation 2015;99:1178–85. domized multicenter kidney transplant study comparing tacrolimus
116. Filler G, Womiloju T, Feber J, et al. Adding sirolimus to tacrolimus- (TAC). Am J Transplant 2001;1:405.
based immunosuppression in pediatric renal transplant recipients 138. Artz MA, Boots JMM, Ligtenberg G, et al. Randomized conver-
reduces tacrolimus exposure. Am J Transplant 2005;5:2005–10. sion from cyclosporine to tacrolimus in renal transplant patients:
117. Gruessner RWG, Sutherland DER, Najarian JS, et al. Solitary pan- improved lipid profile and unchanged plasma homocysteine levels.
creas transplantation for nonuremic patients with labile insulin- Transplant Proc 2002;34:1793–4.
dependent diabetes mellitus. Transplantation 1997;64:1572. 139. Manu M, Tanabe K, Tokumoto T, et al. Impact of tacrolimus on
118. Sutherland DER, Gruessner RWG, Dunn DL, et al. Lessons learned hyperlipidemia after renal transplantation: a Japanese single center
from more than 1,000 pancreas transplants at a single institution. experience. Transplant Proc 2000;32:1736–8.
Ann Surg 2001;233:463. 140. Schnitzler MA, Lowell JA, Brennan DC. New-onset post-renal trans-
119. Burke GW, Ciancio G, Figueiro J, et al. Can acute rejection be pre- plant hyperlipidemia with cyclosporine compared to tacrolimus. In:
vented in SPK transplantation? Transplant Proc 2002;34:1913–4. 2nd International Congress on Immunosuppression. San Diego: ICI.
120. Ciancio G, Lo Monte A, Buscemi G, et al. Use of tacrolimus and myco- 141. Mourer JS, de Koning EJP, Van Zwet EW, et al. Impact of late cal-
phenolate mofetil as induction and maintenance in simultaneous cineurin inhibitor withdrawal on ambulatory blood pressure and
pancreas-kidney transplantation. Transpl Int 2000;13:S191–4. carotid intima media thickness in renal transplant recipients. Trans-
121. Kaufman DB, Leventhal JR, Gallon LG, et al. Technical and immu- plantation 2013;96:49–57.
nologic progress in simultaneous pancreas-kidney transplantation. 142. Holdaas H, de Fijter JW, Cruzado JM, et al. Cardiovascular param-
Surgery 2002;132:545–54. eters to 2 years after kidney transplantation following early
122. Ringers J, van der Torren CR, van de Linde P, et al. Pretransplanta- switch to everolimus without calcineurin inhibitor therapy: an
tion GAD-autoantibody status to guide prophylactic antibody induc- analysis of the randomized ELEVATE study. Transplantation
tion therapy in simultaneous pancreas and kidney transplantation. 2017;101:2612.
Transplant J 2013;96:745–52. 143. Coley KC, Verrico MM, McNamara DM, et al. Lack of tacrolimus-
123. Land W, Malaise J, Sandberg J, et al. Tacrolimus versus cyclosporine induced cardiomyopathy. Ann Pharmacother 2001;35:985–9.
in primary simultaneous pancreas-kidney transplantation: prelimi- 144. Montori VM, Basu A, Erwin PJ, et al. Posttransplantation diabetes.
nary results at 1 year of a large multicenter trial. Transplant Proc Diabetes Care 2002;25:583–92.
2002;34:1911–2. 145. Heisel O, Heisel R, Balshaw R, et al. New onset diabetes mellitus in
124. Bechstein WO, Malaise J, Saudek F, et al. Efficacy and safety of tacro- patients receiving calcineurin inhibitors: a systematic review and
limus compared with cyclosporine microemulsion in primary simul- meta-analysis. Am J Transplant 2004;4:583–95.
taneous pancreas-kidney transplantation: 1-year results of a large 146. Vincenti F, Friman S, Scheuermann E, et al. Results of an interna-
multicenter trial. Transplantation 2004;77:1221. tional, randomized trial comparing glucose metabolism disorders
125. Uemura T, Ramprasad V, Matsushima K, et al. Single dose of alemtu- and outcome with cyclosporine versus tacrolimus. Am J Transplant
zumab induction with steroid-free maintenance immunosuppression 2007;7:1506–14.
in pancreas transplantation. Transplantation 2011;92:678–85. 147. Hricik DE, Anton HAS, Knauss TC, et al. Outcomes of African Ameri-
126. Jordan ML, Chakrabarti P, Luke P, et al. Results of pancreas trans- can kidney transplant recipients treated with sirolimus, tacrolimus,
plantation after steroid withdrawal under tacrolimus immunosup- and corticosteroids. Transplantation 2002;74:189.
pression. Transplantation 2000;69:265. 148. Sato T, Inagaki A, Uchida K, et al. Diabetes mellitus after transplant:
127. Gallon LG, Winoto J, Chhabra D, et al. Long-term renal transplant relationship to pretransplant glucose metabolism and tacrolimus or
function in recipient of simultaneous kidney and pancreas trans- cyclosporine A-based therapy. Transplantation 2003;76:1320.
plant maintained with two prednisone-free maintenance immu- 149. van Duijnhoven EM, Christiaans MHL, Boots JMM, et al. Glucose
nosuppressive combinations: tacrolimus/mycophenolate mofetil metabolism in the first 3 years after renal transplantation in patients
versus tacrolimus/sirolimus. Transplantation 2007;83:10. receiving tacrolimus versus cyclosporine-based immunosuppres-
128. Gruessner RWG, Sutherland DER, Parr E, et al. A prospective, ran- sion. J Am Soc Nephrol 2002;13:213–20.
domized, open-label study of steroid withdrawal in pancreas trans- 150. Kasiske BL, Snyder JJ, Gilbertson D, et al. Diabetes mellitus after
plantation: a preliminary report with 6-month follow-up. Transplant kidney transplantation in the United States. Am J Transplant
Proc 2001;33:1663–4. 2003;3:178–85.
129. Kaufman DB, Leventhal JR, Koffron AJ, et al. A prospective study of 151. Baid-Agrawal S, Delmonico FL, Tolkoff-Rubin NE, et al. Car-
rapid corticosteroid elimination in simultaneous pancreas-kidney diovascular risk profile after conversion from cyclosporine a to
transplantation. Transplantation 2002;73:169. tacrolimus in stable renal transplant recipients. Transplantation
130. Freise C, Kang S, Feng S, et al. Excellent short-term results with ste- 2004;77:1199.
roid-free maintenance immunosuppression in low-risk simultaneous 152. Webster AC, Woodroffe RC, Taylor RS, et al. Tacrolimus versus
pancreas-kidney transplantation. Arch Surg 2003;138:1121–6. ciclosporin as primary immunosuppression for kidney transplant
131. Gruessner AC, Sutherland DER. Pancreas transplant outcomes for recipients: meta-analysis and meta-regression of randomised trial
United States (US) and non-US cases are reported to the United Net- data. BMJ 2005;331:810.
work for Organ Sharing (UNOS) and International Pancreas Trans- 153. Hecking M, Haidinger M, Döller D, et al. Early basal insulin therapy
plant Registry as of October 2002. In: Cecka J, Terasaki P, editors. decreases new-onset diabetes after renal transplantation. J Am Soc
Clinical transplants 2002. Los Angeles: UCLA Tissue Typing Labora- Nephrol 2012;23:739–49.
tory; 2003. p. 41.
260 Kidney Transplantation: Principles and Practice

154. Murakami N, Riella LV, Funakoshi T. Risk of metabolic complica- 157. Armenti VT, Corscia LA, McGrory CH. National Transplantation
tions in kidney transplantation after conversion to mTOR inhibi- Pregnancy Registry looks at outcomes with Neoral and tacrolimus.
tor: a systematic review and meta-analysis. Am J Transplant Nephrol News Issues 2000;14:S11.
2014;14:2317–27. 158. Jain AB, Shapiro R, Scantlebury VP, et al. Pregnancy after kidney
155. Dharnidharka VR, Ho P-L, Stablein DM, et al. Mycophenolate, tacro- and kidney-pancreas transplantation under tacrolimus: a single cen-
limus and post-transplant lymphoproliferative disorder: a report of ter’s experience. Transplantation 2004;77:897.
the North American Pediatric Renal Transplant Cooperative Study. 159. Vincenti F. A decade of progress in kidney transplantation. Trans-
Pediatr Transplant 2002;6:396–9. plantation 2004;77:S52.
156. Trompeter R, Filler G, Webb NJA, et al. Randomized trial of tacro-
limus versus cyclosporin microemulsion in renal transplantation.
Pediatr Nephrol 2002;17:141–9.
18 mTOR Inhibitors: Sirolimus
and Everolimus
CHRISTOPHER J.E. WATSON and MENNA R. CLATWORTHY

CHAPTER OUTLINE Discovery Metabolic Complications


Mechanism of Action Diabetes
Pharmacokinetics Lipids
Therapeutic Blood Monitoring Interstitial Lung Disease (Pneumonitis)
Pharmacogenetics Hemolytic-Uremic Syndrome
Drug Interactions Proteinuria
Use of mTOR Inhibitors Delayed Recovery From Ischemia-
De Novo Therapy With mTOR Inhibitors in Reperfusion Injury
the Absence of Calcineurin Inhibitors Peripheral Edema
De Novo Combination Therapy With mTOR Wound Healing and Lymphocele Formation
Inhibitors and Calcineurin Inhibitors Aphthous Ulcers
Maintenance Therapy With mTOR Inhibitors Rash
mTOR Inhibitors and Malignancy Anemia, Thrombocytopenia, and
mTOR Inhibitors as Antitumor Agents Leukopenia
mTOR Inhibitors and Posttransplant Gastrointestinal Symptoms
Malignancy Thrombosis
mTOR Inhibitors and Nonmelanoma Skin Renal Tubular Effects: Hypokalemia and
Cancer Hypophosphatemia
mTOR Inhibitors and Posttransplantation Bone Effects
Lymphoproliferative Disorder Liver Function Abnormalities
mTOR Inhibitors and Kaposi’s Sarcoma Amenorrhea and Testicular Function
mTOR Inhibitors and BK Virus mTOR inhibition in Pediatric Transplantation
mTOR Inhibitors in Polycystic Kidney Disease Summary and Conclusions
Safety and Side Effects of mTOR Inhibitors
Infection

Sirolimus and its closely related analog everolimus are Discovery


potent immunosuppressive agents that impair lymphocyte
activation and proliferation by inhibiting the mammalian Sirolimus (AY-22989, rapamycin, Rapamune) is a fermen-
target of rapamycin (mTOR), also known as the mechanis- tation product of the bacterium Streptomyces hygroscopicus,
tic target of rapamycin. The mTOR inhibitors emerged in first isolated from soil samples taken in 1965 from Easter
the 1990s as a new class of immunosuppressive agent and Island, which is known locally as Rapa Nui, hence the deci-
a promising alternative to calcineurin inhibitor (CNI)-based sion to subsequently name the drug rapamycin.1 It was first
therapy in organ transplantation, particularly because investigated as an antifungal agent in the mid-1970s2,3
mTOR inhibitors were not believed to cause nephrotoxicity, and in 1977 it was also reported to have immunosuppres-
hitherto the Achilles’ heel of CNIs. Their appeal was further sive effects in the rat, where it prevented the development
enhanced by the suggestion that they may have antitumor of experimental allergic encephalomyelitis and adjuvant
effects and inhibitory effects on vascular smooth muscle arthritis.4 Twelve years later it was reported that admin-
proliferation that might help prevent chronic rejection. istration of rapamycin prolonged the survival of heart
The early promise that mTOR inhibitors would replace CNIs allografts in rats and kidney allografts in pigs and dogs,
as the mainstay of immunosuppression after organ trans- although it was noted that some pigs developed interstitial
plantation has not been fulfilled. Their side effects and limita- pneumonitis,5 an observation that was later to be repeated
tions became increasingly apparent, and as a result their use in the clinic. More worryingly, rapamycin resulted in lethal
in organ transplantation has been more limited than initially vasculitis of the gastrointestinal tract in dogs, and this find-
anticipated. As new information from clinical trials of the ing delayed further clinical evaluation of sirolimus.5 Tacro-
mTOR inhibitors emerges it is clear they still have an impor- limus, which shares structural similarity with sirolimus
tant role and are an effective alternative to CNI-based immu- (Fig. 18.1), also causes vasculitis in the dog,6 but when
nosuppressive therapy in a number of clinical situations. used in humans in 1989 there was no sign of such toxicity,7
261
262 Kidney Transplantation: Principles and Practice

Tacrolimus
O Mechanism of Action
O
O
O The mTOR inhibitors, sirolimus and everolimus, exert their
OH principal immunosuppressive effects by inhibiting the abil-
O N ity of the intracytoplasmic mTORC1 multiprotein enzyme
complex to regulate the growth, proliferation, and survival
of lymphocytes and other immunocompetent cells. mTOR (a
O O 289kDa serine-threonine protein kinase) is a central com-
O ponent of two functionally distinct complexes called mTOR
O
complex 1 and 2 (mTORC1 and mTORC2; Fig. 18.2).9 Both
HO
mTOR complexes exert their effects on other intracellular
OH
signaling molecules including Akt and S6 kinase. Sirolimus
and everolimus are potent inhibitors of mTORC1, whereas
Sirolimus the mTORC2 complex is relatively resistant to these mTOR
FKBP-
inhibitors.
binding After entering into cells, the mTOR inhibitors bind to one
O domain of a family of immunophilins called FK506-binding proteins
O
H OH (FKBPs), particularly the 12-kD FKBP12 (see Fig. 18.2).
O O N Immunophilins are highly conserved and abundantly
FRB- expressed cytosolic proteins whose natural function in the
H
binding cell is the cis/trans isomerization of peptidyl-prolyl bonds
domain and is completely distinct from their ability to act as recep-
O O
H tors for certain immunosuppressant molecules and medi-
O OH
O ate immunosuppressive effects.10 The sirolimus-FKBP12
or everolimus-FKBP12 molecular complex binds to the
O OH FKBP12-binding domain of mTOR and blocks association
O of Raptor (rapamycin-sensitive adapter protein) with mTOR
and mLST8 and other proteins that collectively comprise
the functional mTORC1 complex within the cell.1,11 This
Everolimus
O
interferes with mTORC1-dependent intracellular signaling
O pathways, particularly those regulating cell growth and pro-
H OH
liferation in response to signals from cytokines, growth fac-
O O N tors, nutrients (especially amino acids), stress (e.g., hypoxia)
H and toll-like receptor (TLR) ligand engagement. In lymphoid
cells, the important signals originate from the cell surface
O O and are generated by cytokine receptor binding, such as
H
O
binding of interleukin-2 to the interleukin-2 receptor com-
O OH
plex, and ligand binding to coreceptors such as CD28.
The mTORC2 complex comprises mTOR, Rictor (rapamy-
O OH
O cin insensitive companion of mTOR), mitogen-activated
O
protein kinase associated protein (mSIN1), protein observed
Fig. 18.1 Structure of tacrolimus, sirolimus, and everolimus. Siroli- with Rictor (Protor/PRR5), and mLST8. Whereas mTORC2
mus and everolimus are macrocyclic lactones with structural similarity is resistant to acute inhibition by sirolimus (and everoli-
to tacrolimus (FK506, Prograf). Everolimus has a 2-hydroxyethyl chain mus), increasing evidence suggests that chronic exposure
substitution at position 40 of the sirolimus structure. All three mole-
cules have a common area that binds to a family of intracellular carrier to sirolimus may block mTORC2 function. mTORC2 is
proteins, the FK506-binding proteins (FKBPs), in particular the 12-kD involved in regulating cell morphology and maintaining
protein FKBP12 . FRB, the FKBP-rapamycin binding domain for mTOR. the integrity of the cytoskeleton. It is also an important reg-
ulator of intracellular signaling pathways in both B and T
cells, and mTORC2 inhibition may contribute to immuno-
suppressive effects of sirolimus and everolimus.12
an observation that resulted in a resumption in the clinical Functionally distinct T cell subsets exhibit differential sus-
evaluation of sirolimus. ceptibility to mTOR inhibition13 and, interestingly, mTOR
As the potential of sirolimus as a clinical immunosup- inhibitors appear to preferentially promote the expansion
pressive agent became apparent, chemists at Novartis syn- of T regulatory cells in humans,14,15 which makes them
thesized everolimus (RAD001, SDZRAD, Certican, Zortress, attractive agents to include in experimental protocols
Afinitor, Votubia) by making a 2-hydroxyethyl chain sub- aimed at promoting transplant tolerance.16 The extent to
stitution at position 40 of the sirolimus molecule (see Fig. which this property of mTOR inhibitors contributes to their
18.1), a drug that retained potent immunosuppressive clinical effectiveness as immunosuppressive agents remains
activity with improved oral bioavailability.8 Subsequently to be defined.
more derivatives have been synthesized with the aim of har- Whereas the principal immunosuppressive effect of
nessing its antitumor effects. mTOR inhibitors has been attributed to their inhibitory
18 • mTOR Inhibitors: Sirolimus and Everolimus 263

Immune signals
— antigen
Growth — cytokines
Nutrients factors — co-stimulation

Cell membrane

Sirolimus/
Everolimus
FKBP12

Raptor Deptor Rictor Peptor

mTOR mTOR Proctor

PRAS40 mLST8 mSin1 mLST8

mTORC1 mTORC2

Lipid mRNA
synthesis translation

Protein Cell cycle Glucose Cytoskeleton Cell


synthesis & growth metabolism integrity survival

Fig. 18.2 Highly simplified schematic representation of the mechanism of action of mammalian target of rapamycin (mTOR) inhibitors. The
mTOR inhibitors sirolimus and everolimus form an intracellular complex with FK506-binding protein 12 (FKBP12), and this complex inhibits the func-
tion of the mTORC1 complex, by preventing association of Raptor. mTORC1 is important for cell proliferation in response to growth factor stimulation
and regulates the S6K1 response to stimulation via the CD28 ligand in T cells, for example. mTOR also forms the mTORC2 complex, which is resistant to
sirolimus and everolimus and is involved in cytoskeleton control.

effect on T cell proliferation, with activated lymphocytes from the intestine although both have a relatively low and
showing cell cycle arrest in the late G1 phase, it is now clear variable bioavailability (around 25%). Both sirolimus and
that mTOR functions as a master regulator controlling everolimus have a narrow therapeutic index and therapeu-
many aspects of both innate and adaptive immunity.17 The tic monitoring of blood levels is necessary to ensure effec-
mechanisms of immunosuppression through mTOR inhi- tive and safe immunosuppression. Glutathione-everolimus,
bition are diverse, complex, and still not fully understood. an injectable prodrug of everolimus, has been developed,
For example, mTOR regulates T cell trafficking through largely on the back of everolimus in the oncology field.20
altered expression of cell surface molecules such as CCR7, After absorption, sirolimus is extensively bound to eryth-
and adhesion molecules such as VCAM1 on endothelial rocytes, and less than 5% of the drug remains free in the
cells.18 Therefore mTOR inhibition can alter lymphocyte plasma, where it is associated with the nonlipoprotein
migration patterns significantly. mTOR plays an important fraction.21 It has a long half-life of about 60 hours in renal
role in regulating the differentiation of functionally distinct transplant patients, with rapid absorption time to maximal
CD4+ T cell subsets and mTOR blockade appears as noted concentration at 1 to 2 hours and exposure that is propor-
previously to preferentially promote the development of T tional to dose, but with a large intersubject coefficient of
regulatory cells. mTOR also influences B cell development variance (CV; CV = 52%) and significant intrasubject vari-
and maturation, and inhibiting mTOR may interfere with B ability (CV = 26%).22,23 The pharmacokinetic profiles of the
cell responses. Inhibition of mTOR has been shown to have tablet and liquid formulations of sirolimus are similar apart
both immunosuppressive and immunostimulatory effects from a lower maximal concentration with tablets.24 With
on dendritic cells, reducing and increasing their ability both formulations, the total drug exposure (area under the
to stimulate T cells according to the type of dendritic cell. concentration-time curve [AUC]) correlates well with maxi-
Paradoxically, inhibiting mTOR signaling may also have mal and trough concentrations. Similar to cyclosporine and
immunostimulatory effects on T cells, and sirolimus may, tacrolimus, the pharmacokinetics of sirolimus differ in dif-
for example, promote the generation of long-lived memory ferent ethnic groups, with reduced oral bioavailability in
T cells.19 African Americans.25
Everolimus is more water-soluble than sirolimus, and
this increases its bioavailability. In studies of single doses of
Pharmacokinetics everolimus capsules in renal transplant recipients, evero-
limus was shown to have a much shorter half-life than
Sirolimus and everolimus are only available as oral formu- sirolimus (16–19 hours), a rapid absorption (maximal con-
lations, and both are marketed in tablet form; sirolimus is centration reached within 3 hours), and a good correlation
also available as an oral solution. They are rapidly absorbed between trough and AUC.26 As with sirolimus, ethnicity
264 Kidney Transplantation: Principles and Practice

affects everolimus pharmacokinetics, with a higher dose the concentration of mTOR inhibitors with a reciprocal
requirement in African American patients.27 There is a dif- increase in cyclosporine concentrations.37 This drug inter-
ference in the bioavailability of everolimus depending on action is particularly noticeable when the time interval
the size of tablet taken, with a higher Cmax when five 1 mg between mTOR inhibitor and cyclosporine ingestion varies.
tablets are taken compared with a single 5 mg tablet.28 It is thus important that patients receiving mTOR inhibitors
and cyclosporine adhere to a regular pattern of medica-
tion and do not vary the interval between taking the two
Therapeutic Blood Monitoring agents. Other data suggest that higher doses of everolimus
are required to achieve the same drug exposure when coad-
The low oral bioavailability of mTORs, effects of con- ministered with tacrolimus compared with cyclosporine.38
comitant food and other medicines, and variations in the Conversely, mTOR inhibitors reduce the exposure to tacro-
patient’s condition may cause significant inter- and intra- limus when the two drugs are coadministered.39,40 Other
patient variation in whole blood concentrations of mTORs. groups of drugs with important interactions with the CYP
The requirement to optimize exposure to the drugs to main- pathway are antimicrobials (especially fluconazole and
tain adequate immunosuppressive efficacy while mini- erythromycin) and 3-hydroxy-3-methylglutaryl-coenzyme
mizing side effects mandates monitoring of whole blood A reductase (HMGCoA) inhibitors (statins), both of which
concentrations of the drug,29 and militates against fixed- are widely used in renal transplant recipients.
dose regimens. Whole blood measurements of drug concen- mTOR inhibitors also differ from cyclosporine in the
tration are performed because of the high degree of binding way they interact with other immunosuppressive agents.
of the drug to erythrocytes, although it is the very small free Patients taking sirolimus, like tacrolimus, have a much
drug fraction that is responsible for immunosuppression. higher exposure to mycophenolic acid than do patients tak-
There are two principal methods used in the determination ing mycophenolate and cyclosporine,41 and for patients
of mTOR concentrations, enzyme-linked immunoassays switching from a CNI to mTOR inhibitor the dose of myco-
and high-performance liquid chromatography (HPLC) with phenolic acid may need to be adjusted. Administration of
ultraviolet or mass spectrometry detection. HPLC mea- mTOR inhibitors may also result in a modest reduction in
sures the parent drug; it is very accurate but time consum- exposure (reduced AUC) to prednisolone compared with
ing. Immunoassays, on the other hand, use microparticles cyclosporine.42
coated with antibodies to the mTORs and provide a rapid
assay, but one which overestimates the drug concentration
because of cross-reaction with drug metabolites in addition Use of mTOR Inhibitors
to the parent drug.30,31 Even with the same assay there may
be variations in measured concentrations between labora- mTOR inhibitors have been evaluated for use in renal trans-
tories and over time.32 plantation as an addition to CNI-based therapy and as a
substitute for CNIs. mTOR inhibitors have also been used
as de novo treatment from the time of renal transplanta-
Pharmacogenetics tion, as a later addition to CNIs to enhance immunosup-
pression in response to acute rejection, and as a substitute
Sirolimus and everolimus are metabolized in the liver and for CNIs to avoid CNI toxicity in the maintenance phase of
intestinal wall by the cytochrome P450 (CYP) 3A enzyme immunosuppression.
subfamily (CYP3A4 and CYP3A5) and to a minor extent Early in vitro and in vivo studies suggested that mTOR
by CYP2C8. Polymorphisms of these enzymes are com- inhibitors and CNIs when used together had a synergis-
mon and because of linkage disequilibrium (the genes lie tic immunosuppressive effect.43,44 This might be antici-
adjacent on chromosome 7q21) may occur together. Poly- pated given that CNIs and mTOR inhibitors each block
morphisms of CYP3A enzymes are associated with lower different signals during T cell activation and affect differ-
drug concentration-to-dose ratios in patients expressing ent stages of the cell cycle. Initially, it was envisaged that
the least common genotypes.33,34 An association between mTOR inhibitors might be best used along with CNIs to
CYP3A5 genotype and dose requirement for sirolimus has exploit this synergistic immunosuppressive effect, optimiz-
been noted in some but not all studies, and a recent study ing immunosuppression and minimizing agent-specific side
in renal transplant patients found no association between effects. Evidence from rodent studies suggested, however,
CYP3A5 genotype and everolimus pharmacokinetics.35 It that sirolimus may exacerbate cyclosporine nephrotoxic-
is likely that CYP3A5 contributes much less to sirolimus ity,45 a finding that was subsequently confirmed in clinical
metabolism than CYP3A4, and that many of the studies of studies.46 Most of the initial work with sirolimus was done
the CYP3A5 interaction have been underpowered.36 in conjunction with cyclosporine rather than tacrolimus
because it was believed that competition for binding to the
immunophilin FKBP12 would preclude coadministration of
Drug Interactions tacrolimus and sirolimus. It later became evident that there
is an abundance of FKBP12 in the cytoplasm, and in vitro
Because sirolimus and everolimus are metabolized pri- studies suggest that less than 5% of the available FKBP
marily by CYP3A4, and to a lesser extent by CYP3A5 and needs to be bound to cause half-maximal immunosup-
CYP 2C8, drugs that affect these enzyme pathways alter pression.47 Tacrolimus and sirolimus can be administered
the metabolism of the mTORs. Important among these are simultaneously at therapeutic doses in humans without
the CNIs, particularly cyclosporine, which can increase significant competition for FKBP12.48
18 • mTOR Inhibitors: Sirolimus and Everolimus 265

DE NOVO THERAPY WITH mTOR INHIBITORS IN insight into the toxicity profile of sirolimus in humans and
THE ABSENCE OF CALCINEURIN INHIBITORS suggested a side effect profile that was different from that
associated with CNIs (Table 18.1).
Sirolimus has been investigated in numerous studies where Subsequent studies further explored the use of sirolimus
it was the principal immunosuppressant. The first such with MMF, together with anti-CD25 monoclonal antibody
studies were phase 2 trials conducted in Europe that exam- induction therapy. Early data suggested that the combination
ined concentration-controlled sirolimus dose regimens, of sirolimus and MMF was superior to a cyclosporine-based
rather than fixed-dose regimens. When sirolimus was regimen.52 A subsequent randomized trial comparing siroli-
administered as a component of triple therapy with aza- mus and tacrolimus (each given along with MMF and pred-
thioprine and prednisolone, it was associated with a simi- nisolone) showed the two regimens to be comparable in terms
lar incidence of acute rejection to that observed in patients of acute rejection rate and graft function.53 A registry analysis
on the old nonmicroemulsion Sandimmune preparation of suggested that renal allograft recipients treated with a combi-
cyclosporine (41% vs. 38% at 12 months).49 A follow-up nation of sirolimus and MMF had a higher rate of acute rejec-
study substituted azathioprine with mycophenolate mofetil tion and reduced allograft survival compared with recipients
(MMF) and showed no significant difference in the inci- receiving alternative immunosuppressive regimens.54
dence of acute rejection between sirolimus and cyclospo- A systematic review of randomized trials in which mTOR
rine, although there were numerically more acute rejection inhibitors were used in place of CNIs as initial therapy
episodes in the sirolimus arm (27.5% vs. 18.5%).50 Patient after kidney transplantation in studies published up to
and graft survival were similar in the two study groups, 2005 (eight different trials with a total of 750 participants)
although the studies were insufficiently powered to detect revealed that there was no difference in the incidence of
small differences. Pooled data from both studies showed acute rejection at 1 year, but the level of serum creatinine
significantly better renal function in patients receiving siro- (a possible surrogate endpoint for long-term graft survival)
limus.51 These two early studies provided the first detailed was lower in patients receiving mTOR inhibitors.55

TABLE 18.1 Adverse Effects of Sirolimus Identified in Phase 2 Studies of Sirolimus Compared With Cyclosporine
Sirolimus Cyclosporine + Azathioprine Cyclosporine + MMF
(n = 41 + 40) (n = 42) (n = 38)
METABOLIC
Hypertriglyceridemia 21 + 29 = 50 (63%) 5 (12%) 19 (50%)
Hypercholesterolemia 18 + 26 = 44 (54%) 6 (14%) 17 (45)
Hyperglycemia 8 + 6 = 14 (17%) 3 (7%) 6 (16%)
IDDM 1 + 1 = 2 (2%) 1 (2%) 1 (3%)
ALT increase 8 + 8 = 16 (20%) 1 (2%) 3 (8%)
Hypokalemia 14 + 8 = 22 (27%) 0 6 (16%)
Hypophosphatemia 6 + 6 = 12 (15%) 0 1 (3%)
Hyperuricemia 1 (3%) — 7 (18%)
HEMATOLOGIC
Thrombocytopenia 15 + 18 = 33 (41%) 0 3 (8%)
Leukopenia 16 + 11 = 27 (33%) 6 (14%) 7 (18%)
Anemia 15 + 17 = 32 (40%) 10 (24%) 11 (29%)
INFECTIONS
CMV viremia 6 + 2 = 8 (10%) 5 (12%) 8 (21%)
Herpes simplex 10 + 6 = 16 (20%) 4 (10%) 6 (16%)
Herpes zoster 0 + 1 = 1 (1%) 1 (2%) 1 (3%)
Oral Candida 3 + 5 = 8 (10%) 0 3 (8%)
PCP 0+0 1 (2%) 0
Pyelonephritis/UTI 17 + 17 = 34 (42%) 12 (29%) 15 (39%)
Septicemia 6 + 2 = 8 (10%) 1 (2%) 1 (3%)
Pneumonia 7 + 6 = 13 (16%) 1 (2%) 2 (5%)
Wound infection 4 + 2 = 6 (7%) 2 (5%) 3 (8%)
OTHER
Hypertension 7 + 16 = 23 (16%) 14 (33%) 18 (47%)
Arthralgia 8 (20%) 0 —
Tremor 1 + 2 = 3 (4%) 7 (14%) 8 (21%)
Gingival hyperplasia 0+0 4 (10%) 3 (8%)
Hirsutism 1 (3%) — 4 (11%)
Diarrhea 15 (38%) — 4 (11%)
Malignancies 0 2 (5%) 0

ALT, alanine aminotransferase; CMV, cytomegalovirus; IDDM, insulin-dependent diabetes mellitus; MMF, mycophenolate mofetil; PCP, Pneumocystis jirovecii
pneumonia; UTI, urinary tract infection.
Data from Groth CG, Backman L, Morales JM, et al. Sirolimus (rapamycin)-based therapy in human renal transplantation: similar efficacy and different toxic-
ity compared with cyclosporine. Sirolimus European Renal Transplant Study Group. Transplantation 1999;67:1036; and Kreis H, Cisterne JM, Land W, et al.
Sirolimus in association with mycophenolate mofetil induction for the prevention of acute graft rejection in renal allograft recipients. Trans p lantation
2000;69:1252.
266 Kidney Transplantation: Principles and Practice

Reports from two large-scale trials (ORION and SYM- and standard dose cyclosporine groups (91.7% and 91.9%,
PHONY) suggested that the combination of sirolimus and respectively) compared with the low-dose tacrolimus group
MMF is inferior to a low-dose tacrolimus and MMF-based (96.4%). Withdrawal from the study, mainly because of
triple therapy. treatment failure (use of additional immunosuppressive
The ORION study was an open-label randomized mul- agents and discontinuation of the study drug), was high-
ticenter international comparison of two sirolimus-based est in recipients randomized to low-dose sirolimus (48.9%).
regimens with tacrolimus and MMF in adult first- or second- Serious adverse events occurred in 53.2% of patients in the
time recipients of living or deceased donor kidneys.56 The low-dose sirolimus group compared with around 44% for
study recruited 469 patients in 65 centers in North Amer- the other study groups. In this study, therefore, low-dose
ica, Europe, and Australia and randomly assigned them to sirolimus resulted in higher rates of biopsy-proven rejection
one of three groups. Group one received sirolimus (initial and no improvement in renal function compared with the
target trough levels 8–15 ng/mL) plus tacrolimus (target cyclosporine-based regimens.
trough level 6–15 ng/mL), with stepwise withdrawal of In a subsequent report on this study, with an additional
tacrolimus after week 13 and continuation on sirolimus 2 years of follow-up based on around half of the patients
(maintenance trough levels of 12–20 ng/mL). Group two originally recruited to the (ELiTE)-Symphony study,59 renal
received sirolimus (initial target trough levels of 10–15 ng/ function remained relatively stable overall and the inci-
mL, reducing after 13 weeks to 8–15 ng/mL and after 26 dence of graft rejection and graft loss remained low. The
weeks to 5–15 ng/mL) plus MMF (up to 2 g/day). Group low-dose tacrolimus group continued to have the highest
three received tacrolimus (target trough levels of 8–15 ng/ GFR but the difference compared with the other groups was
mL to week 26 and 5–15 ng/mL thereafter) and MMF (up to no longer significant. However, the study was not powered
2 g/day). The primary endpoint of the study was glomeru- to detect differences in the key endpoints at 3 years. The
lar filtration rate (GFR) at 12 months (using the Nankivell57 number of patients in the low-dose sirolimus group who
formula) and secondary endpoints included GFR at 2 years, remained on sirolimus for the entire 3-year study period
patient and graft survival, and biopsy-­ confirmed acute was low, although their average renal function at 3 years
rejection. Study group two (Sirolimus + MMF) was pre- was numerically but not significantly slightly higher than
maturely discontinued because of a higher than expected that of patients remaining on the original agents in the
rate of acute rejection. No significant differences in the pri- other three groups.
mary endpoint were found. Patient survival was similar in Eight-year follow-up of the 145 patients randomized in
all three groups (2-year patient survival 94.4% in group the SPIESSER study, comparing de novo cyclosporine with
one, 94.5% in group two, and 97% in group three). Graft de novo sirolimus, together with MMF and steroids, showed
loss was numerically higher in the groups receiving siroli- similar results to ELiTe.60 There were more discontinuations
mus but not significantly so (2-year graft survival 88.5% of sirolimus than cyclosporine (47% vs. 27%) and more
in group one, 89.9% in group two, and 95.4% in group adverse events in the sirolimus group, but no difference in
three). The incidence of biopsy-proven acute rejection in patient or graft survival, or acute rejection incidence; inten-
the first 2 years was significantly higher in the CNI-free tion-to-treat analysis showed that the sirolimus arm had
groups (group one 17.4%, group two 32.8%, and group better renal function (modification of diet in renal disease
three 12.3%). Adverse events were the primary reason for study [MDRD] GFR 62.5 mL/min sirolimus [SRL], 47.8 mL/
patients not continuing in the study, and this occurred in min cyclosporine [CsA], P = 0.004).
34.2% patients in group one, 33.6% patients in group two,
and 22.3% patients in group three. The authors concluded DE NOVO COMBINATION THERAPY WITH mTOR
that sirolimus-based regimens were not associated with INHIBITORS AND CALCINEURIN INHIBITORS
improved outcomes after kidney transplantation.
The Efficacy Limiting Toxicity Elimination (ELiTE)-Sym- One of the first studies of sirolimus in renal transplantation
phony study,58 was an open label multicenter international to be performed was a dose-ranging study that combined
randomized four-arm trial that examined the efficacy of different fixed doses of sirolimus in conjunction with a high-
reduced dose sirolimus, tacrolimus, or cyclosporine when dose or low-dose Sandimmune cyclosporine (concentration
combined with CD25 monoclonal antibody induction controlled).26 All groups received steroids but no azathio-
(daclizumab), MMF, and corticosteroids in adult renal prine or MMF. Small numbers of patients in the study and an
transplant recipients. Both living donor and deceased donor unequal distribution of African Americans between the six
transplants were included. The outcomes were also com- study groups meant that the results were difficult to inter-
pared with those in recipients given standard dose cyclospo- pret. Nevertheless, the study showed that the combination
rine, MMF, and corticosteroids. The primary endpoint was of sirolimus and cyclosporine was more potent than cyclo-
estimated GFR 12 months after transplantation. The study sporine alone in the prevention of acute rejection and that
recruited 1645 patients from 15 participating countries. half-dose cyclosporine and sirolimus was as efficacious as
The study group that was given low-dose tacrolimus, MMF, full-dose cyclosporine and sirolimus. The higher incidence
and steroids had a significantly better 12-month GFR com- of acute rejection seen in African Americans in this study
pared with the other three study groups. Acute rejection was also observed in subsequent studies.61 The other impor-
(biopsy-proven) in the first 6 months was three times higher tant finding to emerge from this study was a high incidence
in the low-dose sirolimus group (35.3%) compared with the of pneumocystis pneumonia in sirolimus-treated patients,
low-dose tacrolimus group (11.3%). Allograft survival at particularly where routine prophylaxis against Pneumo-
12 months (censored for death with a functioning trans- cystis jirovecii was not given. Two large phase 3 studies of
plant) was also significantly lower in the low-dose sirolimus sirolimus followed shortly afterward, one conducted in the
18 • mTOR Inhibitors: Sirolimus and Everolimus 267

TABLE 18.2 Outcome of Two Phase 3 Sirolimus Adjuvant Therapy Studies


US Study (n = 719) Global Study (n = 576)
Aza SRL 2 mg SRL 5 mg Placebo SRL 2 mg SRL 5 mg
(n = 161) (n = 284) (n = 274) (n = 130) (n = 227) (n = 219)
Acute rejection (%) 29.8 16.9a 12b 41.5 24.7c 19.2d
Creatinine clear- 68.8 62.3 59.2 62.6 56.4
ance (mL/min)
Graft survival (%) 94.4 94.3 92.7 87.7 89.9 90.9
Patient survival (%) 98.1 97.2 96 94.6 96.5 95

Aza, azathioprine; SRL, sirolimus.


Note: Acute rejection incidence and creatinine clearance (Nankivell formula) are 6-month values; graft and patient survivals are 12-month values.
aP = 0.002 relative to the azathioprine arm.
bP < 0.001 relative to the azathioprine arm.
cP = 0.003 relative to the placebo arm.
dP < 0.001 relative to the placebo arm.

From MacDonald A, Rapamune Global Study Group. A worldwide, phase 3, randomized, controlled, safety and efficacy study of a sirolimus/cyclosporine regimen
for prevention of acute rejection in recipients of primary mismatched renal allografts. Transplantation 2001;71:271.

US61 and the second worldwide (Table 18.2).62 Similar early multicenter clinical trial conducted predominantly in
to earlier studies, these studies used a fixed dose of siroli- Europe compared two fixed doses of everolimus (1.5 and 3
mus (2 or 5 mg/day) in combination with concentration- mg/day) against MMF, with all three arms receiving corti-
controlled cyclosporine. In the US study, the two different costeroids and full dose cyclosporine with target trough lev-
doses of sirolimus were compared with azathioprine, and all els of 150 to 400 ng/mL until week 4 and then 100 to 300
groups received steroids but no induction therapy.61 Only ng/mL.64,65 There was no significant difference between
patients with functioning renal allografts were recruited, in the three groups in the primary composite endpoint at 6
contrast to the global study in which function of the graft months (biopsy-proven acute rejection, graft loss, death,
was not a prerequisite for enrolment.62 The other major dif- and loss to follow-up). Significantly worse renal function
ference between the US and the global study was that the in both everolimus groups led to a protocol revision at 12
comparator in the global study was placebo rather than months with reduction in the target trough cyclosporine
azathioprine. Both studies showed a clear benefit in terms levels to 50–75 ng/mL. At 3 years, renal function was simi-
of reduction in the rate of acute rejection for patients receiv- lar in the low-dose everolimus and MMF groups, but signifi-
ing sirolimus, an effect that was more marked in patients cantly worse in the higher dose everolimus group. A similar
receiving a higher dose of sirolimus. There was a difference trial conducted predominantly in North America with the
in acute rejection rates, patient survival, and graft survival same composite endpoint at 36 months and the same proto-
in the US study compared with the global study in all treat- col amendment showed that all three groups were similarly
ment arms, which likely reflects the different enrollment efficacious but creatinine clearance was lower at 12 and 36
requirements, with only recipients with functioning grafts months in the everolimus groups.66 Another multicenter
being entered into the US study. randomized phase 2 study undertaken in the US and Europe
These two pivotal studies in the development of siroli- evaluated the efficacy and safety of everolimus 3 mg/day
mus reveal much about how best to use sirolimus and its together with basiliximab induction, prednisolone, and
drawbacks. There was a high incidence of lymphocele for- either full dose or reduced dose cyclosporine (trough levels
mation (12%–15% vs. 3% in the azathioprine control group 125–250 ng/mL, respectively) although a protocol amend-
in the US study) and wound infection compared with the ment at 12 months reduced the cyclosporine trough target
control arm. Of particular importance was the observation to 50–75 ng/mL in all patients.67 The primary endpoint
that the renal function of patients on a combination of siro- was efficacy (composite of biopsy-proven acute rejection,
limus and cyclosporine was worse than that of patients on graft loss, death, or loss to follow-up) and efficacy failure
cyclosporine alone, with a 12-month calculated creatinine was significantly higher in the full dose cyclosporine arm
clearance of 67.5 mL/min in the azathioprine control group at 36 months (35.8% vs. 17.2%). Creatinine clearance was
compared with 62 mL/min and 55.5 mL/min in the 2 mg similar in the two groups at 36 months.
and 5 mg sirolimus groups. A similar effect was seen in the Sirolimus has also been evaluated in combination with
global study. tacrolimus after an initial report suggesting that the theo-
The immunosuppressive synergy between cyclosporine retical misgivings about the combination are not seen in
and sirolimus in these studies was analyzed by median clinical practice.68 As noted previously, the ORION study56
effect analysis of the pooled data.46 This analysis showed included a group that received sirolimus plus tacrolimus
that administration of sirolimus permitted a 2.2-fold reduc- but with stepwise withdrawal of tacrolimus after week 13
tion in cyclosporine exposure, and reciprocally, cyclospo- and continuation on sirolimus.
rine permits a 5-fold reduction in sirolimus dose to achieve Sirolimus in combination with tacrolimus was compared
the same immunosuppressive efficacy. Experimental data with tacrolimus and MMF as primary immunosuppression
also suggest that synergism accounts for the increased after kidney transplantation in a randomized multicenter
nephrotoxicity.63 trial in the US.69 A total of 361 patients were recruited. Both
The combination of everolimus and cyclosporine has arms of the study received corticosteroids and sirolimus
been evaluated in several multicenter clinical trials. An was dosed to achieve blood levels of 4 to 12 ng/mL. At 1
268 Kidney Transplantation: Principles and Practice

year renal function was superior in the tacrolimus arm and graft loss (1.3% EVL + LTac, 3.9% Tac + MMF, P < 0.05)
patients receiving sirolimus had a higher incidence of study was less with the everolimus plus low-dose tacrolimus com-
drug discontinuation (26.5% vs. 14.8%). The inferiority of bination. There was no difference in MDRD GFR at 1 year.74
sirolimus/tacrolimus in terms of renal function observed in In conclusion, there is no convincing evidence that use
this study was not found in a more recent European mul- of mTOR inhibitors as de novo therapy offers any advan-
ticenter randomized phase 2 clinical trial involving 734 tage over CNI-based therapy. The 2009 KDIGO clinical
patients.70 In this study primary immunosuppression using practice guidelines for the care of kidney transplant recipi-
sirolimus and tacrolimus was compared with tacrolimus and ents recommend that the combined use of mTOR inhibi-
MMF. Sirolimus was given according to a fixed-dose regimen tors and CNIs should be avoided because they potentiate
of 2 mg for 28 days and 1 mg thereafter. Both study groups nephrotoxicity, particularly if used in the early period after
received steroids initially but these were tapered and discon- transplantation.75
tinued after day 90. Renal function at 6 months, the primary
endpoint, was similarly good in the two study groups. Acute MAINTENANCE THERAPY WITH mTOR
rejection and graft survival were similar in both groups, but INHIBITORS
premature withdrawal because of adverse events was twice
as high in the sirolimus/tacrolimus arm (15.1% vs. 6.3%). Although available data suggest that mTOR inhibitors are
Although not comparing de novo combination of siro- almost as efficacious in terms of immunosuppressive potency
limus and tacrolimus, in the ADHERE study patients were as CNIs when used as the principal immunosuppressive
given prolonged-release tacrolimus and converted from agent, their use immediately after renal transplantation is
MMF to sirolimus with low-dose tacrolimus early post- undesirable because of their effects on wound healing and
transplant, on day 28.71 Of 730 patients randomized, 625 lymphocele formation. Because mTOR inhibitors used in
patients completed the study 11 months later. There was the absence of CNIs do not have the same nephrotoxicity
no difference in the primary endpoint of iohexol-measured concerns as CNIs they are potentially attractive agents for
GFR at 12 months (40.73 MMF vs. 41.75 mL/min/1.73m2 use in the maintenance phase of the posttransplant course,
SRL, P = 0.405), but there were more patients withdrawing especially in patients with CNI-associated problems, includ-
from the sirolimus arm because of adverse events than the ing chronic allograft nephropathy. The first major study to
MMF arm (14.4% vs. 5.2%). examine the efficacy of mTOR inhibitors in this context used
Everolimus plus tacrolimus has also been evaluated for sirolimus combined with cyclosporine and steroids as initial
use as primary immunosuppression after renal transplan- therapy, with the cyclosporine being stopped at 3 months
tation. The first prospective study to evaluate this combi- in half the patients. Sirolimus was shown to provide suffi-
nation was a 6-month multicenter study performed in the cient immunosuppression during the maintenance phase,
US. Ninety-two de novo renal transplant recipients were with superior calculated creatinine clearance compared
randomized to receive everolimus, steroids, and basiliximab with patients remaining on sirolimus and cyclosporine.
together with either standard or low-exposure tacrolimus.72 Although the acute rejection rate was slightly higher in the
The differences in tacrolimus exposure achieved in the two no-cyclosporine group, this did not translate into poorer
study arms were relatively small, but the study suggested renal function.76 Longer follow-up confirmed the sustained
that everolimus/tacrolimus-based immunosuppression was benefit of sirolimus maintenance therapy.77,78 This study
safe and effective and gave good renal function at 6 months. had no standard control group, and the subsequent finding
In a global study performed in 13 countries (ASSET) of enhanced nephrotoxicity when CNIs are combined with
everolimus was given along with tacrolimus (target lev- sirolimus casts a shadow over the results.61,62
els of 4–7 ng/mL) for the first 3 months in all patients and Smaller studies also suggested a benefit of sirolimus over
thereafter the two study arms comprised one maintained at CNIs as maintenance therapy after renal transplantation.
the same target level of tacrolimus and a tacrolimus mini- A dual-center randomized controlled trial suggested that
mization group with target tacrolimus levels of 1.5 to 3.0 conversion to sirolimus in patients with impaired graft
ng/mL.73 The study failed to demonstrate any benefit in the function results in a rapid improvement in measured glo-
tacrolimus minimization group, possibly as a result of over- merular filtration rate at 3 months, which was sustained
lapping of achieved tacrolimus exposure in the two groups, to 2 years, whereas patients who remained on CNIs expe-
but suggested that everolimus in combination with early rienced deteriorating graft function.79 Because of concerns
tacrolimus minimization was associated with a low rejec- about triggering acute rejection during the conversion from
tion rate, good graft survival and renal function, and an CNIs to sirolimus, some investigators have used a period of
acceptable safety profile. overlap of immunosuppression,80 or covered the transition
The lower rejection rate in ASSET is in contrast to the period with additional agents such as basiliximab,81 but in
findings of a more recent noninferiority study, where patients who are greater than 6 months posttransplanta-
everolimus (target 3–8 ng/mL) and low-dose tacrolimus tion, it is unlikely that this is necessary.
(4–7 ng/mL) were compared with MMF and standard dose Late conversion to sirolimus is associated with three dom-
tacrolimus (8–12 ng/mL) in 613 recipients of deceased inant side effects that might limit its usefulness as a main-
donor kidney transplants, including extended criteria and tenance agent, in addition to the other side effects that are
circulatory death donor kidneys.74 This study showed infe- well recognized with sirolimus (see later). First, more than
riority in the composite endpoint (biopsy-proven rejection, half of patients in some studies experience a rash, either an
graft loss, and death) of the everolimus/tacrolimus arm; acneiform rash or a dermatitis-like rash affecting the hands
The incidence of biopsy-proven acute rejection was greater and, in particular, the fingers. Second, the period of con-
(19.1% EVL + LTac, 11.2% Tac + MMF, P < 0.05) although version to sirolimus is associated with the development of
18 • mTOR Inhibitors: Sirolimus and Everolimus 269

mouth ulcers that, in most patients, resolve within 4 weeks. mycophenolate, and prednisolone. At 4.5 months after
If such ulcers fail to resolve, herpes simplex should be con- transplantation recipients were randomized (n = 300 or
sidered. Finally, patients with suboptimal renal function, 60% of the total recruited) to continue on cyclosporine
particularly patients with proteinuria, are prone to develop (n = 145) or switch to an everolimus-based regimen (n =
marked proteinuria after conversion (see later).80,82 155) with everolimus trough concentrations of 6 to 10
Despite the potential drawbacks in terms of side effects, ng/mL. Those recipients who were switched to everolimus
evidence is accumulating that conversion from CNIs to showed a significant improvement in 12-month GFR (the
mTOR inhibitors may be worthwhile in patients with primary endpoint) compared with those who remained
interstitial fibrosis and tubular atrophy or histologic signs on cyclosporine (71.8 mL/min/1.73 m2 vs. 61.9 mL/
of CNI toxicity and, at least in the short term, may lead to min/1.73 m2). Conversion to everolimus was associated
improved graft function.83 The optimal time for conver- with a 6% higher rate of postrandomization acute rejection
sion in such patients is unclear, but early rather than late than remaining on cyclosporine (10% vs. 3%) but over the
conversion may be best, before irreversible fibrosis and entire study period acute rejection rates were similar (15%).
tubular atrophy become extensive. With the increasing use The overall efficacy and safety were similar in the two study
of organs from more marginal donors that already have groups. Five-year follow-up of the ZEUS study showed a sus-
some damage at baseline, cessation of CNIs and replace- tained benefit in terms of renal function in the everolimus
ment with mTOR inhibitors may become a useful strategy arm (intention-to-treat: Nankivell GFR 66.2 mL/min/1.73
to prevent further decline in graft function from a subopti- m2 in EVL arm, 60.9 mL/min/1.73 m2 in CsA arm), with
mal baseline. Switching to mTOR inhibitors in patients who greater benefit when analyzed in terms of those remaining
are experiencing other side effects from CNIs, such as neu- on everolimus.89 This benefit is in spite of the cumulative
rotoxicity and diabetes, may also be a reasonable option. incidence of acute rejection postrandomization over the
Conversion from CNIs to mTOR inhibitors for patients who 5-year follow-up period being nonsignificantly higher in
develop hemolytic-uremic syndrome may also be consid- the everolimus arm (13.6% EVL, 7.5% CsA P = 0.095).
ered, although sirolimus itself has been identified as a cause The ZEUS trial contrasts with the results of ELEVATE, a
of this condition.84,85 Because mTOR inhibitors lead to an similar large multicenter trial randomizing 715 de novo
increased urinary excretion of uric acid, a further possible patients to stay on their CNI or switch to everolimus at 3
indication for use of mTOR inhibitors in the management of months.90 Patients received basiliximab induction and
severe gout in patients taking CNIs. either tacrolimus or cyclosporine-based immunosuppres-
The 3C study was a two-step trial, looking first to com- sion together with MPS and steroids: 360 were random-
pare the incidence of rejection in patients receiving either ized to everolimus, with 357 randomized to CNIs, of which
alemtuzumab or basiliximab induction, followed by a sec- 231 were on tacrolimus and 125 on cyclosporine, with one
ond randomization at 6 months to either sirolimus-based dropout starting on renal replacement in each arm. The pri-
immunosuppression or continuation on tacrolimus.86 The mary endpoint was change in MDRD GFR at 12 months,
alemtuzumab arm had no steroids and lower dose tacroli- and there was no significant difference between study arms
mus, in addition to mycophenolate sodium (MPS); the basi- (0.3 mL/min/1.73 m2 EVL, −1.5 mL/min/1.73 m2 CNI).
liximab arm had full dose tacrolimus, MPS, and steroids. However, there was a significantly better estimated glomer-
The study enrolled 852 patients, and the initial phase of ular filtration rate (eGFR) at 12 and 24 months in everoli-
the study showed that alemtuzumab was associated with mus-treated patients (24-month GFRs: 62.5 mL/min/1.73
less rejection but no difference in transplant survival at 6 m2 EVL, 57.4 mL/min/1.73 m2 CNI, P = 0.005). There was
months. In the subsequent randomization, 197 patients a greater proportion of biopsy-proven acute rejection on
were allocated to stay on tacrolimus whereas 197 switched everolimus compared with CNI overall (9.7% EVL, 4.8%
to sirolimus.87 There was no difference in GFR at 18 months CNI, P = 0.014); when the CNIs were analyzed separately,
(12 months postrandomization) but a significantly higher everolimus- and cyclosporine-treated patients had a similar
(RR 5.15) incidence of acute rejection in the sirolimus- incidence (8.8%), whereas tacrolimus patients had a sig-
treated patients (14.7% SRL, 3% Tac, P < 0.001); the inci- nificantly lower incidence (2.6%) than everolimus-treated
dence of rejection on sirolimus was no different whether patients. As with other mTOR studies, there were more dis-
the patient had alemtuzumab or basiliximab induction. continuations in the everolimus group than the CNI group
In addition, 49% of patients discontinued sirolimus-based (23.6% EVL, 8.4% CNI). Other observations were a signifi-
therapy compared with 6% tacrolimus-based therapy after cantly greater mean urinary protein/creatinine ratio in the
randomization. Serious infections were also more common everolimus patients than the CNI patients (36.4 mg/mmol
on sirolimus (48.2% SRL, 35.5% Tac, P = 0.008). The high EVL, 19.1 mg/mmol CNI, P < 0.001), with no difference
discontinuation rate on sirolimus and the high rejection between tacrolimus- and cyclosporine-treated patients.
rate probably account for why the patients randomized to
sirolimus saw no benefit in GFR on an intention-to-treat
analysis.
The ZEUS study, a multicenter, open-label, randomized
mTOR Inhibitors and Malignancy
controlled trial of everolimus-based, CNI-free immunosup- mTOR INHIBITORS AS ANTITUMOR AGENTS
pression suggested that early replacement of CNIs with
everolimus-based immunosuppression is advantageous The P13K/Akt signaling pathway is often dysregulated
for renal function.88 The study recruited 503 adult recipi- in malignant cells, causing activation of mTOR and thus
ents of renal transplants, and they received initial therapy stimulating cell proliferation, tumor growth, and produc-
with basiliximab induction together with cyclosporine, tion of growth factors, such as vascular endothelial growth
270 Kidney Transplantation: Principles and Practice

factor, that stimulate angiogenesis, a key component of recipients and is an important consideration when consid-
many tumors. Because of the central role of mTOR in regu- ering the optimal immunosuppressive regimen, especially
lating cellular processes in malignant cells it is an attractive for patients considered at particularly high risk because of
therapeutic target.91 Although intuitively the immunosup- previous skin cancers or premalignant skin lesions.100 One
pressive effects of mTOR might be expected to outweigh any of the earliest reports in this context was a single-center
beneficial effect on limiting tumor cell growth in patients randomized controlled trial that showed that renal trans-
with malignancy, this seems not to be the case and a number plant recipients with premalignant skin lesions who were
of new sirolimus derivatives (rapalogs) have been developed switched to sirolimus-based immunosuppression had less
specifically for use as antitumor agents, including temsiroli- progression and in some cases even regression of prema-
mus (CCI-779), a sirolimus derivative formulated for intra- lignant skin lesions.101 There were 25 patients assigned to
venous administration. mTOR inhibitors have been licensed the sirolimus arm and 19 to the control arm, and of the
for a number of different tumors, including renal cell carci- 9 patients who developed histologically confirmed NMSC
noma, gliomas, neuroendocrine tumors, and advanced hor- during the 12-month follow-up period, only one was in the
mone receptor positive/HER2 negative breast cancer.92–95 sirolimus arm. Another multicenter study (the TUMOR-
The use of mTOR inhibitors is currently being tested in many APA study) randomly assigned renal transplant recipients
of the other common types of cancer including colorectal, who had at least one invasive posttransplant cutaneous
ovarian, prostatic, gastric, and pancreatic cancer. squamous cell carcinoma either to remain on CNI-based
immunosuppression or to switch to sirolimus.102 New
mTOR INHIBITORS AND POSTTRANSPLANT squamous cell cancers developed in 14 (22%) of those who
switched to sirolimus and 22 (39%) of those randomized to
MALIGNANCY
stay on calcineurin blockers with a median time to onset
Because patients after renal transplantation are at increased of 16 versus 7 months respectively (P = 0.02). Switching
risk of developing most types of malignancy, particularly to sirolimus appears to be an effective strategy for reducing
squamous cell cancer of the skin and lymphoma, the anti- the risk of recurrent skin cancer and is a reasonable treat-
cancer effects of mTOR inhibitors are of great relevance. ment option although not without potential side effects.
Several reports suggest that maintenance immunosuppres- In the TUMORAPA study, twice as many serious adverse
sion with mTOR inhibitors after renal transplantation may events occurred in those who switched to sirolimus and
be associated with a reduced risk of posttransplant malig- 23% of patients discontinued sirolimus because of adverse
nancy. It is, however, important to bear in mind that this events.
could, at least in part, reflect the ability of CNIs to increase
the risk of malignancy rather than any direct protective mTOR INHIBITORS AND POSTTRANSPLANTATION
effect of mTOR inhibitors. A multivariate analysis of post- LYMPHOPROLIFERATIVE DISORDER
transplant malignancy in 33,249 renal allograft recipients
in the US revealed that the incidence rates of any type of Everolimus and sirolimus have been shown to markedly
posttransplant malignancy were 0.6% in patients taking inhibit the growth of human posttransplant lymphoprolif-
mTOR inhibitors, 0.6% for patients taking mTOR inhibitors erative disorder (PTLD)-derived cell lines and Epstein–Barr
plus CNIs, and 1.8% for patients taking CNIs alone.96 Simi- virus-transformed B lymphocytes in vitro and in vivo.103–
larly, the incidence of posttransplant malignancy in adults 105 Nevertheless there is relatively limited evidence to sup-

randomly assigned to remain on sirolimus and CNIs was port the use of mTOR inhibitors in the management of
found to be greater than in subjects randomly assigned to PTLD, although a renal transplant recipient in whom dis-
early CNI withdrawal and an increased dose of sirolimus.97 seminated PTLD resolved completely after conversion of
A more recent meta-analysis of 20 randomized controlled immunosuppression to sirolimus has been reported.106 In a
trials and two observational studies, comprising 39,039 pooled analysis from nine European centers, 19 renal trans-
patients, suggested there was a protective effect of sirolimus, plant recipients with PTLD were studied after conversion
at least for nonmelanoma skin cancer (incidence rate ratio to mTOR inhibitors and subsequent withdrawal or mini-
[IRR] 0.49; 95% CI 0.32, 0.76), particularly where the com- mization of CNIs.107 Remission of disease was observed in
parator was cyclosporine.98 There was also a suggestion of 15 of the recipients, although because 12 of the recipients
a lower incidence of kidney cancer (IRR 0.40; 95% CI 0.20, also received rituximab or chemotherapy with cyclophos-
0.81) and a higher rate of prostate cancer (IRR 1.85; 95% phamide, doxorubicin, vincristine, and prednisone (CHOP)
CI 1.17, 2.91). In a separate, earlier analysis of 21 trials and it is difficult to know how much of the benefit was directly
5876 patients, a similar effect was seen for sirolimus in reduc- attributable to use of mTOR inhibitors. Sirolimus did not
ing the incidence of nonmelanoma skin cancer (NMSC), par- appear to modify the risk of developing PTLD in an analysis
ticularly where the patient was converted to sirolimus from of 25,127 patients who underwent renal transplantation in
another regimen.99 This study also showed a reduction in the the US, 34 of whom developed PTLD.108
risk of other cancers (risk reduction [RR] 0.52, 0.38–0.69),
but worryingly indicated an increased risk of death on siroli- mTOR INHIBITORS AND KAPOSI’S SARCOMA
mus compared with controls.
mTOR inhibitors may have a useful role in the treatment of
mTOR INHIBITORS AND NONMELANOMA SKIN renal transplant recipients who develop Kaposi’s sarcoma
associated with herpesvirus-8, especially if the disease is
CANCER
confined to the skin. In a study of 15 patients who devel-
The most frequent type of cancer after renal transplan- oped cutaneous Kaposi’s sarcoma after renal transplanta-
tation is NMSC, which affects over half of all transplant tion while taking cyclosporine, switching them to sirolimus
18 • mTOR Inhibitors: Sirolimus and Everolimus 271

led to complete, histologically confirmed remission in all


patients for the duration of the study (6 months) with pres-
Safety and Side Effects of mTOR
ervation of graft function.109 However, response varies and Inhibitors
may depend on the severity of disease. A retrospective anal-
ysis in which 14 renal transplant recipients with Kaposi’s It was not until the phase 2 studies of sirolimus by Groth
sarcoma (including several with visceral or advanced dis- and Kreis and colleagues49,50 that the sirolimus-specific side
ease) were switched from CNIs to sirolimus showed that the effects became clear, because until then the agent had been
switch was generally well tolerated. Complete remission used in conjunction with CNIs. Table 18.1 shows the prin-
was seen in two patients, and a partial response was seen cipal side effects found in these studies. In contrast to CNIs,
in a further eight, although three of the partial responders it was notable that although mTOR inhibitors may cause
with advanced disease relapsed after several months.110 In a range of agent-specific side effects, these did not include
patients who do not respond to mTOR inhibitors or show neurotoxicity, hypertension, or gingival hyperplasia.
significant side effects it has been suggested that a combina-
tion of mTOR inhibitor and leflunomide (which inhibits Akt INFECTION
signaling upstream from mTOR) may act synergistically in
the treatment of Kaposi’s sarcoma.111 The use of mTOR- The incidence and pattern of infections reported in patients
based immunosuppression, however, does not necessarily receiving mTOR inhibitors is broadly similar to patients
prevent the development of posttransplant Kaposi’s sar- receiving CNI-based immunosuppression. Neither the US
coma.112 Further studies are needed to evaluate the role of study nor the global studies of de novo sirolimus use (see
mTOR inhibitors in the treatment of Kaposi’s sarcoma and Table 18.2) identified any particular problem with infection
to determine the optimal treatment strategy for patients over and above that observed in the comparator groups,
with more advanced disease. although the global study noted an increase in the inci-
dence of mucosal lesions attributed (but without virologic
confirmation) to herpes simplex virus.61,62 A meta-analy-
mTOR INHIBITORS AND BK VIRUS
sis of mTOR inhibitor use as primary immunosuppression
Polyoma virus nephropathy caused by BK virus is a after kidney transplantation confirmed the overall safety of
well-recognized cause of kidney graft dysfunction. The inci- mTOR inhibitors in terms of infection and noted that when
dence in patients with sirolimus-based immunosuppression mTOR inhibitors were substituted for antimetabolites, there
has been reported to be relatively low, compared with other was a reduction in the incidence of cytomegalovirus infec-
immunosuppressive regimens,113,114 although this reduced tion.55 Some studies have suggested that the incidence of
incidence may not be the case where mTORs are combined pneumonia may be greater in patients receiving mTOR
with CNIs.115 Early reports that a sirolimus-based regimen inhibitors, but the evidence for this remains inconclu-
may be associated with early resolution of nephropathy sive and is confounded by the occurrence of drug-induced
have been confirmed by later reports using everolimus- pneumonitis.
based regimens.116–118
These observations are supported by in vitro evidence METABOLIC COMPLICATIONS
suggesting that sirolimus may be working by blocking intra-
cellular protein kinase pathways used by the virus,119,120 Diabetes
an effect that is complemented by the addition of lefluno- New-onset diabetes after transplantation (NODAT) is a
mide.120 Clinical efficacy of the combination of everolimus common complication of CNIs, but is also associated with
and leflunomide has been reported in two cases.121 mTOR use. A meta-analysis of trials where patients were
converted to an mTOR from a CNI showed a similar inci-
MTOR INHIBITORS IN POLYCYSTIC KIDNEY dence in patients converted to mTORs (5.5%) as those on
CNIs (5.05%).130 An earlier analysis of the US Renal Data
DISEASE
System had shown that patients treated with sirolimus and
Autosomal dominant polycystic kidney disease (ADPKD) is an antimetabolite were at higher risk of NODAT compared
a monogenic disorder characterized by the development of with patients on cyclosporine (hazard ratio [HR]1.36, 95%
cysts in the kidneys that results in loss of functioning neph- CI 1.09–1.69; P < 0.01),131 and oncology trials also sug-
ron mass such that patients develop kidney failure in adult- gest a high incidence of hyperglycemia.132 The mechanism
hood.122 Patients with ADPKD frequently have cysts in other of the mTOR effect is complex and includes inhibition of the
organs, such as the liver that can compromise hepatic func- intracellular response to insulin, decreased glucose trans-
tion. Studies have implicated mTOR in cyst pathogenesis, port, decreased glycogen synthesis, and increased glycoly-
and mTOR inhibitors have been shown to slow cyst devel- sis in addition to beta cell toxicity.133–136
opment in animal models.123–125 In human patients with
ADPKD who went on to receive a kidney transplant, mTOR Lipids
inhibition was associated with a reduction in hepatic cysts The mTORC1 plays an important role in regulating lipid
in some subjects.126 Randomized controlled trials of mTOR metabolism,137 so it is no surprise that mTOR inhibition
inhibitors in nontransplanted ADPKD patients have failed is associated with abnormalities of lipid metabolism.130 In
to demonstrate efficacy in preventing cyst formation.127,128 the ORION study, 42.8% of patients on sirolimus and MMF
However, recent data suggest that combined phosphoinosit- developed hyperlipidemia compared with 20.1% on tacro-
ide 3-kinase (PI3K) and mTOR inhibition may be a more limus.56 In a similar study comparing cyclosporine and
effective strategy,129 but this remains to be tested in patients. sirolimus, 25.5% of sirolimus-treated patients and 21.7% of
272 Kidney Transplantation: Principles and Practice

cyclosporine-treated patients developed hypercholesterol-


emia, and 43.2% of sirolimus patients developed hyperlipid-
emia compared with just 26.1% on cyclosporine.138 The full
significance of the increased lipids associated with mTOR
inhibitors is unclear, but it is a long-term concern. There
is evidence in animal models that sirolimus inhibits graft
vasculopathy,139 an observation confirmed with everoli-
mus in heart transplant recipients.140 Sirolimus also seems
able to prevent the accelerated vascular disease seen in
cholesterol-fed, apolipoprotein E-deficient mice despite the
high-cholesterol diet.141 The occurrence of lipid abnormali-
ties seems to be, at least in part, genetically determined with
polymorphisms in apolipoprotein A being implicated.142

INTERSTITIAL LUNG DISEASE (PNEUMONITIS)


A
Pneumonitis is one of the most feared complications of
mTOR inhibitors and may progress to pulmonary failure. It
may occur at any time during treatment, but is most com-
mon within 6 months of starting treatment. It presents
with progressive dyspnea, dry cough, fatigue, hypoxia and
fever.143–146 Imaging reveals bilateral pulmonary infil-
trates (Fig. 18.3), most commonly in the lower lobes, and
pulmonary function tests may show a restrictive pattern
with reduction in the diffusing capacity of lung for carbon
monoxide (DLCO) being an early feature. Open lung biop-
sies have revealed granulomata in some cases. The effect is
reversible with discontinuation of the mTOR inhibitor. The
true incidence of mTOR-associated pneumonitis is unclear,
and it is probably underrecognized and underreported.
The first reports of sirolimus-associated pneumonitis were
in 2000,147 and these were followed by a disclosure from
the US Food and Drug Administration (FDA) of 31 other
cases of interstitial pneumonitis associated with sirolimus
use.148 Earlier studies had reported an increased incidence
of pneumonia (see Table 18.1),49 and one of the first studies
reported an excess of Pneumocystis jirovecii pneumonia149; it
B
is possible that some of these were in fact sirolimus-induced
pneumonitis. Ten years previously, the complication had
been noted as the principal cause of death in pigs undergoing
renal transplantation with sirolimus.5 Pneumonitis has also
been shown to occur with other mTOR inhibitors including
everolimus.150 There is evidence that radiologic changes
may occur in asymptomatic patients, making true incidence
difficult to quantify unless active surveillance is taking place.
This has occurred in an oncology setting in Japan, where an
incidence of 17.4% for everolimus was reported.151
The etiology of the pneumonitis that occurs with mTOR
inhibitors is unclear. Reports suggest that it is more com-
mon in patients switching from a CNI to sirolimus, or hav-
ing a CNI withdrawn from sirolimus/CNI combination, and
having a high drug concentration.143,148,152 A mortality
of 12% was noted in the FDA report, although early recog-
nition of the problem, with immediate discontinuation of C
the mTOR inhibitor, should reduce the mortality from this Fig. 18.3 Sirolimus-induced pneumonitis. (A) Plain chest radio-
complication.146 graph shows bilateral interstitial infiltration. (B) High-resolution com-
puted tomography scan shows patchy ground-glass opacification
and interstitial reticular change. (C) Immunohistologic analysis of
HEMOLYTIC-UREMIC SYNDROME transbronchial lung biopsy specimen in sirolimus-induced pneumo-
nitis shows heavy interstitial CD4 T-cell infiltrate (immunoperoxidase,
One of the main attractions of mTOR inhibitor therapy is ×400). (A and B, courtesy Dr. A. Tasker; C, courtesy Dr. M. Griffiths.)
its perceived lack of nephrotoxicity, but it is not entirely
18 • mTOR Inhibitors: Sirolimus and Everolimus 273

devoid of adverse effects on the kidney. The most serious


of these is its association with hemolytic-uremic syndrome
(HUS; thrombotic microangiopathy). HUS was identified
as a potential problem in patients taking cyclosporine and
sirolimus,153,154 but subsequent reports indicated that it
could occur with everolimus,66 and also with sirolimus in
the absence of CNIs.84,85 It also occurs in the native kidneys
of nonrenal transplant recipients.155,156

PROTEINURIA
Proteinuria is a common manifestation of mTOR inhibitor
toxicity in patients converted to mTOR for renal impair-
ment, with nephrotic range proteinuria being common.157
It is most common in patients who already have a degree of
proteinuria at the time of conversion,158 with the absence
of proteinuria being the best indicator of improvement in
renal function after conversion.159,160 It has thus been sug-
gested that proteinuria has been masked by the CNI only
to be revealed with its withdrawal.161 The actual cause of
the proteinuria is unclear. In one study of four patients who
developed proteinuria, biopsy specimens revealed glomeru-
lonephritis (membranoproliferative glomerulonephritis in
one, membranous glomerulonephritis in another, and IgA Fig. 18.4 Sirolimus-induced erythema and limb edema. Acute ery-
nephropathy in the other two).162 The proteinuria resolved thema and swelling in the limbs associated with sirolimus. The symp-
when the patients were converted back to CNIs and the toms resolved after administration of steroids.
sirolimus was stopped. In a separate study of liver trans-
plant recipients who had developed renal impairment, no nontransplant settings.176 Although sirolimus is associated
proteinuria was seen on conversion to sirolimus, suggest- with a higher incidence of delayed graft function and pro-
ing that preexisting renal damage may be a prerequisite for longed recovery of function, renal function in the long term
the development of proteinuria,163 although a change in does not seem to suffer.177,178
glomerular hemodynamics may contribute.164 Proteinuria
has also been observed in patients undergoing islet trans- PERIPHERAL EDEMA
plantation and receiving sirolimus in whom underlying
diabetic nephropathy may have been contributory.165 The occurrence of edema in patients taking mTOR inhibi-
The actual pathogenesis of the proteinuria is unclear. tors is well described. Most edema affects the lower limb
It may relate to inhibition of vascular endothelial growth and may be unilateral or bilateral (Fig. 18.4);179 it is not
factor (VEGF) secretion and activity, tubular epithelial necessarily ipsilateral to the kidney transplant. Angio-
cell protein endocytosis, and podocyte integrity and func- edema affecting the eyelids and tongue has also been
tion.166–168 Blockade of the angiotensin system may be use- described.180–183 It may be less common with everolimus
ful to limit proteinuria.164,169 than sirolimus.184 The condition appears to be related to
disrupted lymphatic drainage, and its pathogenesis may
DELAYED RECOVERY FROM ISCHEMIA- be linked to disruption of VEGF action, which play a role in
lymphangiogenesis.185 It may resolve completely on early
REPERFUSION INJURY
discontinuation of mTOR inhibition.
Delayed recovery of normal kidney function (delayed graft
function) is a common manifestation of ischemia-reperfu- WOUND HEALING AND LYMPHOCELE
sion injury, more common in recipients of kidneys donated FORMATION
after circulatory death where both warm ischemia and cold
ischemia contribute to renal injury. Reduced exposure to One of the most concerning complications of de novo
CNIs in the early posttransplant period is practiced in many mTOR inhibitor use is the potentially detrimental effect
transplant centers, so “nonnephrotoxic agents” such as they have on the operative site. mTOR inhibitors not
mTOR inhibitors were considered to be an attractive alter- only impair wound healing but are also associated with
native. However, early experimental work in rats showed a high incidence of lymphoceles after renal transplanta-
delayed recovery from ischemia-reperfusion injury,170 and tion. Wound problems include fluid collections around
this has subsequently been observed in the clinic in small the graft and beneath the skin, superficial infections,
retrospective studies and registry analyses.171–173 Everoli- and late incisional hernia.186 Anastomotic healing has
mus may have less of an effect on delay of recovery.174 not been reported as a problem after renal transplanta-
The mechanism underlying this observation presumably tion, but poor healing of the airway anastomosis has been
relates to the inhibition of cell proliferation affecting tubu- cited after lung transplantation,187,188 and there is evi-
lar repair.175 However, high doses of mTOR inhibitors have dence in the pig that ureteric anastomoses are less strong
been associated with new onset acute tubular necrosis in when performed in the presence on mTOR inhibitors.189
274 Kidney Transplantation: Principles and Practice

The problems observed with wound healing may result lesions were all mild and resolved spontaneously without
from mTOR inhibition reducing the fibroblast response discontinuing sirolimus.62
to fibroblast growth factor,190 and to a lack of neovascu- Mouth ulcers are also common in patients converted to
larization of wounds because of decreased production of mTOR inhibitors. Again, such ulcers usually resolve spon-
vascular endothelial growth factor. A systematic review of taneously, but they can be problematic. In one prospective
randomized controlled trials of wound complications and randomized study in which renal transplant recipients were
lymphoceles after solid organ transplantation supported converted at 1 year from a steroid-free regimen of tacrolimus
the view that mTOR inhibitors are best avoided during the and MMF to sirolimus and MMF, oral ulceration occurred
first few months after transplantation.191 Pooled analysis in 9 of 15 converted patients. The mucosal lesions healed
showed a higher incidence of wound complications (odds within 2 weeks of discontinuing sirolimus, but the problem
ratio [OR] 1.77, CI 1.31–2.37) and lymphoceles (OR 2.07, led to premature cessation of the study.148 The authors pos-
CI 1.62–2.65) for kidney transplant recipients receiving tulated that the high incidence of oral ulceration may have
mTOR inhibitors along with calcineurin blockers. Wound been attributable to overimmunosuppression during con-
complications (OR 3.00, CI 1.61–5.59) and lympho- version, the use of oral emulsion of sirolimus rather than
celes (OR 2.13, CI 1.57–2.90) were also more common tablets, and the lack of corticosteroids.148 In a randomized
in recipients receiving mTOR inhibitors along with anti- study of conversion from CNIs to sirolimus after renal trans-
metabolites. A nonsystematic review of wound healing plantation, aphthous-type mouth ulcers occurred in one-
complications in relation to use of mTOR inhibitors sug- third of patients during the first 2 weeks after conversion,
gested that wound complications may be less of a problem although all resolved with adjustment of sirolimus to the
with the lower exposure regimens of mTOR inhibitors that lower end of the target range of 5 to 15 ng/mL.79
have now largely superseded the historical high-exposure Aphthous-type ulcers are also common with other mTOR
regimens.192 It has also been suggested that obesity may inhibitor analogs used in oncology, and they are distinct
adversely influence wound healing in patients receiving from the mucositis commonly seen with chemotherapeutic
mTOR inhibitors193 and that it would be prudent to avoid agents.194 The association between mTOR inhibitors and
their use in patients undergoing surgery, including trans- mucosal ulceration may be attributable predominantly to
plantation, whose body mass index is greater than 32 kg/ their detrimental effect on wound healing, rather than any
m2 until more data are available.192 direct effect on initiating ulcer formation. Although they
may resolve with time, topical clobetasol has been reported
to induce prompt resolution.195 More recently, colchicine,
APHTHOUS ULCERS
which is active against Behçet’s disease, has been shown
Oral ulceration (stomatitis) manifesting as painful gingival to be effective against severe everolimus-induced oral
or buccal ulceration of mucosa leading to pain on eating, ulceration.196
is a well-documented and troublesome side effect of mTOR
inhibition (Fig. 18.5). The ulcers are usually small but mul- RASH
tiple, and in some cases, may be related to herpes simplex
virus infection. In the global phase 3 study of de novo treat- As already noted, rash is a common complication of mTOR
ment with sirolimus, ulceration of the oral mucosa was inhibitor therapy and most commonly takes the form of
observed in 19% of patients randomly assigned to 5 mg/ an inflammatory acneiform eruption197 or a dermatitis-
day of sirolimus, 10% of patients assigned to 2 mg/day of like rash affecting the hands and, in particular, the fin-
sirolimus, and 9% of patients in the placebo group.62 The gers (Fig. 18.6). This side effect was apparent in both the

Fig. 18.5 Sirolimus-induced oral ulceration. Solitary aphthous-type


ulcer on the undersurface of the tongue occurring several days after
late (>6 months) conversion from calcineurin inhibitor to sirolimus. Fig. 18.6 Sirolimus-induced rash. After conversion from a calcineu-
Such lesions are often multiple and painful and can be distressing for rin inhibitor to sirolimus, patients commonly develop skin problems
the patient. They usually resolve rapidly after adjustment of sirolimus that may take the form of a dermatitis-like rash affecting the hands and,
to the lower end of the target range of 5 to 10 ng/mL. in particular, the fingers.
18 • mTOR Inhibitors: Sirolimus and Everolimus 275

early multicenter randomized trials of de novo sirolimus marrow.203 It was suggested instead that sirolimus may
treatment after renal transplantation. In the US study, an have a direct effect on iron homeostasis.203
acneiform rash was observed in 25% of recipients on 2 mg/ Thrombocytopenia was identified as a side effect of siro-
day, 19% of recipients on 5 mg/day, and 11% in the aza- limus in the global and the US phase 3 randomized trials
thioprine control group.61 In the global phase 3 study, rash of de novo sirolimus and seemed to be dose related.61,62 In
was observed in 14% of patients randomly assigned to 5 both studies, a few patients randomly assigned to the higher
mg/day of sirolimus, 4% of patients assigned to 2 mg/day of dose (5 mg/day) of sirolimus had to have sirolimus dis-
sirolimus, and 5% of patients in the placebo group.62 In the continued because of thrombocytopenia (6 of 208 [2.8%]
ORION study the incidence of acne was 9.2% and 12.5% in in the global study and 3 of 274 [1.1%] in the US study),
the sirolimus arms, compared with just 2.2% in the tacro- although none of the patients experienced severe throm-
limus arm.56 Two systematic reviews in oncology suggest bocytopenia or were reported to have had related hemor-
that the incidence of any rash with everolimus is around rhage. mTOR inhibitors may reduce circulating platelets as
28%.198,199 part of their inhibitory effect on hematopoietic cytokines.
In studies in which an in depth dermatologic analysis has In addition, sirolimus has been shown to promote agonist-
been undertaken, the incidence of dermatologic side effects induced platelet aggregation in vitro,204 and conceivably if
is considerably higher. A cross-sectional study of cutaneous increased platelet aggregation occurs in vivo, it may pro-
adverse events in renal transplant recipients receiving long- mote increased removal of platelets by the spleen.
term sirolimus-based immunosuppression reported the pres- Although it is well recognized that mTOR inhibitors may
ence of an acne-like eruption in 46%, scalp folliculitis in 26%, reduce the platelet count, this is not usually of clinical sig-
and hidradenitis suppurativa in 12% of patients.200 In the nificance and it rarely requires cessation of therapy. Throm-
absence of a control group, it is difficult to attribute such find- bocytopenia most often occurs within the first month of
ings exclusively to mTOR inhibitors, but both studies indicate starting sirolimus, and its occurrence correlates with whole
the high frequency of dermatologic complications associated blood trough levels of sirolimus that exceed 16 ng/mL. If the
with mTOR inhibitors. Although rashes are usually mild, platelet count falls significantly, it usually responds well to
they may be a reason for discontinuing mTOR inhibitors. dose reduction without the need to withdraw mTOR inhibi-
The pathophysiology of rash in patients taking mTOR inhibi- tors.205 Finally, mTOR inhibitors may produce mild leuko-
tors is unclear, but may be attributable to their effect on the penia, which is usually transient and dose related.
epidermal growth factor receptor, which is important in the
differentiation and development of the hair follicle.197 GASTROINTESTINAL SYMPTOMS
ANEMIA, THROMBOCYTOPENIA, AND Gastrointestinal side effects include abdominal pain, nau-
sea, and vomiting, but the most common symptom is diar-
LEUKOPENIA
rhea, which is usually mild, dose related, and does not
Anemia, thrombocytopenia, and leukopenia are all well rec- require mTOR withdrawal. In the pivotal phase 3 studies
ognized side effects of mTOR inhibitor use. Although throm- of de novo sirolimus, mild diarrhea was observed in 27%
bocytopenia attracted the most attention in early studies, to 32% of patients receiving 5 mg/day of sirolimus, 16% to
anemia has emerged as the most significant clinical problem. 20% of those receiving 2 mg/day of sirolimus, and 11% to
Anemia is a common complication during the first 6 months 13% of patients in the control groups.61,62 Mild diarrhea is
after renal transplantation regardless of the immunosuppres- particularly common in patients receiving a combination
sive regimen,201 but the incidence is increased with use of on mTOR inhibitors and MMF50 and may be related to the
mTOR inhibitors. In the global study of de novo sirolimus in pharmacokinetic interaction between the two agents;206
renal transplantation, anemia was observed in 16% of recipi- concentration-controlled administration of MMF markedly
ents taking 2 mg/day and 27% of recipients taking 5 mg/day reduces gastrointestinal symptoms.206
sirolimus. The incidence of anemia in the 5 mg/day group
was significantly higher than in the placebo group (13%) THROMBOSIS
receiving cyclosporine and steroids.62 mTOR inhibitor dose
adjustment may be required, and in some patients adminis- Sirolimus, like CNIs, may increase platelet aggregation
tration of erythropoietin may be necessary. in vitro,204 and it has been suggested that sirolimus, when
Anemia after renal transplantation most often results used in combination with CNIs, may increase the risk of
from iron deficiency and defective erythropoietin produc- hepatic artery thrombosis after liver transplantation.207
tion, but the mechanisms responsible for mTOR inhibitor- There is no published evidence that mTOR inhibitors are
induced anemia are unclear. Sirolimus blocks the in vitro associated with an increased risk of thromboembolic events
response of bone marrow cells to several hematopoietic after renal transplantation, although mTOR-coated coro-
cytokines, including granulocyte colony-stimulating fac- nary stents are believed to have an increased incidence of
tor, interleukin-3, and kit ligand.202 Although mTOR thrombosis, but the evidence for this is poor.208 In a ret-
inhibitor-induced suppression of nonerythroid bone mar- rospective single-center analysis of deep vein thrombo-
row cells contributes to leukopenia and thrombocytopenia, sis, graft thrombosis, and pulmonary embolism in renal
the extent to which they cause anemia by suppression of transplant recipients, the addition of sirolimus in recipients
erythrocyte production is uncertain. Sirolimus has been taking cyclosporine did not increase the risk of postopera-
observed to reduce hemoglobin levels without reducing the tive thrombotic events.209 A strong correlation between
erythrocyte count in renal transplant recipients, arguing the development of deep vein thrombosis and lymphocele
against a direct antiproliferative effect on erythroid bone was observed in patients receiving sirolimus,209 and the
276 Kidney Transplantation: Principles and Practice

increased risk of deep vein thrombosis in patients develop-


ing lymphocele should be kept in mind.
In vitro sirolimus has been shown to reduce the expres-
sion of tissue plasminogen activator and induce the
expression of plasminogen activator inhibitor by human
umbilical vein endothelial cells, providing a possible under-
lying mechanism for the increased thromboembolic disease
seen with mTOR inhibitors.210

RENAL TUBULAR EFFECTS: HYPOKALEMIA AND


HYPOPHOSPHATEMIA
mTOR inhibitors may contribute to hypokalemia after renal
transplantation, and in the phase 2 and 3 trials of primary
treatment with sirolimus, values of serum potassium less
than the normal range were recorded during the first 3
months in about half of patients.51 Hypokalemia is usually
mild and only about 10% of patients required a period of
potassium supplementation, which readily corrected the A B
problem.51 Hypokalemia may be partially related to the
Fig. 18.7 Radionuclide bone scan in a patient with sirolimus-
dose of mTOR inhibitors given and seems to be a result of induced bone pain. The patient complained of pain affecting the
mTOR inhibitor-induced alterations in tubular function feet, ankles, and knees. The diagnosis was confirmed by radioisotope
leading to increased tubular secretion of potassium.211 scanning that revealed areas of increased uptake in the knees and at
Hypophosphatemia is also common in the first few weeks the ankles (arrows, A). After sirolimus dosage reduction, the symptoms
resolved, and the bone scan returned to normal (B).
after renal transplantation and is multifactorial in etiol-
ogy. Although reduced serum phosphate levels may be in vitro and reduces bone loss in an oophorectomized rat
observed more often during the first 3 months in patients model.215 Markers of bone turnover (serum osteocalcin and
receiving mTOR inhibitors, this is rarely a clinically sig- urinary N-telopeptides) also are significantly lower in renal
nificant problem, and values return to normal with time transplant recipients taking de novo sirolimus compared
and dose adjustment.51 The mechanism underlying mTOR with recipients taking cyclosporine.216 However, conflict-
inhibitor-associated hypophosphatemia are not completely ing evidence exists. For example, in one retrospective study
understood, but mTOR inhibitors may impair renal tubular mTOR inhibitors were associated with osteoporosis,217
phosphate reabsorption, prolonging the phosphate leak.212 and in rodent models of senile osteoporosis sirolimus has
been shown to both cause bone loss,218 and reduce it.219
BONE EFFECTS Such data suggest that more extensive clinical studies with
extended follow-up are needed.
Arthralgia was identified as a side effect of mTOR inhibitors
in the global phase 3 study of sirolimus. It was observed in
27% of recipients on the higher dose (5 mg/day) of siroli- LIVER FUNCTION ABNORMALITIES
mus compared with 16% and 13% of recipients on low-dose Sirolimus tends to cause increased levels of transaminases
sirolimus or placebo.62 Similar to the CNI-induced pain syn- (alanine aminotransferase and aspartate aminotransferase)
drome, bone pain associated with sirolimus affects weight- and lactate dehydrogenase. Whether this is clinically signifi-
bearing areas, particularly the feet, ankles, and knees, cant is unclear. There are two reports of hepatotoxicity in a
although the pain may be unrelated to weight bearing. It renal transplant recipient220,221 and a series of 10 liver trans-
is generally bilateral and symmetric. The problem is much plant recipients in whom sirolimus was thought to be respon-
less common when lower doses of mTOR inhibitors are sible for abnormal liver function tests, with 2 of the patients
used, and symptoms may improve after dosage reduction or having liver biopsy specimens with eosinophilia and sinusoi-
respond to treatment with bisphosphonates or alfacalcidol. dal congestion.222 This latter group of 10 patients underwent
mTOR inhibitor-induced bone pain and CNI-induced pain transplantation for hepatitis C, which had reinfected their
syndrome are likely caused by a combination of increased grafts, making a clear association with sirolimus difficult.
adipocyte volume, reduced intraosseous perfusion, and
marrow edema giving rise to a “bone compartment syn- AMENORRHEA AND TESTICULAR FUNCTION
drome.”213 The diagnosis usually can be confirmed by
radionuclide bone scan (Fig. 18.7) or magnetic resonance In a study of conversion from CNIs to sirolimus, it was noted
imaging that reveals hyperemia and marrow edema. that all three female patients younger than 40 years of age
Osteoporosis and bone loss are common after renal trans- who were switched to sirolimus developed amenorrhea for
plantation, and there is evidence from preclinical and early a variable length of time and then resumed irregular men-
clinical studies that mTOR inhibitors may have bone-spar- ses.79 Disturbance of menstruation has been described in
ing properties compared with CNIs. Although sirolimus 50% women receiving sirolimus in a study of its effect on
is associated with bone-remodeling, it does not result in a polycystic kidneys; an increased incidence of ovarian cysts
loss of trabecular bone volume in rat studies, in contrast to was also noted in sirolimus-treated patients.223 Menstrual
CNIs.214 Similarly, everolimus inhibits osteoclast activity irregularities have also been noted with everolimus.224
18 • mTOR Inhibitors: Sirolimus and Everolimus 277

mTOR inhibitors have also been suggested to impair tes- broadly comparable with that of patients receiving stan-
ticular function and cause impaired spermatogenesis and dard CNI-based therapy. The agent-specific side effect pro-
reduced fertility in males.225,226 They have been noted to file of the mTOR inhibitors is also now well established and
cause a fall in testosterone levels with concomitant rise has relatively little overlap with that of CNIs.
in luteinizing hormone and follicle-stimulating hormone Overall, the clinical effect of mTOR inhibitors for immu-
levels.227,228 Although it has been suggested that amen- nosuppression after renal transplantation has been less
orrhoea may have resulted from a direct effect on the than anticipated when they were first introduced. They do
endometrium (e.g., inhibition of VEGF), observed effects on not share the same nephrotoxicity as CNIs (although they
ovaries and testes suggests that mTOR inhibitors are either can cause proteinuria) and there is some evidence that they
having an effect on the hypothalamic-pituitary-gonadal may limit chronic allograft nephropathy. However, the
axis or a direct effect on gonadal function. problems of impaired wound healing and lymphocele for-
mation argue against the immediate use of mTOR inhibi-
mTOR INHIBITION IN PEDIATRIC tors after renal transplantation, and the adverse effects of
mTOR inhibitors on the lipid profile together with the low
TRANSPLANTATION
but significant risk of life-threatening pneumonitis are con-
mTOR inhibitors have been used in pediatric kidney trans- cerns with long-term use.
plantation, but concerns remain regarding potential effects What then is their role? The combination of CNI and
on growth and maturation, and particularly in light of mTOR inhibitor is probably more powerful than the combi-
observed effects on sex hormone levels. nation of a CNI and mycophenolate, but the side effect profile
Short-term effects on growth were explored in matched makes them difficult to tolerate. They have little role in the
cohort study of 14 subjects receiving mycophenolate and absence of a CNI as the first agent posttransplant. As a proto-
cyclosporine, and 14 on low-dose everolimus and cyclospo- colled switch in the maintenance phase, there is conflicting
rine.229 Over the 2-year follow-up period there was no sig- evidence as to their efficacy. Where they are tolerated and
nificant difference in change in height standard deviation where conversion to mTOR inhibitor is not associated with
score, which takes into account age and sex. Such studies acute rejection, there is evidence of improvement in renal
are complex and need to take into account the onset and function, particularly when converted from cyclosporine.
duration of the pubertal growth spurt. However, their benefit compared with continuing on lower
In a small retrospective study of 15 adolescents aged 11 dose tacrolimus and mycophenolate is less clear, because the
to 15, who were switched to sirolimus at a mean 81 months latter combination is associated with less acute rejection and
posttransplant and followed for 3 years, testosterone levels a side effect profile the patient has already accepted.
post-switching were just below the normal range (348 and mTOR inhibitors are an attractive alternative for patients
360 ng/dL).230 However, interpretation is made difficult by who cannot tolerate CNIs. They are also an appealing option
the absence of true presirolimus levels on all the subjects. for recipients whose graft function is declining because of
The effect of mTOR inhibitors on the onset of puberty and CNI toxicity, or as a strategy for CNI-avoidance in patients
subsequent sex hormone levels has been studied.231 Of 108 receiving a marginal graft with suboptimal function. They
patients aged under 14 years who were analyzed, 67 received also have a role for recipients who have a high risk of malig-
an mTOR inhibitor (56 everolimus, 11 sirolimus) with 35 nant disease particularly those with previous skin cancer,
starting it in the first posttransplant year. All but 3 patients and they may be a useful option in recipients with trouble-
had concomitant CNI therapy at low dose, combined with some BK nephropathy.
low-dose mTOR inhibitor. Of the 41 not receiving an mTOR The optimal timing for the introduction of mTOR inhibi-
inhibitor, all but 4 received mycophenolate and a CNI. All 27 tors after renal transplantation is still unclear and better
prepubertal girls for whom data were available attained men- ways to manage the troublesome mucosal and dermato-
arche, doing so at a median age of 12.5 years (range 10–16), logic complications that are commonly encountered need
consistent with the general population with no difference to be found. Long-term data are now emerging to suggest
between mTOR and CNI groups. Of the 20 boys for whom the benefits of conversion in terms of improved function
data were available (14 mTOR, 6 CNI), all achieved sper- are maintained in the longer term (5 years), but it remains
marche with no difference between groups. Analysis of sex unclear whether these benefits translate into improved
hormones of boys and girls who had passed puberty showed graft survival.232 There is also still a need to determine the
no difference between CNI and mTOR inhibitor groups. extent to which any such benefits outweigh the long-term
Overall the retrospective nature, together with the size side effects of mTOR inhibitors, particularly their adverse
and duration of follow-up of the studies of mTOR inhibitors effects on the lipid profile.
in children, are too limited to draw firm conclusions on the
longer term effects of the drugs. References
1. Blenis J. TOR, the gateway to cellular metabolism, cell growth, and
disease. Cell 2017;171:10–3.
Summary and Conclusions 2. Baker H, Sidorowicz A, Sehgal SN, et al. Rapamycin (AY-22,989), a
new antifungal antibiotic. III. In vitro and in vivo evaluation. J Anti-
biot (Tokyo) 1978;31:539–45.
The mTOR inhibitors sirolimus and everolimus represent a 3. Sehgal SN, Baker H, Vezina C. Rapamycin (AY-22,989), a new anti-
distinct class of immunosuppressive agents and have been fungal antibiotic. II. Fermentation, isolation and characterization.
shown to be effective in preventing acute renal allograft J Antibiot (Tokyo) 1975;28:727–32.
4. Martel RR, Klicius J, Galet S. Inhibition of the immune response by
rejection and preserving renal function. Their safety pro- rapamycin, a new antifungal antibiotic. Can J Physiol Pharmacol
file in terms of posttransplant infection is satisfactory and 1977;55:48–51.
278 Kidney Transplantation: Principles and Practice

5. Calne RY, Collier DS, Lim S, et al. Rapamycin for immunosuppres- 31. Zochowska D, Bartlomiejczyk I, Kaminska A, et al. High-perfor-
sion in organ allografting. Lancet 1989;2:227. mance liquid chromatography versus immunoassay for the mea-
6. Collier DS, Thiru S, Calne R. Kidney transplantation in the dog surement of sirolimus: comparison of two methods. Transplant Proc
receiving FK-506. Transplant Proc 1987;19:62. 2006;38:78–80.
7. Starzl TE, Todo S, Fung J, et al. FK 506 for liver, kidney, and pancreas 32. Holt DW, Mandelbrot DA, Tortorici MA, et al. Long-term evaluation
transplantation. Lancet 1989;2:1000–4. of analytical methods used in sirolimus therapeutic drug monitor-
8. Schuler W, Sedrani R, Cottens S, et al. SDZ RAD, a new rapamycin ing. Clin Transplant 2014;28:243–51.
derivative: pharmacological properties in vitro and in vivo. Trans- 33. Anglicheau D, Le Corre D, Lechaton S, et al. Consequences of
plantation 1997;64:36–42. genetic polymorphisms for sirolimus requirements after renal trans-
9. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and plant in patients on primary sirolimus therapy. Am J Transplant
metabolism. Cell 2006;124:471–84. 2005;5:595–603.
10. Lucke C, Weiwad M. Insights into immunophilin structure and func- 34. Le Meur Y, Djebli N, Szelag JC, et al. CYP3A5*3 influences sirolimus
tion. Curr Med Chem 2011;18:5333–54. oral clearance in de novo and stable renal transplant recipients. Clin
11. Jhanwar-Uniyal M, Amin AG, Cooper JB, et al. Discrete signaling Pharmacol Ther 2006;80:51–60.
mechanisms of mTORC1 and mTORC2: connected yet apart in cel- 35. Picard N, Rouguieg-Malki K, Kamar N, et al. CYP3A5 genotype
lular and molecular aspects. Adv Biol Regul 2017;64:39–48. does not influence everolimus in vitro metabolism and clinical
12. Lazorchak AS, Su B. Perspectives on the role of mTORC2 in B lym- pharmacokinetics in renal transplant recipients. Transplantation
phocyte development, immunity and tumorigenesis. Protein Cell 2011;91:652–6.
2011;2:523–30. 36. Emoto C, Fukuda T, Venkatasubramanian R, et al. The impact of
13. Zeiser R, Leveson-Gower DB, Zambricki EA, et al. Differential impact of CYP3A5 3 polymorphism on sirolimus pharmacokinetics: insights
mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ from predictions with a physiologically-based pharmacokinetic
regulatory T cells compared with conventional CD4+ T cells. Blood model. Br J Clin Pharmacol 2015;80:1438–46.
2008;111:453–62. 37. Zimmerman JJ, Harper D, Getsy J, et al. Pharmacokinetic interac-
14. Battaglia M, Stabilini A, Migliavacca B, et al. Rapamycin promotes tions between sirolimus and microemulsion cyclosporine when
expansion of functional CD4+CD25+FOXP3+ regulatory T cells orally administered jointly and 4 hours apart in healthy volunteers.
of both healthy subjects and type 1 diabetic patients. J Immunol J Clin Pharmacol 2003;43:1168–76.
2006;177:8338–47. 38. Rostaing L, Christiaans MHL, Kovarik JM, et al. The pharmacoki-
15. Ruggenenti P, Perico N, Gotti E, et al. Sirolimus versus cyclosporine netics of everolimus in de novo kidney transplant patients receiv-
therapy increases circulating regulatory T cells, but does not pro- ing tacrolimus: an analysis from the randomized ASSET study. Ann
tect renal transplant patients given alemtuzumab induction from Transplant 2014;19:337–45.
chronic allograft injury. Transplantation 2007;84:956–64. 39. Baldan N, Rigotti P, Furian L, et al. Co-administration of sirolimus
16. Hester J, Schiopu A, Nadig SN, et al. Low-dose rapamycin treatment alters tacrolimus pharmacokinetics in a dose-dependent manner in
increases the ability of human regulatory T cells to inhibit transplant adult renal transplant recipients. Pharmacol Res 2006;54:181–5.
arteriosclerosis in vivo. Am J Transplant 2012;12:2008–16. 40. Pascual J, del Castillo D, Cabello M, et al. Interaction between evero-
17. Saemann MD, Haidinger M, Hecking M, et al. The multifunctional limus and tacrolimus in renal transplant recipients: a pharmacoki-
role of mTOR in innate immunity: implications for transplant immu- netic controlled trial. Transplantation 2010;89:994–1000.
nity. Am J Transplant 2009;9:2655–61. 41. Buchler M, Lebranchu Y, Beneton M, et al. Higher exposure to myco-
18. Wang C, Qin L, Manes TD, et al. Rapamycin antagonizes TNF induc- phenolic acid with sirolimus than with cyclosporine cotreatment.
tion of VCAM-1 on endothelial cells by inhibiting mTORC2. J Exp Clin Pharmacol Ther 2005;78:34–42.
Med 2014;211:395–404. 42. Jusko WJ, Ferron GM, Mis SM, et al. Pharmacokinetics of predniso-
19. Araki K, Ellebedy AH, Ahmed R. TOR in the immune system. Curr lone during administration of sirolimus in patients with renal trans-
Opin Cell Biol 2011;23:707–15. plants. J Clin Pharmacol 1996;36:1100–6.
20. Wang H, Zheng X, Cai Z, et al. Synthesis and evaluation of an inject- 43. Kahan BD, Gibbons S, Tejpal N, et al. Synergistic effect of the
able everolimus prodrug. Bioorg Med Chem Lett 2017;27:1175–8. rapamycin-cyclosporine combination: median effect analysis of
21. Yatscoff R, LeGatt D, Keenan R, et al. Blood distribution of rapamy- in vitro immune performances by human T lymphocytes in PHA,
cin. Transplantation 1993;56:1202–6. CD3, and MLR proliferative and cytotoxicity assays. Transplant Proc
22. Ferron GM, Mishina EV, Zimmerman JJ, et al. Population pharmaco- 1991;23:1090–1.
kinetics of sirolimus in kidney transplant patients. Clin Pharmacol 44. Kimball PM, Kerman RH, Kahan BD. Production of synergistic but
Ther 1997;61:416–28. nonidentical mechanisms of immunosuppression by rapamycin and
23. MacDonald A, Scarola J, Burke JT, et al. Clinical pharmacoki- cyclosporine. Transplantation 1991;51:486–90.
netics and therapeutic drug monitoring of sirolimus. Clin Ther 45. Andoh TF, Lindsley J, Franceschini N, et al. Synergistic effects of
2000;22(Suppl. B):B101–21. cyclosporine and rapamycin in a chronic nephrotoxicity model.
24. Kelly PA, Napoli K, Kahan BD. Conversion from liquid to solid Transplantation 1996;62:311–6.
rapamycin formulations in stable renal allograft transplant recipi- 46. Kahan BD, Kramer WG. Median effect analysis of efficacy ver-
ents. Biopharm Drug Dispos 1999;20:249–53. sus adverse effects of immunosuppressants. Clin Pharmacol Ther
25. Dirks NL, Huth B, Yates CR, et al. Pharmacokinetics of immunosup- 2001;70:74–81.
pressants: a perspective on ethnic differences. Int J Clin Pharmacol 47. Dumont FJ, Kastner C, Iacovone Jr F, et al. Quantitative and tempo-
Ther 2004;42:701–18. ral analysis of the cellular interaction of FK-506 and rapamycin in
26. Kahan BD, Wong RL, Carter C, et al. A phase I study of a 4-week T-lymphocytes. J Pharmacol Exp Ther 1994;268:32–41.
course of SDZ-RAD (RAD) quiescent cyclosporine-prednisone-treated 48. Vu MD, Qi S, Xu D, et al. Tacrolimus (FK506) and sirolimus (rapamy-
renal transplant recipients. Transplantation 1999;68:1100–6. cin) in combination are not antagonistic but produce extended graft
27. Kovarik JM, Kahan BD, Rajagopalan PR, et al. Population pharma- survival in cardiac transplantation in the rat. Transplantation
cokinetics and exposure-response relationships for basiliximab in 1997;64:1853–6.
kidney transplantation. The US Simulect Renal Transplant Study 49. Groth CG, Backman L, Morales JM, et al. Sirolimus (rapamycin)-
Group. Transplantation 1999;68:1288–94. based therapy in human renal transplantation: similar efficacy and
28. Thudium K, Gallo J, Bouillaud E, et al. Bioavailability of everolimus different toxicity compared with cyclosporine. Sirolimus European
administered as a single 5 mg tablet versus five 1 mg tablets: a ran- Renal Transplant Study Group. Transplantation 1999;67:1036–42.
domized, open-label, two-way crossover study of healthy volun- 50. Kreis H, Cisterne JM, Land W, et al. Sirolimus in association with myco-
teers. Clin Pharmacol 2015;7:11–7. phenolate mofetil induction for the prevention of acute graft rejection
29. Shipkova M, Hesselink DA, Holt DW, et al. Therapeutic drug in renal allograft recipients. Transplantation 2000;69:1252–60.
monitoring of everolimus: a consensus report. Ther Drug Monit 51. Morales JM, Wramner L, Kreis H, et al. Sirolimus does not exhibit
2016;38:143–69. nephrotoxicity compared to cyclosporine in renal transplant recipi-
30. Johnson-Davis KL, De S, Jimenez E, et al. Evaluation of the Abbott ents. Am J Transplant 2002;2:436–42.
ARCHITECT i2000 sirolimus assay and comparison with the Abbott 52. Flechner SM, Goldfarb D, Modlin C, et al. Kidney transplantation
IMx sirolimus assay and an established liquid chromatography-­tandem without calcineurin inhibitor drugs: a prospective, randomized trial
mass spectrometry method. Ther Drug Monit 2011;33:453–9. of sirolimus versus cyclosporine. Transplantation 2002;74:1070–6.
18 • mTOR Inhibitors: Sirolimus and Everolimus 279

53. Larson TS, Dean PG, Stegall MD, et al. Complete avoidance of calci- 74. Qazi Y, Shaffer D, Kaplan B, et al. Efficacy and safety of everoli-
neurin inhibitors in renal transplantation: a randomized trial com- mus plus low-dose tacrolimus versus mycophenolate mofetil plus
paring sirolimus and tacrolimus. Am J Transplant 2006;6:514–22. standard-dose tacrolimus in de novo renal transplant recipients:
54. Srinivas TR, Schold JD, Guerra G, et al. Mycophenolate mofetil/siro- 12-month data. Am J Transplant 2017;17:1358–69.
limus compared to other common immunosuppressive regimens in 75. Kidney Disease: Improving Global Outcomes (KDIGO) Transplant
kidney transplantation. Am J Transplant 2007;7:586–94. Work Group. KDIGO clinical practice guideline for the care of kidney
55. Webster AC, Lee VW, Chapman JR, et al. Target of rapamycin inhibi- transplant recipients. Am J Transplant 2009;9(Suppl. 3):S1–157.
tors (sirolimus and everolimus) for primary immunosuppression of 76. Johnson RW, Kreis H, Oberbauer R, et al. Sirolimus allows early
kidney transplant recipients: a systematic review and meta-analysis cyclosporine withdrawal in renal transplantation resulting in
of randomized trials. Transplantation 2006;81:1234–48. improved renal function and lower blood pressure. Transplantation
56. Flechner SM, Glyda M, Cockfield S, et al. The ORION study: com- 2001;72:777–86.
parison of two sirolimus-based regimens versus tacrolimus and 77. Kreis H, Oberbauer R, Campistol JM, et al. Long-term benefits with
mycophenolate mofetil in renal allograft recipients. Am J Trans- sirolimus-based therapy after early cyclosporine withdrawal. J Am
plant 2011;11:1633–44. [Erratum appears in Am J Transplant. Soc Nephrol 2004;15:809–17.
2012;12(1):268.]. 78. Oberbauer R, Segoloni G, Campistol JM, et al. Early cyclosporine
57. Nankivell BJ, Gruenewald SM, Allen RD, et al. Predicting glomeru- withdrawal from a sirolimus-based regimen results in better renal
lar filtration rate after kidney transplantation. Transplantation allograft survival and renal function at 48 months after transplanta-
1995;59:1683–9. tion. Transpl Int 2005;18:22–8.
58. Ekberg H, Tedesco-Silva H, Demirbas A, et al. Reduced exposure 79. Watson CJ, Firth J, Williams PF, et al. A randomized controlled
to calcineurin inhibitors in renal transplantation. N Engl J Med trial of late conversion from CNI-based to sirolimus-based immu-
2007;357:2562–75. nosuppression following renal transplantation. Am J Transplant
59. Ekberg H, Bernasconi C, Tedesco-Silva H, et al. Calcineurin inhibitor 2005;5:2496–503.
minimization in the Symphony study: observational results 3 years 80. Diekmann F, Budde K, Oppenheimer F, et al. Predictors of success
after transplantation. Am J Transplant 2009;9:1876–85. in conversion from calcineurin inhibitor to sirolimus in chronic
60. Gatault P, Bertrand D, Buchler M, et al. Eight-year results of the allograft dysfunction. Am J Transplant 2004;4:1869–75.
Spiesser study, a randomized trial comparing de novo sirolimus and 81. Sundberg AK, Rohr MS, Hartmann EL, et al. Conversion to siroli-
cyclosporine in renal transplantation. Transpl Int 2016;29:41–50. mus-based maintenance immunosuppression using daclizumab
61. Kahan BD. Efficacy of sirolimus compared with azathioprine for reduc- bridge therapy in renal transplant recipients. Clin Transplant
tion of acute renal allograft rejection: a randomised multicentre study. 2004;18(Suppl. 12):61–6.
The Rapamune US Study Group. Lancet 2000;356:194–202. 82. Letavernier E, Pe’raldi MN, Pariente A, et al. Proteinuria following
62. MacDonald AS. A worldwide, phase III, randomized, controlled, a switch from calcineurin inhibitors to sirolimus. Transplantation
safety and efficacy study of a sirolimus/cyclosporine regimen for pre- 2005;80:1198–203.
vention of acute rejection in recipients of primary mismatched renal 83. Diekmann F, Campistol JM. Conversion from calcineurin inhibitors
allografts. Transplantation 2001;71:271–80. to sirolimus in chronic allograft nephropathy: benefits and risks.
63. Podder H, Stepkowski SM, Napoli KL, et al. Pharmacokinetic inter- Nephrol Dial Transplant 2006;21:562–8.
actions augment toxicities of sirolimus/cyclosporine combinations. 84. Barone GW, Gurley BJ, Abul-Ezz SR, et al. Sirolimus-induced throm-
J Am Soc Nephrol 2001;12:1059–71. botic microangiopathy in a renal transplant recipient. Am J Kidney
64. Vitko S, Margreiter R, Weimar W, et al. Three-year efficacy and safety Dis 2003;42:202–6.
results from a study of everolimus versus mycophenolate mofetil in 85. Sartelet H, Toupance O, Lorenzato M, et al. Sirolimus-induced
de novo renal transplant patients. Am J Transplant 2005;5:2521– thrombotic microangiopathy is associated with decreased expres-
30. sion of vascular endothelial growth factor in kidneys. Am J Trans-
65. Vitko S, Margreiter R, Weimar W, et al. Everolimus (Certican) plant 2005;5:2441–7.
12-month safety and efficacy versus mycophenolate mofetil in de 86. The 3C Study Collaborative Group. Alemtuzumab-based induc-
novo renal transplant recipients. Transplantation 2004;78:1532– tion treatment versus basiliximab-based induction treatment in
40. kidney transplantation (the 3C Study): a randomised trial. Lancet
66. Lorber MI, Mulgaonkar S, Butt KM, et al. Everolimus versus myco- 2014;384:1684–90.
phenolate mofetil in the prevention of rejection in de novo renal 87. Haynes R, Blackwell L, Staplin N, et al. Campath, calcineurin inhibi-
transplant recipients: a 3-year randomized, multicenter, phase III tor reduction and chronic allograft nephropathy (the 3C Study)—
study. Transplantation 2005;80:244–52. results of a randomized controlled clinical trial. Am J Transplant
67. Nashan B, Curtis J, Ponticelli C, et al. Everolimus and reduced-expo- 2018;18:1424–34.
sure cyclosporine in de novo renal-transplant recipients: a three-year 88. Budde K, Becker T, Arns W, et al. Everolimus-based, calcineurin-
phase II, randomized, multicenter, open-label study. Transplanta- inhibitor-free regimen in recipients of de-novo kidney transplants:
tion 2004;78:1332–40. an open-label, randomised, controlled trial. Lancet 2011;377:837–
68. McAlister VC, Gao Z, Peltekian K, et al. Sirolimus-tacrolimus combi- 47.
nation immunosuppression. Lancet 2000;355:376–7. 89. Budde K, Lehner F, Sommerer C, et al. Five-year outcomes in kidney
69. Mendez R, Gonwa T, Yang HC, et al. A prospective, randomized transplant patients converted from cyclosporine to everolimus: the
trial of tacrolimus in combination with sirolimus or mycophenolate randomized zeus study. Am J Transplant 2015;15:119–28.
mofetil in kidney transplantation: results at 1 year. Transplantation 90. de Fijter JW, Holdaas H, Oyen O, et al. Early conversion from cal-
2005;80:303–9. cineurin inhibitor- to everolimus-based therapy following kidney
70. Van Gurp E, Bustamante J, Franco A, et al. Comparable renal func- transplantation: results of the randomized ELEVATE trial. Am J
tion at 6 months with tacrolimus combined with fixed-dose siro- Transplant 2017;17:1853–67.
limus or MMF: results of a randomized multicenter trial in renal 91. LoPiccolo J, Blumenthal GM, Bernstein WB, et al. Targeting the
transplantation. J Transplant 2010;2010:731426. Available online PI3K/Akt/mTOR pathway: effective combinations and clinical con-
at: https://doi.org/10.1155/2010/731426. siderations. Drug Resist Updat 2008;11:32–50.
71. Rummo OO, Carmellini M, Rostaing L, et al. ADHERE: randomized 92. Baselga J, Campone M, Piccart M, et al. Everolimus in postmeno-
controlled trial comparing renal function in de novo kidney trans- pausal hormone-receptor-positive advanced breast cancer. N Engl J
plant recipients receiving prolonged-release tacrolimus plus MMF or Med 2012;366:520–9.
sirolimus. Transplant Int 2016;30:83–95. 93. Duzgun Z, Eroglu Z, Biray Avci C. Role of mTOR in glioblastoma.
72. Chan L, Greenstein S, Hardy MA, et al. Multicenter, random- Gene 2016;575:187–90.
ized study of the use of everolimus with tacrolimus after renal 94. Pal SK, Quinn DI. Differentiating mTOR inhibitors in renal cell carci-
transplantation demonstrates its effectiveness. Transplantation noma. Cancer Treat Rev 2013;39:709–19.
2008;85:821–6. 95. Panzuto F, Rinzivillo M, Fazio N, et al. Real-world study of everoli-
73. Langer RM, Hene R, Vitko S, et al. Everolimus plus early tacrolimus mus in advanced progressive neuroendocrine tumors. Oncologist
minimization: a phase III, randomized, open-label, multicentre trial 2014;19:966–74. [Erratum appears in Oncologist. 2015;20(5):570.
in renal transplantation. Transpl Int 2012;25:592–602. PMID:25956900.]
280 Kidney Transplantation: Principles and Practice

96. Kauffman HM, Cherikh WS, Cheng Y, et al. Maintenance immu- 118. Wali RK, Drachenberg C, Hirsch HH, et al. BK virus-associated
nosuppression with target-of-rapamycin inhibitors is associated nephropathy in renal allograft recipients: rescue therapy by sirolimus-
with a reduced incidence of de novo malignancies. Transplantation based immunosuppression. Transplantation 2004;78:1069–73.
2005;80:883–9. 119. Hirsch HH, Yakhontova K, Lu M, et al. BK polyomavirus replication
97. Campistol JM, Eris J, Oberbauer R, et al. Sirolimus therapy after early in renal tubular epithelial cells is inhibited by sirolimus, but acti-
cyclosporine withdrawal reduces the risk for cancer in adult renal vated by tacrolimus through a pathway involving FKBP-12. Am J
transplantation. J Am Soc Nephrol 2006;17:581–9. Transplant 2016;16:821–32.
98. Yanik EL, Siddiqui K, Engels EA. Sirolimus effects on cancer inci- 120. Liacini A, Seamone ME, Muruve DA, et al. Anti-BK virus mecha-
dence after kidney transplantation: a meta-analysis. Cancer Med nisms of sirolimus and leflunomide alone and in combination:
2015;4:1448–59. toward a new therapy for BK virus infection. Transplantation
99. Knoll GA, Kokolo MB, Mallick R, et al. Effect of sirolimus on malig- 2010;90:1450–7.
nancy and survival after kidney transplantation: systematic review 121. Jaw J, Hill P, Goodman D. Combination of leflunomide and everolimus
and meta-analysis of individual patient data. BMJ 2014;349:g6679. for treatment of BK virus nephropathy. Nephrology 2017;22:326–9.
100. Euvrard S, Kanitakis J, Claudy A. Skin cancers after organ transplan- 122. Harris PC, Torres VE. Polycystic kidney disease. Annu Rev Med
tation. N Engl J Med 2003;348:1681–91. 2009;60:321–37.
101. Salgo R, Gossmann J, Schofer H, et al. Switch to a sirolimus-based 123. Novalic Z, van der Wal AM, Leonhard WN, et al. Dose-dependent
immunosuppression in long-term renal transplant recipients: effects of sirolimus on mTOR signaling and polycystic kidney disease.
reduced rate of (pre-)malignancies and nonmelanoma skin cancer in J Am Soc Nephrol 2012;23:842–53.
a prospective, randomized, assessor-blinded, controlled clinical trial. 124. Shillingford JM, Piontek KB, Germino GG, et al. Rapamycin amelio-
Am J Transplant 2010;10:1385–93. rates PKD resulting from conditional inactivation of Pkd1. J Am Soc
102. Euvrard S, Morelon E, Rostaing L, et al. Sirolimus and secondary Nephrol 2010;21:489–97.
skin-cancer prevention in kidney transplantation. N Engl J Med 125. Wu M, Wahl PR, Le Hir M, et al. Everolimus retards cyst growth and
2012;367:329–39. preserves kidney function in a rodent model for polycystic kidney dis-
103. Majewski M, Korecka M, Joergensen J, et al. Immunosuppres- ease. Kidney Blood Press Res 2007;30:253–9.
sive TOR kinase inhibitor everolimus (RAD) suppresses growth of 126. Qian Q, Du H, King BF, et al. Sirolimus reduces polycystic liver vol-
cells derived from posttransplant lymphoproliferative disorder at ume in ADPKD patients. J Am Soc Nephrol 2008;19:631–8.
allograft-protecting doses. Transplantation 2003;75:1710–7. 127. Serra AL, Poster D, Kistler AD, et al. Sirolimus and kidney growth
104. Majewski M, Korecka M, Kossev P, et al. The immunosuppressive in autosomal dominant polycystic kidney disease. N Engl J Med
macrolide RAD inhibits growth of human Epstein-Barr virus-trans- 2010;363:820–9.
formed B lymphocytes in vitro and in vivo: a potential approach to 128. Walz G, Budde K, Mannaa M, et al. Everolimus in patients with
prevention and treatment of posttransplant lymphoproliferative dis- autosomal dominant polycystic kidney disease. N Engl J Med
orders. Proc Natl Acad Sci USA 2000;97:4285–90. 2010;363:830–40.
105. Nepomuceno RR, Balatoni CE, Natkunam Y, et al. Rapamycin inhib- 129. Liu Y, Pejchinovski M, Wang X, et al. Dual mTOR/PI3K inhibition
its the interleukin 10 signal transduction pathway and the growth limits PI3K-dependent pathways activated upon mTOR inhibition in
of Epstein Barr virus B-cell lymphomas. Cancer Res 2003;63:4472– autosomal dominant polycystic kidney disease. Sci Rep 2018;8:5584.
80. 130. Murakami N, Riella LV, Funakoshi T. Risk of metabolic complica-
106. Cullis B, D’Souza R, McCullagh P, et al. Sirolimus-induced remission tions in kidney transplantation after conversion to mTOR inhibi-
of posttransplantation lymphoproliferative disorder. Am J Kidney Dis tor: a systematic review and meta-analysis. Am J Transplant
2006;47:e67–72. 2014;14:2317–27.
107. Pascual J. Post-transplant lymphoproliferative disorder—the 131. Johnston O, Rose CL, Webster AC, et al. Sirolimus is associated
potential of proliferation signal inhibitors. Nephrol Dial Transplant with new-onset diabetes in kidney transplant recipients. J Am Soc
2007;22(Suppl. 1):i27–35. Nephrol 2008;19:1411–8.
108. Caillard S, Dharnidharka V, Agodoa L, et al. Posttransplant lym- 132. Goldman JW, Mendenhall MA, Rettinger SR. Hyperglycemia associ-
phoproliferative disorders after renal transplantation in the United ated with targeted oncologic treatment: mechanisms and manage-
States in era of modern immunosuppression. Transplantation ment. Oncologist 2016;29:29.
2005;80:1233–43. 133. Bussiere CT, Lakey JR, Shapiro AM, et al. The impact of the mTOR
109. Stallone G, Schena A, Infante B, et al. Sirolimus for Kaposi’s sarcoma inhibitor sirolimus on the proliferation and function of pancreatic
in renal-transplant recipients. N Engl J Med 352:1317-1323. islets and ductal cells. Diabetologia 2006;49:2341–9.
110. Lebbe C, Euvrard S, Barrou B, et al. Sirolimus conversion for 134. Guo S. Insulin signaling, resistance, and the metabolic syndrome:
patients with posttransplant Kaposi’s sarcoma. Am J Transplant insights from mouse models into disease mechanisms. J Endocrinol
2006;6:2164–8. 2014;220:T1–23.
111. Basu G, Mohapatra A, Manipadam MT, et al. Leflunomide with 135. Laplante M, Sabatini DM. mTOR signaling in growth control and dis-
low-dose everolimus for treatment of Kaposi’s sarcoma in a renal ease. Cell 2012;149:274–93.
allograft recipient. Nephrol Dial Transplant 2011;26:3412–5. 136. Teutonico A, Schena PF, Di Paolo S. Glucose metabolism in renal
112. Babel N, Eibl N, Ulrich C, et al. Development of Kaposi’s sarcoma transplant recipients: effect of calcineurin inhibitor withdrawal and
under sirolimus-based immunosuppression and successful treat- conversion to sirolimus. J Am Soc Nephrol 2005;16:3128–35.
ment with imiquimod. Transpl Infect Dis 2008;10:59–62. 137. Caron A, Richard D, Laplante M. The roles of mTOR complexes in
113. Benavides CA, Pollard VB, Mauiyyedi S, et al. BK virus-associated lipid metabolism. Annu Rev Nutr 2015;35:321–48.
nephropathy in sirolimus-treated renal transplant patients: inci- 138. Flechner SM, Gurkan A, Hartmann A, et al. A randomized, open-
dence, course, and clinical outcomes. Transplantation 2007;84:83– label study of sirolimus versus cyclosporine in primary de novo renal
8. allograft recipients. Transplantation 2013;95:1233–41.
114. Tohme FA, Kalil RS, Thomas CP. Conversion to a sirolimus-based 139. Ikonen TS, Gummert JF, Hayase M, et al. Sirolimus (rapamycin)
regimen is associated with lower incidence of BK viremia in low-risk halts and reverses progression of allograft vascular disease in non-
kidney transplant recipients. Transpl Infect Dis 2015;17:66–72. human primates. Transplantation 2000;70:969–75.
115. Radtke J, Dietze N, Fischer L, et al. Incidence of BK polyomavirus 140. Eisen HJ, Tuzcu EM, Dorent R, et al. Everolimus for the prevention
infection after kidney transplantation is independent of type of of allograft rejection and vasculopathy in cardiac-transplant recipi-
immunosuppressive therapy. Transpl Infect Dis 2016;18:850–5. ents. N Engl J Med 2003;349:847–58.
116. Belliere J, Kamar N, Mengelle C, et al. Pilot conversion trial from 141. Elloso MM, Azrolan N, Sehgal SN, et al. Protective effect of the immu-
mycophenolic acid to everolimus in ABO-incompatible kidney- nosuppressant sirolimus against aortic atherosclerosis in apo E-defi-
transplant recipients with BK viruria and/or viremia. Transpl Int cient mice. Am J Transplant 2003;3:562–9.
2016;29:315–22. 142. Maluf DG, Mas VR, Archer KJ, et al. Apolipoprotein E genotypes as
117. Polanco N, Gonzalez Monte E, Folgueira MD, et al. Everolimus-based predictors of high-risk groups for developing hyperlipidemia in kid-
immunosuppression therapy for BK virus nephropathy. Transplant ney transplant recipients undergoing sirolimus treatment. Trans-
Proc 2015;47:57–61. plantation 2005;80:1705–11.
18 • mTOR Inhibitors: Sirolimus and Everolimus 281

143. Champion L, Stern M, Israel-Biet D, et al. Brief communication: siro- 165. Senior PA, Paty BW, Cockfield SM, et al. Proteinuria developing after
limus-associated pneumonitis: 24 cases in renal transplant recipi- clinical islet transplantation resolves with sirolimus withdrawal and
ents. Ann Intern Med 2006;144:505–9. increased tacrolimus dosing. Am J Transplant 2005;5:2318–23.
144. Haydar AA, Denton M, West A, et al. Sirolimus-induced pneumo- 166. Cina DP, Onay T, Paltoo A, et al. Inhibition of mTOR disrupts
nitis: three cases and a review of the literature. Am J Transplant autophagic flux in podocytes. J Am Soc Nephrol 2012;23:412–
2004;4:137–9. 20.
145. Solazzo A, Botta C, Nava F, et al. Interstitial lung disease after kid- 167. Letavernier E, Bruneval P, Vandermeersch S, et al. Sirolimus inter-
ney transplantation and the role of everolimus. Transplant Proc acts with pathways essential for podocyte integrity. Nephrol Dial
2016;48:349–51. Transplant 2009;24:630–8.
146. Willemsen AE, Grutters JC, Gerritsen WR, et al. mTOR inhibitor- 168. Vuiblet V, Birembaut P, Francois A, et al. Sirolimus-based regimen is
induced interstitial lung disease in cancer patients: comprehen- associated with decreased expression of glomerular vascular endo-
sive review and a practical management algorithm. Int J Cancer thelial growth factor. Nephrol Dial Transplant 2012;27:411–6.
2016;138:2312–21. 169. Oroszlan M, Bieri M, Ligeti N, et al. Sirolimus and everolimus reduce
147. Morelon E, Stern M, Kreis H. Interstitial pneumonitis associated albumin endocytosis in proximal tubule cells via an angiotensin II-
with sirolimus therapy in renal-transplant recipients. N Engl J Med dependent pathway. Transpl Immunol 2010;23:125–32.
2000;343:225–6. 170. Fuller TF, Freise CE, Serkova N, et al. Sirolimus delays recovery of rat
148. Singer SJ, Tiernan R, Sullivan EJ. Interstitial pneumonitis associated kidney transplants after ischemia-reperfusion injury. Transplanta-
with sirolimus therapy in renal-transplant recipients. N Engl J Med tion 2003;76:1594–9.
2000;343:1815–6. 171. McTaggart RA, Gottlieb D, Brooks J, et al. Sirolimus prolongs recov-
149. Kahan BD, Julian BA, Pescovitz MD, et al. Sirolimus reduces the inci- ery from delayed graft function after cadaveric renal transplanta-
dence of acute rejection episodes despite lower cyclosporine doses in tion. Am J Transplant 2003;3:416–23.
caucasian recipients of mismatched primary renal allografts: a phase 172. Simon JF, Swanson SJ, Agodoa LY, et al. Induction sirolimus and
II trial. Rapamune Study Group. Transplantation 1999;68:1526– delayed graft function after deceased donor kidney transplantation
32. in the United States. Am J Nephrol 2004;24:393–401.
150. David S, Kumpers P, Shin H, et al. Everolimus-associated intersti- 173. Smith KD, Wrenshall LE, Nicosia RF, et al. Delayed graft function
tial pneumonitis in a patient with a heart transplant. Nephrol Dial and cast nephropathy associated with tacrolimus plus rapamycin
Transplant 2007;22:3363–4. use. J Am Soc Nephrol 2003;14:1037–45.
151. Saito Y, Gemma A. Current status of DILD in molecular targeted 174. Dantal J, Berthoux F, Moal MC, et al. Efficacy and safety of de
therapies. Int J Clin Oncol 2012;17:534–41. novo or early everolimus with low cyclosporine in deceased-
152. Garrean S, Massad MG, Tshibaka M, et al. Sirolimus-associated donor kidney transplant recipients at specified risk of delayed graft
interstitial pneumonitis in solid organ transplant recipients. Clin function: 12-month results of a randomized, multicenter trial.
Transplant 2005;19:698–703. Transpl Int 2010;23:1084–93. [Erratum appears in Transpl Int.
153. Langer RM, Van Buren CT, Katz SM, et al. De novo hemolytic uremic 2012;25(1):138.]
syndrome after kidney transplantation in patients treated with cyclo- 175. Loverre A, Ditonno P, Crovace A, et al. Ischemia-reperfusion induces
sporine-sirolimus combination. Transplantation 2002;73:756–60. glomerular and tubular activation of proinflammatory and anti-
154. Robson M, Cote I, Abbs I, et al. Thrombotic micro-angiopathy apoptotic pathways: differential modulation by rapamycin. J Am Soc
with sirolimus-based immunosuppression: potentiation of calci- Nephrol 2004;15:2675–86.
neurin-inhibitor-induced endothelial damage? Am J Transplant 176. Izzedine H, Escudier B, Rouvier P, et al. Acute tubular necrosis asso-
2003;3:324–7. ciated with mTOR inhibitor therapy: a real entity biopsy-proven.
155. Hachem RR, Yusen RD, Chakinala MM, et al. Thrombotic microan- Ann Oncol 2013;24:2421–5.
giopathy after lung transplantation. Transplantation 2006;81:57– 177. McTaggart RA, Tomlanovich S, Bostrom A, et al. Comparison of out-
63. comes after delayed graft function: sirolimus-based versus other cal-
156. Lovric S, Kielstein JT, Kayser D, et al. Combination of everolimus cineurin-inhibitor sparing induction immunosuppression regimens.
with calcineurin inhibitor medication resulted in post-transplant Transplantation 2004;78:475–80.
haemolytic uraemic syndrome in lung transplant recipients—a case 178. Stallone G, Di Paolo S, Schena A, et al. Addition of sirolimus to cyclo-
series. Nephrol Dial Transplant 2011;26:3032–8. sporine delays the recovery from delayed graft function but does not
157. Diekmann F, Andres A, Oppenheimer F. mTOR inhibitor-associ- affect 1-year graft function. J Am Soc Nephrol 2004;15:228–33.
ated proteinuria in kidney transplant recipients. Transplant Rev 179. Aboujaoude W, Milgrom ML, Govani MV. Lymphedema associ-
(Orlando) 2012;26:27–9. ated with sirolimus in renal transplant recipients. Transplantation
158. Ruiz JC, Diekmann F, Campistol JM, et al. Evolution of proteinuria 2004;77:1094–6.
after conversion from calcineurin inhibitors (CNI) to sirolimus (SRL) 180. Fuchs U, Zittermann A, Berthold HK, et al. Immunosuppressive ther-
in renal transplant patients: a multicenter study. Transplant Proc apy with everolimus can be associated with potentially life-threaten-
2005;37:3833–5. ing lingual angioedema. Transplantation 2005;79:981–3.
159. Bumbea V, Kamar N, Ribes D, et al. Long-term results in renal 181. Mohaupt MG, Vogt B, Frey FJ. Sirolimus-associated eyelid edema in
transplant patients with allograft dysfunction after switching kidney transplant recipients. Transplantation 2001;72:162–4.
from calcineurin inhibitors to sirolimus. Nephrol Dial Transplant 182. Stallone G, Infante B, Di Paolo S, et al. Sirolimus and angiotensin-con-
2005;20:2517–23. verting enzyme inhibitors together induce tongue oedema in renal
160. Diekmann F, Fritsche L, Neumayer HH, et al. Sirolimus dosage dur- transplant recipients. Nephrol Dial Transplant 2004;19:2906–8.
ing and after conversion from calcineurin inhibitor therapy to siro- 183. Wadei H, Gruber SA, El-Amm JM, et al. Sirolimus-induced angio-
limus in chronic kidney transplant patients. Kidney Blood Press Res edema. Am J Transplant 2004;4:1002–5.
2004;27:186–90. 184. Moro JA, Almenar L, Martinez-Dolz L, et al. mTOR inhibitors and
161. Faul C, Donnelly M, Merscher-Gomez S, et al. The actin cytoskeleton unilateral edema. Rev Esp Cardiol 2008;61:987–8.
of kidney podocytes is a direct target of the antiproteinuric effect of 185. Gharbi C, Gueutin V, Izzedine H. Oedema, solid organ transplanta-
cyclosporine A. Nat Med 2008;14:931–8. tion and mammalian target of rapamycin inhibitor/proliferation sig-
162. Dittrich E, Schmaldienst S, Soleiman A, et al. Rapamycin-associ- nal inhibitors (mTOR-I/PSIs). Clin Kidney J 2014;7:115–20.
ated post-transplantation glomerulonephritis and its remission 186. Dean PG, Lund WJ, Larson TS, et al. Wound-healing complications
after reintroduction of calcineurin-inhibitor therapy. Transpl Int after kidney transplantation: a prospective, randomized comparison
2004;17:215–20. of sirolimus and tacrolimus. Transplantation 2004;77:1555–61.
163. Dervaux T, Caillard S, Meyer C, et al. Is sirolimus responsible for pro- 187. Dutly AE, Gaspert A, Inci I, et al. The influence of the rapamycin-
teinuria? Transplant Proc 2005;37:2828–9. derivate SDZ RAD on the healing of airway anastomoses. Eur J Car-
164. Saurina A, Campistol JM, Piera C, et al. Conversion from calcineurin diothorac Surg 2003;24:154–8; discussion 8.
inhibitors to sirolimus in chronic allograft dysfunction: changes in 188. King-Biggs MB, Dunitz JM, Park SJ, et al. Airway anastomotic dehis-
glomerular haemodynamics and proteinuria. Nephrol Dial Trans- cence associated with use of sirolimus immediately after lung trans-
plant 2006;21:488–93. plantation. Transplantation 2003;75:1437–43.
282 Kidney Transplantation: Principles and Practice

189. Kahn D, Spearman CW, Mall A, et al. The effect of rapamycin on the 210. Ma Q, Zhou Y, Nie X, et al. Rapamycin affects tissue plasminogen
healing of the ureteric anastomosis and wound healing. Transplant activator and plasminogen activator inhibitor I expression: a poten-
Proc 2005;37:830–1. tial prothrombotic mechanism of drug-eluting stents. Angiology
190. Nair RV, Huang X, Shorthouse R, et al. Antiproliferative effect of 2012;63:330–5.
rapamycin on growth factor-stimulated human adult lung fibro- 211. Morales JM, Andres A, Dominguez-Gil B, et al. Tubular function in
blasts in vitro may explain its superior efficacy for prevention and patients with hypokalemia induced by sirolimus after renal trans-
treatment of allograft obliterative airway disease in vivo. Transplant plantation. Transplant Proc 2003;35:154S–6S.
Proc 1997;29:614–5. 212. Schwarz C, Bohmig GA, Steininger R, et al. Impaired phosphate han-
191. Pengel LH, Liu LQ, Morris PJ. Do wound complications or lympho- dling of renal allografts is aggravated under rapamycin-based immu-
celes occur more often in solid organ transplant recipients on mTOR nosuppression. Nephrol Dial Transplant 2001;16:378–82.
inhibitors? A systematic review of randomized controlled trials. 213. Elder GJ. From marrow oedema to osteonecrosis: common paths in
Transplant Int 2011;24:1216–30. the development of post-transplant bone pain. Nephrology (Carlton)
192. Nashan B, Citterio F. Wound healing complications and the use 2006;11:560–7.
of mammalian target of rapamycin inhibitors in kidney trans- 214. Romero DF, Buchinsky FJ, Rucinski B, et al. Rapamycin: a bone spar-
plantation: a critical review of the literature. Transplantation ing immunosuppressant? J Bone Miner Res 1995;10:760–8.
2012;94:547–61. 215. Kneissel M, Luong-Nguyen NH, Baptist M, et al. Everolimus sup-
193. Tiong HY, Flechner SM, Zhou L, et al. A systematic approach to presses cancellous bone loss, bone resorption, and cathepsin K
minimizing wound problems for de novo sirolimus-treated kidney expression by osteoclasts. Bone 2004;35:1144–56.
transplant recipients. Transplantation 2009;87:296–302. 216. Campistol JM, Holt DW, Epstein S, et al. Bone metabolism in renal
194. Sonis S, Treister N, Chawla S, et al. Preliminary characterization transplant patients treated with cyclosporine or sirolimus. Transpl
of oral lesions associated with inhibitors of mammalian target of Int 2005;18:1028–35.
rapamycin in cancer patients. Cancer 2010;116:210–5. 217. Gregorini M, Sileno G, Pattonieri EF, et al. Understanding bone dam-
195. Chuang P, Langone AJ. Clobetasol ameliorates aphthous ulcer- age after kidney transplantation: a retrospective monocentric cross
ation in renal transplant patients on sirolimus. Am J Transplant sectional analysis. Transplant Proc 2017;49:650–7.
2007;7:714–7. 218. Rubert M, Montero M, Guede D, et al. Sirolimus and tacrolimus
196. Ropert S, Coriat R, Verret B, et al. Colchicine is an active treatment rather than cyclosporine A cause bone loss in healthy adult male
for everolimus-induced oral ulcers. Eur J Cancer 2017;87:209–11. rats. Bone Rep 2015;2:74–81.
197. Mahe E, Morelon E, Lechaton S, et al. Acne in recipients of renal 219. Luo D, Ren H, Li T, et al. Rapamycin reduces severity of senile
transplantation treated with sirolimus: clinical, microbiologic, his- osteoporosis by activating osteocyte autophagy. Osteoporos Int
tologic, therapeutic, and pathogenic aspects. J Am Acad Dermatol 2016;27:1093–101.
2006;55:139–42. 220. Jacques J, Dickson Z, Carrier P, et al. Severe sirolimus-induced acute
198. Ramirez-Fort MK, Case EC, Rosen AC, et al. Rash to the mTOR inhib- hepatitis in a renal transplant recipient. Transpl Int 2010;23:967–
itor everolimus: systematic review and meta-analysis. Am J Clin 70.
Oncol 2014;37:266–271. 221. Niemczyk M, Wyzgal J, Perkowska A, et al. Sirolimus-associated hep-
199. Shameem R, Lacouture M, Wu S. Incidence and risk of rash to mTOR atotoxicity in the kidney graft recipient. Transpl Int 2005;18:1302–
inhibitors in cancer patients—a meta-analysis of randomized con- 3.
trolled trials. Acta Oncol 2015;54:124–32. 222. Neff GW, Ruiz P, Madariaga JR, et al. Sirolimus-associated hepatotox-
200. Mahe E, Morelon E, Lechaton S, et al. Cutaneous adverse events icity in liver transplantation. Ann Pharmacother 2004;38:1593–6.
in renal transplant recipients receiving sirolimus-based therapy. 223. Braun M, Young J, Reiner CS, et al. Ovarian toxicity from sirolimus.
Transplantation 2005;79:476–82. N Engl J Med 2012;366:1062–4.
201. Afzali B, Al-Khoury S, Shah N, et al. Anemia after renal transplanta- 224. Kawaguchi Y, Maruno A, Kawashima Y, et al. Amenorrhea as a rare
tion. Am J Kidney Dis 2006;48:519–36. drug-related adverse event associated with everolimus for pancreatic
202. Quesniaux VF, Wehrli S, Steiner C, et al. The immunosuppressant neuroendocrine tumors. World J Gastroenterol 2014;20:15920–4.
rapamycin blocks in vitro responses to hematopoietic cytokines and 225. Boobes Y, Bernieh B, Saadi H, et al. Gonadal dysfunction and infer-
inhibits recovering but not steady-state hematopoiesis in vivo. Blood tility in kidney transplant patients receiving sirolimus. Int Urol
1994;84:1543–52. Nephrol 2010;42:493–8.
203. Maiorano A, Stallone G, Schena A, et al. Sirolimus interferes with 226. Zuber J, Anglicheau D, Elie C, et al. Sirolimus may reduce fertility in
iron homeostasis in renal transplant recipients. Transplantation male renal transplant recipients. Am J Transplant 2008;8:1471–9.
2006;82:908–12. 227. Fritsche L, Budde K, Dragun D, et al. Testosterone concentrations
204. Babinska A, Markell MS, Salifu MO, et al. Enhancement of human and sirolimus in male renal transplant patients. Am J Transplant
platelet aggregation and secretion induced by rapamycin. Nephrol 2004;4:130–1.
Dial Transplant 1998;13:3153–9. 228. Huyghe E, Zairi A, Nohra J, et al. Gonadal impact of target of rapamy-
205. Hong JC, Kahan BD. Sirolimus-induced thrombocytopenia and leu- cin inhibitors (sirolimus and everolimus) in male patients: an over-
kopenia in renal transplant recipients: risk factors, incidence, pro- view. Transpl Int 2007;20:305–11.
gression, and management. Transplantation 2000;69:2085–90. 229. Billing H, Burmeister G, Plotnicki L, et al. Longitudinal growth on
206. Flechner SM, Feng J, Mastroianni B, et al. The effect of 2-gram versus an everolimus- versus an MMF-based steroid-free immunosuppres-
1-gram concentration controlled mycophenolate mofetil on renal sive regimen in paediatric renal transplant recipients. Transpl Int
transplant outcomes using sirolimus-based calcineurin inhibitor 2013;26:903–9.
drug-free immunosuppression. Transplantation 2005;79:926–34. 230. Cavanaugh TM, Schoenemen H, Goebel J. The impact of sirolimus on
207. Asrani SK, Wiesner RH, Trotter JF, et al. De novo sirolimus and sex hormones in male adolescent kidney recipients. Pediatr Trans-
reduced-dose tacrolimus versus standard-dose tacrolimus after liver plant 2012;16:280–5.
transplantation: the 2000-2003 phase II prospective randomized 231. Forster J, Ahlenstiel-Grunow T, Zapf A, et al. Pubertal development
trial. Am J Transplant 2014;14:356–66. in pediatric kidney transplant patients receiving mammalian tar-
208. Palmerini T, Biondi-Zoccai G, Della Riva D, et al. Stent thrombosis get of rapamycin inhibitors or conventional immunosuppression.
with drug-eluting and bare-metal stents: evidence from a compre- Transplantation 2016;100:2461–70.
hensive network meta-analysis. Lancet 2012;379:1393–402. 232. Isakova T, Xie H, Messinger S, et al. Inhibitors of mTOR and risks
209. Langer RM, Kahan BD. Sirolimus does not increase the risk for post- of allograft failure and mortality in kidney transplantation. Am J
operative thromboembolic events among renal transplant recipi- Transplant 2013;13(1):100–10. Available online at: https://doi.
ents. Transplantation 2003;76:318–23. org/10.1111/j.1600-6143.2012.04281.x.
19 Antilymphocyte Globulin,
Monoclonal Antibodies, and
Fusion Proteins
EILEEN T. CHAMBERS and ALLAN D. KIRK

CHAPTER OUTLINE Historical Perspective Rituximab (Humanized Anti-CD20)


Antibody Structure and Function Induction
Fusion Protein Structure and Function Rescue
General Clinical Considerations for the Administration and Adverse Effects
Use of Antibody and Fusion Protein Belatacept
Preparations Maintenance
Polyclonal Antibody Preparations Administration and Adverse Effects
Specific Clinical Applications of Polyclonal Monoclonal Antibodies and Fusion
Antibody Preparations Proteins in Clinical Transplantation
Induction Investigation
Rescue CD2-Specific Approaches
Administration and Adverse Effects CD3-Specific Antibodies
Monoclonal Antibody and Fusion Protein CD4-Specific Antibodies
Preparations Costimulation-Based Therapies
Monoclonal Antibodies and Fusion Cytokine-Based Approaches
Proteins in Clinical Transplantation
Practice Targeting Cell Adhesion
Muromonab (OKT3; Murine Anti-CD3) Targeting the T Cell Receptor
Induction Targeting Complement
Rescue Other Experimental Antibodies and
Fusion Proteins
Interleukin-2 Receptor (CD25)-Specific
Monoclonal Antibodies Targeting CD5
Induction Targeting CD6
Administration and Adverse Effects Targeting CD7
Alemtuzumab (Humanized Anti-CD52) Targeting CD8
Induction Targeting CD45
Rescue Immunotoxins
Administration and Adverse Effects Conclusion

Renal transplantation is the preferred treatment for most with unambiguous specificity. Although they may medi-
end-stage renal diseases. Transplantation’s success, how- ate diverse effects through associated downstream signal-
ever, has been counterbalanced, by its dependence on ing pathways, their function is characterized by fidelity to
immunosuppressive drugs with their related infectious, distinct binding motifs. This trait has been long recognized
metabolic, and malignant complications. Consequently, a as having great potential for targeted therapeutic use with
common goal throughout the history of clinical transplan- minimal unintended effects, and organ transplantation his-
tation has been the minimization and individualization of torically has been a preferred testing ground for receptor-
immunosuppressive therapy. Typically, drugs with highly based therapeutics, such as monoclonal antibodies (MAbs),
specific mechanisms of action have been preferred over polyclonal antibody preparations, and engineered glyco-
drugs with broad effects, and the search for increasingly protein receptor–antibody hybrids known as fusion pro-
specific drugs has provided a major impetus for the devel- teins; these are collectively known as biologics. The initial
opment of immunosuppressive therapies in general, and of success of biologics in transplantation has more recently
antibodies and fusion proteins in particular. led to an explosion in the number developed for clinical
Antibodies and other glycoprotein cell surface receptors use.1 In addition to transplant-related indications, biologics
are defined by their ability to bind to a particular ligand have been developed for the treatment of many oncologic

283
284 Kidney Transplantation: Principles and Practice

and autoimmune conditions, and now at least 300 prepa- and cytokines, producing a syndrome of flu-like and, in
rations are in some level of clinical or preclinical develop- extreme cases, septic-like symptoms, subsequently termed
ment.2,3 Importantly, although renal allograft rejection cytokine release syndrome.
was the original indication for MAb therapy,4 most modern In the 1970s, Kohler and Milstein13 presented a land-
development has been spurred by indications serving larger mark development in the field of protein therapeutics—a
population bases. In addition to using agents developed for means of producing antibody preparations with a single,
transplantation, clinicians are increasingly adopting thera- genetically defined monoclonal specificity. The develop-
pies from other immunologically relevant indications. This ment of MAbs addressed many of the shortcomings associ-
so-called off-label use is now increasingly common and is ated with polyclonal preparations, particularly specificity
becoming a primary means of biologics development for and variability. The first such preparation approved for clin-
transplantation. ical use in 1985 was muromonab (OKT3), a MAb of mouse
This chapter provides an overview of antibody- and recep- origin specific for human cluster of differentiation (CD) 3
tor-based therapies for kidney transplantation. Drugs devel- (described later).4 OKT3 rapidly and specifically cleared T
oped and approved for use in transplantation are described, cells from the peripheral circulation and was shown to be an
as well as drugs with relevant actions developed for other effective treatment for allograft rejection.4,14–17 Although
indications but evaluated in transplantation. In addition, many of the problems associated with the diffuse nature
clinically tested investigational agents are reviewed. of polyclonal antibodies were addressed, some were not.
The immune response against heterologous animal pro-
teins and the cytokine release syndrome remained. OKT3’s
Historical Perspective heightened specificity for the T cell receptor (TCR) not only
produced more reliable T cell clearance but also more reli-
Early experiences in renal transplantation were marked able T cell activation and cytokine release. The antimouse
by high rates of rejection and complications related to antibody response also limited prolonged dosing in a subset
the effects of the two available immunosuppressants of of patients.18
the day, glucocorticosteroids and azathioprine; this, com- With the genetic engineering advances of the 1980s, the
bined with the recognition that lymphocytes were the production of MAbs became much more efficient, theoreti-
predominant effectors in rejection, stimulated interest in cally allowing any surface molecules to be targeted. Effort
alternative lymphocyte-directed strategies. By the mid- was redirected from pan-T cell depletion toward fine tar-
1960s, several investigators had shown that animals geting of relevant T cell subsets and blockade of functions
injected with lymphocytes would produce sera contain- unique to effector T cell activation. An example was the
ing lymphocyte-specific antibodies, which could be used high-affinity interleukin (IL)-2 receptor CD25 (described
to reduce the lymphocyte counts when injected into other later), expressed predominantly on activated T cells. Addi-
experimental animals. This technology gave rise to the tionally, methods of genetic engineering were developed to
initial lymphocyte depletion trials using antilymphocyte allow DNA encoding for binding sites from heterologous
antibody preparations: antilymphocyte serum, antilym- proteins to be grafted onto genetic sequences encoding
phocyte globulin, and antithymocyte globulin.5–8 These the monomorphic scaffold of human antibodies to create
agents were collectively called polyclonal preparations chimeric or humanized MAbs.19–21 These techniques also
because they were composed of antibodies with many, allowed for unique fusion proteins to be created, combin-
largely undefined specificities. Their ability to prevent and ing the Fc portions of antibodies with nonantibody recep-
reverse rejection, particularly in patients refractory to the tors and ligands and allowing for cell surface molecules to
drugs of the day, led to their increasing use over the ensu- be created in a soluble form with prolonged half-lives.
ing decade.9 The humanization of antibodies and the use of human-
The increased use of polyclonals made many of their derived receptors has practically eliminated the problem
limitations apparent. The imprecise in vivo methods for of antibody clearance and opened the possibility for pro-
producing polyclonal antibodies resulted in preparations longed treatment regimens. More recently, the production
with promiscuous binding to many nonlymphocyte cell of fully human antihuman antibodies has become a prac-
types. Although each antibody in the preparation bound tical reality.22 Techniques including phage display muta-
to a single target, collectively the preparation bound to a genesis and the transgenic production of mice containing
broad array of cell surface molecules. Cross-reactivity with human immunoglobulin genes that respond to immu-
many hematopoietic cells made anemia, neutropenia, and nization with human antibody now offer the promise of
thrombocytopenia dose-limiting. The method of produc- highly specific, nonimmunogenic, well-tolerated protein
tion also led to wide batch-to-batch variability. The clinical reagents. Human and humanized biologics are now mak-
effect of the agent varied considerably, making it difficult to ing possible prolonged therapy with highly specific thera-
establish prospectively proper dosages and to estimate the peutic agents.
magnitude of anticipatable side effects. In addition, because Multiple surface molecules have been targeted by bio-
the preparations were made in animals, usually rabbits logics investigationally, and several are now accepted as
or horses, they contained proteins that were antigenic to clinical therapies in transplantation and other indications.
humans.10,11 They had the potential to induce a neutral- Biologic therapy is being increasingly adopted into stan-
izing antibody response and evoke adverse effects, such as dard practice, with 95% of kidney transplants performed
serum sickness or anaphylaxis.12 Finally, some lympho- in the US now using some form of prophylactic antibody
cyte cell surface receptors, when bound by antibody, would therapy.23 Despite this trend, however, it has not been
induce cell activation, leading to a release of anaphylatoxins established whether this strategy is necessary in all cases.
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 285

Although antibody induction reduces acute rejection rates antibodies are sufficiently similar to their human counter-
in the first year after transplantation, the lasting effects of parts to facilitate most physiologic effector functions when
induction remain incompletely defined.24–26 The modern used in humans. Antibodies produced by mice, rabbits, and
era is now characterized by the availability of many promis- horses can be used in humans and still evoke biologically
ing agents and the challenge of understanding their most important effects. There is no animal that is a priori supe-
appropriate clinical use. rior, however, and all heterologous antibodies have the
potential to induce a neutralizing antibody response.
Antibodies can have a broad range of effects when they
Antibody Structure and Function bind (Fig. 19.2). They can mimic the native ligand of a mol-
ecule and lead to signal transduction, or they can bind to
The clinical effects of MAbs in transplantation relate closely the molecule in such a way as to prevent it from binding to
to the physiologic effects and structural characteristics of its intended ligand.29,30 Antibodies can be either activating
antibodies in general. Antibodies are one of two common or inhibiting, and the predominant effect can be determined
glycoprotein antigen receptors that result from somatic only through empirical in vivo analysis. Antibodies can
gene rearrangements in specialized lymphocytes, the other bind to cells in such a way as to have no appreciable effect.31
being TCRs.27,28 Five different heavy-chain loci (μ, γ, α, ε, Thus antibody binding cannot be equated with functional
and δ) and two light-chain loci (κ and λ), each with vari- significance. In some cases, a combined effect occurs
able, diversity, or junctional (V, D, or J) and constant (C) whereby the antibody activates the targeted molecule but
regions, are brought together randomly by the recombina- induces surface molecule internalization, effectively clear-
tion-associated gene (RAG)-1 and RAG-2 apparatus to form ing the molecule from the cell surface and inhibiting its
a functional antigen receptor with highly variable binding subsequent function.32 This transient activation effect can
ability. Antibodies have a basic structure of two identical lead to a burst of target cell activity (e.g., cytokine release),
heavy chains and two identical light chains (Fig. 19.1). resulting in undesirable side effects, or it can simply lead to
The heavy-chain usage defines the immunoglobulin type surface modulation of the targeted molecule. Antibodies
as being IgM, IgG, IgA, IgE, or IgD. This structure forms cannot target molecules that are not present on the cell sur-
two identical antigen-binding sites brought together on a face. Although they can influence intracellular pathways,
common region known as the Fc portion of the antibody. they cannot directly bind intracellular molecules in vivo.
Although all of these subtypes have therapeutic potential, Antibodies also activate the classical complement cas-
IgG antibodies have been the most commonly used clini- cade and in doing so can induce complement-mediated
cally. IgG molecules are the most common result of periph- lysis of a targeted cell. In addition, many phagocytic cells
eral immunization and are structurally easier to produce have receptors for the constant Fc region of antibodies and
and manipulate. preferentially engulf cells coated with antibody through a
Physiologically, antibodies exist as surface molecules on process known as antibody-dependent cellular cytotoxicity
B cells, facilitating their antigen-specific activation and, (ADCC). Both of these activities facilitate the most notice-
importantly, are secreted into the serum to bind to and able effect of antibody therapies: target cell depletion. Deple-
neutralize circulating antigens. Heterologous nonhuman tion is only the most obvious effect of antibody therapy,
however, and should not be assumed to be the most rele-
vant or desired. Additionally, these effects depend on their
Antigen binding sites antigen-binding region and their nonvariable Fc region
for effectiveness.33 The importance of Fc segment effects is
shown by nonspecific antibody infusion, which can medi-
Light chains ate important effects, presumably by neutralizing comple-
ment or saturating Fc receptors.34,35
It has become apparent that the maturation state of the
Heavy chains targeted cells also can influence the response to antibody
Fab treatments. Specifically, cells that have matured into a
memory phenotype have some degree of resistance to anti-
body-mediated depletion.36 The mechanisms involved in
depletion resistance remain to be defined, but memory cells
differ from naïve cells in many potentially relevant ways,
including enhanced antiapoptotic and complement regula-
tory gene expression. The ultimate effect of antibody ther-
apy may vary not only with the antibody preparation but
also with the phenotype of the targeted cell and even the
FC recipient’s immune history.
All of these effects can alter the function of molecules
and cells, giving antibodies broad therapeutic potential.
This array of effects makes antibody development difficult,
however. Minor changes in antibody structure can radi-
cally alter the effect, and at present it is impossible to predict
Fig. 19.1 General antibody structure. The prototypic structure of an an antibody’s properties based solely on structure. Certain
IgG molecule is shown. IgG isotypes support complement and ADCC functions
286 Kidney Transplantation: Principles and Practice

Blockade Antibody-dependent cell cytotoxicity (ADCC)

Phagocytosis

Fc
receptor

Internalization/activation Mimicry

Inert

Fusion
protein
Cytokine
release

Lysis Monoclonal vs. polyclonal

Complement
C1q

Membrane attack Single specificity Broad specificity


complex
Fig. 19.2 Mechanisms of action for antibody and fusion protein function. Antibodies can work via many mechanisms.

better than others, but generally an antibody must be tested provides another salutary property. In transplantation, this
in vivo to determine which of its many potential effects secondary molecule is typically the Fc portion of an IgG mol-
would be dominant.37 Specifically, new antibodies undergo ecule that gives the receptor an antibody-like half-life and/
detailed and rigorous analysis of structure, posttransla- or opsonization properties.40–42 Fusion proteins also can
tional modifications, and cellular functions during develop- involve the fusion of a specific toxin to a MAb to facilitate
ment.38 This can, at times, have serious consequences that epitope-directed drug delivery.43 Fusion proteins are similar
make the introduction of new antibodies into phase I clini- to MAbs in that they have a single homogeneous specificity
cal trials challenging.39 and can be composed of human or humanized components,
limiting their immune clearance and opening their use for
prolonged administration. Currently, only a single fusion
Fusion Protein Structure and protein is approved for transplantation, belatacept, which
Function is a mutated form of the receptor CTLA4 (CD152) fused to
IgG Fc domain. Belatacept, along with notable examples of
Fusion proteins are molecules engineered from a single recep- transplant-relevant fusion proteins in development, will be
tor targeting a ligand of interest fused to another protein that discussed subsequently.
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 287

General Clinical Considerations bolus methylprednisolone induction and maintenance with


cyclosporine, azathioprine, and prednisone.24,25 Despite
for the Use of Antibody and these benefits, there is no evidence that biologics alter long-
Fusion Protein Preparations term patient or graft survival in the era of modern immuno-
suppression. Additionally, the use of biologics increases the
Immunosuppressive regimens used for organ transplanta- risk of patient toxicity, including cytomegalovirus (CMV)
tion can be generally characterized as induction, mainte- disease, thrombocytopenia, and leukopenia.24–26,44 Long-
nance, or rescue therapies. Induction immunosuppression term analysis suggests that a measurable effect in kidney
is intense treatment designed to inhibit immune respon- transplantation disappears after 5 years. This analysis may
siveness prophylactically at the time of transplantation. It indicate that the side effects of maintenance therapy or
is usually potent to the point that prolonged use is prohibi- patient comorbidities supersede early graft outcome and are
tively toxic. Maintenance immunosuppression is of lesser the dominant determinants of outcome over time.
potency, but is tolerable for long-term use and forms the Antibody preparation use does not generally influence
basis of most immunosuppressive regimens. Rescue ther- the rate of technical complications47 but seems to reduce
apy is similar to induction in that it is intense, effective, and the risk of graft thrombosis in children.48 Several induc-
chronically intolerable, but differs in that it is used to reverse tion strategies, in particular polyclonal antibodies and
established rejection. Immunosuppressive medications can OKT3, have been shown to measurably increase the risk
conceivably fall into any or all of these categorizations based of posttransplantation lymphoproliferative disease (PTLD)
on the dose and route used. Currently, biologics are primar- and death from malignancy when combined with conven-
ily indicated as rescue agents and are used in approximately tional maintenance immunosuppression.49–53 PTLD is a
30% of all acute rejection episodes.44 Their use as induction product of the intensity of the overall immunosuppressive
agents predominates: 95% of patients undergoing kidney therapy in combination with the recipient’s preexisting
transplantation now receive biologic induction.23 immunity to the causative agent, Epstein-Barr virus (EBV).
Antibody preparations have been generally classified as Specifically, the expected PTLD rate is 0.5% in patients
depleting or nondepleting based on whether or not they who do not receive antibody induction or who receive
deplete cells expressing the targeted antigen. Generally, T CD25-specific therapy. OKT3 induction carries a signifi-
cell depleting antibody preparations are indicated for the cantly higher rate of 0.85%, as does polyclonal depletion at
treatment of refractory (e.g., steroid-resistant) acute cellular 0.81%, particularly in recipients newly exposed to EBV at
rejections, acute rejections occurring in high-risk settings transplantation.49 Interestingly, the use of alemtuzumab,
(e.g., marginal kidneys), and particularly aggressive vascu- a CD52-specific MAb with potent depletional properties
lar (e.g., Banff grade 2 or 3) rejections. Depleting antibod- similar in magnitude but differing in spectrum to polyclonal
ies are also used as induction agents, although this is often preparations or OKT3, has been shown to have a low PTLD
an off-label use. Nondepleting antibody preparations and incidence that approximates that of the nondepletional
fusion proteins have been most commonly studied as induc- CD25-specific MAbs.54 This likely relates to alemtuzumab’s
tion agents and typically have less efficacy in rescue indica- B cell depleting properties, with B cells being the dominant
tions. Applications of biologics have been made possible by reservoir of EBV, although practice patterns associated with
the ability to produce human biologics and so are gradu- alemtuzumab also may account for the difference.
ally being incorporated into maintenance immunosuppres- Other early complications, including cardiovascular
sion. The first biologic maintenance agent, belatacept, was and infectious deaths, correlate with antibody use, but the
approved for use as a calcineurin inhibitor (CNI) replace- interpretation of this relationship is confounded by the pref-
ment for kidney transplantation in June 2011.41,45,46 It is erential use of antibodies in high-risk patients.51,55 Viral
a fusion protein with specificity for the B7 costimulatory infection is a substantial concern, however, when using
molecules and will be considered fully later in this chapter. potent antibody therapy, particularly agents associated
Randomized trials have studied many depleting and with T cell depletion. When used for induction or rescue,
nondepleting antibody preparations. These agents are effi- antibody preparations should be accompanied by broad
cacious in reducing the rate of acute rejection when used prophylaxis against opportunistic infection. Antiviral
as induction agents and combined with standard mainte- therapy, such as ganciclovir or acyclovir,56–58 should be
nance regimens, compared with bolus methylprednisolone initiated and continued for at least 3 months. The choice
induction. Few prospective studies compare the prominent of agent is based on the pretransplant status of the donor
agents, however, and no agent has distinguished itself as and recipient. Oral candidiasis prophylaxis with nystatin or
clearly superior in all clinical circumstances. Most trials clotrimazole and Pneumocystis therapy with trimethoprim/
have used the surrogate end-point of acute rejection, rather sulfamethoxazole also should be considered for several
than more definitive outcome measures, such as patient or months. Individual clinical risks often dictate substantially
graft survival. longer periods of prophylaxis. Each antibody preparation
When considered as a whole, biologics have been con- has a unique side effect profile and indication, which are
vincingly shown to be more effective than steroids in revers- discussed subsequently.
ing first acute cellular rejection, but offer minimal benefit The use of antibody preparations for maintenance ther-
for humoral rejection episodes.44 When used as induction apy had been limited until more recently by the immune
agents, they reduce the incidence of acute rejection in the response formed against the antibody itself. Recombinant
first 6 months of transplantation in kidney recipients, partic- humanized or chimeric antibodies and fusion proteins have
ularly recipients who are sensitized or experiencing delayed essentially eliminated this as a concern. Long-term benefits
graft function, compared with the historical standard of with belatacept as a maintenance agent are now gaining
288 Kidney Transplantation: Principles and Practice

recognition and other antibodies are currently in clinical purification, but now all commercially available products
trials for sustained preventive therapy (discussed later in are purified to obtain only IgG isotypes. Even so, polyclonal
this chapter). antibody preparations are not fractionated to separate rel-
evant from irrelevant antibodies preexistent from the envi-
ronmental immune responses of the immunized animals.
Polyclonal Antibody Preparations More than 90% of antibodies found in polyclonal prepa-
rations are likely not involved in therapeutically relevant
Heterologous antibody preparations can be derived from antigen binding.59–62
many animals immunized with human tissues, cells (e.g., Many groups have prepared polyclonal antibody prepa-
human lymphocytes), or cell lines (e.g., Jurkatt cells). When rations for their own institutional use, and this practice
reinfused into humans, these antibodies bind to antigens gave rise to a highly variable literature with little standard-
expressed on the original immunogen, where they mediate ization or objective comparison between products.63–66
the effects discussed earlier. Given that these preparations More recently, three dominant commercial polyclonal
are produced through whole-cell immunization, the result- preparations have emerged: two rabbit-derived antibody
ing preparations contain a vast array of antibodies binding preparations, antithymocyte globulin–rabbit (ATG-R, Thy-
many epitopes expressed on the immunogen cells—some moglobulin, Genzyme-Sanofi) and antithymocyte globu-
intended, and some not. Because each animal produces a lin–Fresenius (ATG-F, Fresenius), and one horse-derived
unique immune response to an antigen, clinical-grade prep- product (ATGAM, Pfizer). Of these, Thymoglobulin is used
arations are generally the result of pooled responses from most commonly in North America,23 with both rabbit prep-
many animals. For practical reasons, most polyclonal prep- arations used in Europe. ATGAM is primarily used for the
arations are derived from rabbit or horse immunizations. treatment of aplastic anemia, but can be used for patients
Ideally, a single renewable cell type equivalent to the who have a hypersensitivity to rabbits.
effector cell in rejection could be used as a reproducible As discussed earlier, antibodies can mediate many effects
immunogen free from elements such as stromal tissue and when they bind to their target antigen, and a significant fac-
neutrophils. No such cell has yet to be identified or devel- tor determining their effect is the antigenic specificity of the
oped. Commercially available polyclonal preparations con- preparation. By their very nature, polyclonal preparations
tinue to be made using heterogeneous cell populations or are composed of a wide variety of antibodies, and complete
tissues such as thymus obtained from deceased donors or characterization has remained elusive.59–61,67 Detected
surgical specimens or from the Jurkatt T cell line, which specificities include many T cell molecules involved in
is thought to approximate the antigenic spectrum of allo- antigen recognition (CD3, CD4, CD8, and TCR), adhesion
specific T cells. After immunization, immunized animals (CD2, lymphocyte function antigen (LFA)-1, and intracel-
are bled to obtain hyperimmune serum. The serum is typi- lular adhesion molecule (ICAM)-1), costimulation (CD28,
cally absorbed against platelets, erythrocytes, and selected CD40, CD80, CD86, and CD154), non-T cell molecules
proteins to remove antibodies that could result in unde- (CD16, CD38, CD138, and CD20) and class I and II major
sirable effects such as thrombocytopenia. Historically, histocompatibility complex (MHC) molecules (Fig. 19.3).
hyperimmune serum was administered without additional Although all of these targets hypothetically can influence

Fig. 19.3 Sites of action for antibody and fusion proteins in clinical use. Shown are the surface molecules that have been targeted in clinical transplant
trials and their respective ligands when known. APC, antigen-presenting cell; ICAM, intracellular adhesion molecule; LFA, lymphocyte function antigen;
MHC, major histocompatibility complex; TCR, T cell receptor; TNF-α, tumor necrosis factor-α.
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 289

an immune response, and when studied individually they Most modern trials have evaluated polyclonal antibod-
do, it is unclear which of these specificities is crucial to the ies added to an otherwise rigorous maintenance regimen
ultimate therapeutic effect. This broad reactivity with adhe- (typically triple immunosuppressive therapy). This intense
sion molecules and other receptors upregulated on activated regimen has statistically reduced acute rejection rates, but
endothelium has led many authors to advocate the prefer- has reciprocated with increased infectious morbidity with-
ential use of polyclonal antibody preparations in situations, out changing the long-term outcome.26,78,79 The increased
such as prolonged ischemic times, where endothelial acti- infectious risk may be acceptable in selected higher-risk
vation and ischemia–reperfusion injury is anticipated.68,69 patient populations, such as recipients of donation after car-
Most polyclonal antibodies have prolonged serum half- diac death donors, recipients of organs with kidney donor
lives of several weeks.70,71 Nondepleted cells have been profile index >85%, formerly extended criteria donors, and
shown to be coated with heterologous antibody for months, patients with a high risk of rejection, such as African Ameri-
suggesting that these preparations could influence the func- can recipients, retransplant recipients, and recipients with
tion of lymphocytes long after treatment has stopped. Lym- delayed graft function,64,68,69,80–85 particularly when avoid-
phocyte subsets are abnormal for years after therapy, with ance of prolonged CNIs is desired.86–88 A seminal random-
particularly low CD4+ T cell counts.72 It also is reasonable to ized trial between ATG-R and CD25-specific (basiliximab)
assume that antibodies targeting differing specificities would induction showed that ATG-R reduced the incidence and
have variable effective half-lives based on the rates of surface severity of acute rejection but not the incidence of delayed
molecule recycling, the affinity of the binding interaction, graft function.89 Recently, another randomized study with
and the mechanism of action. Stimulating antibodies may ATG-R and daclizumab induction revealed sustained superi-
have effects whenever they are bound, whereas inhibitory ority of ATG-R to prevent acute rejection in highly sensitized
compounds could mediate an effect only when the natural patients.90 In contrast, a randomized trial for rapid steroid
ligand being antagonized is present. Polyclonal preparations withdrawal in nonsensitized patients showed no superior-
likely have mechanisms of action that vary by batch, cir- ity of ATG-R over basiliximab in preventing acute rejec-
cumstance of use, and degradation state. It is unlikely that tion at 1 year after renal transplantation.91 Although early
any single generalized mechanism exists. For the purposes of patient and graft survival were not influenced by the choice
following the clinical effect, bulk T cell depletion is used as of induction regimen, long-term studies have suggested both
a general estimate of antibody potency, and polyclonal anti- a patient and graft survival benefit with ATG-R.92–94
body preparations are considered depletional agents. Other trials have attempted to address the increased infec-
tious risk by pairing aggressive polyclonal induction with
substantially reduced maintenance therapy. Two pilot stud-
Specific Clinical Applications of ies have shown that ATG-R induction facilitates reduced
Polyclonal Antibody Preparations maintenance immunosuppression in highly selected,
closely followed patients, leading to graft and patient sur-
Polyclonal antibody preparations have been used in vivals comparable to the current standard.95,96 These stud-
transplantation to achieve immunosuppression since the ies have emphasized administration before reperfusion,
1960s.8 They are used as induction and rescue therapies, theoretically to take maximal advantage of antiadhesion
but the immune response to the proteins has precluded molecule effects, and relatively high-dose therapy, to limit
attempts to use them as maintenance drugs. As discussed the proinflammatory effects of reperfusion and to achieve
previously, no single mechanism of action has been estab- rapid and lasting T cell depletion. Although these studies
lished, and they likely mediate their antirejection properties indicate that such an approach is possible, it remains to be
through depletion and other effects, including costimula- seen if it can be generalized to noninvestigational settings.
tion blockade, adhesion molecule modulation, and, to a
lesser extent, B cell depletion.59–61,67,73
RESCUE
INDUCTION Although induction therapy remains an off-label indication
for polyclonal antibodies, their use for the treatment of ste-
Historically, polyclonal antibody preparations were used to roid-refractory rejection is an established indication. Many
bolster the effect of steroids and azathioprine in an attempt polyclonal preparations have shown their utility in this set-
to reduce the unacceptably high rejection rates typical of ting, spanning several decades of associated maintenance
the 1960s and 1970s. Generally, a 2- to 3-week course regimens. The first randomized trial showing that antilym-
of a polyclonal antibody delayed the onset of acute rejec- phocyte serum was superior to high-dose steroids for the
tion and reduced the requirement for high-dose steroids in treatment of established rejection was reported in 1979.97
the early postoperative period without significantly alter- In the context of azathioprine and prednisone maintenance
ing long-term survival.4,6,65,74,75 After the introduction of immunosuppression, antilymphocyte serum reversed rejec-
cyclosporine, the use of polyclonal antibody induction fell tion faster than bolus glucocorticosteroids, reduced the rate of
from favor with the realization that this potent combination recurrent rejection, and led to improved survival at 1 year.98
was associated with increased infectious and malignant Most rejection episodes in the cyclosporine era and beyond
morbidity.76,77 With improved viral prophylaxis, a better respond to bolus steroids. Polyclonal agents have been indi-
understanding of the infectious etiology of PTLD, and more cated as a second-line therapy for steroid-resistant acute cel-
standardized commercial polyclonal products, there has lular rejection.99–102 Recurrent rejection can be treated with
been a marked resurgence of interest in polyclonal antibody repeated courses of polyclonal antibodies in situations where
induction.23 antirabbit (or antihorse) antibodies have not formed.103,104
290 Kidney Transplantation: Principles and Practice

Of the currently available polyclonal preparations, Generally, rabbit-derived polyclonal preparations seem
ATG-R is used most commonly for rescue. It has been to be significantly better tolerated and more efficacious
shown to be superior to ATGAM in terms of reversal of ste- than ATGAM when used in a quadruple regimen for renal
roid-resistant rejection and persistence of a rejection-free transplantation.109,110 The most common acute symptoms
state.100 This difference has not been shown, however, to associated with polyclonal antibody use are the result of
influence patient or graft survival. transient cytokine release. Chills and fever occur in at least
Non-T cell-specific polyclonal antibody preparations 20% of patients and are generally treatable by premedica-
also reverse established cellular acute rejection. Although tion with methylprednisolone, antipyretics, and antihis-
not typically considered alongside T cell depleting poly- tamines. The use of polyclonal antibodies, particularly in
clonal antibody preparations, high-dose human IgG the treatment of rejection, has been associated with an
fractions (intravenous immunoglobulin) are polyclonal increase in the reactivation and development of primary
antibodies of random specificity pooled from human viral disease caused by CMV, herpes simplex virus, EBV,
donors. Because they are not derived from animals, are and varicella.111,112 It is likely, however, that this is not a
not the products of heterologous immunization, and do class-specific association, but rather an indication of more
not target a specific cell type, most of the adverse effects intensive immunosuppression in general.
associated with polyclonal antibodies are not applicable. Dosage adjustment is warranted to counter leukope-
Nevertheless, high-dose human IgG fractions have been nia and thrombocytopenia. Peripheral cell counts drawn
shown to reverse rejection despite the absence of any T immediately after infusion tend to exaggerate cytopenic
cell depleting abilities. Although a course of polyclonal effects, and most side effects are promptly remedied by time.
anti-T cell antibody typically consists of 5 to 20 mg/kg T cell counts or, more easily, absolute lymphocyte counts
given over several days, intravenous immunoglobulin is can be monitored to ensure that the preparation is achiev-
infused at much higher doses, 500 to 1000 mg/kg over ing its desired effect. Absolute lymphocyte counts less than
1 to 3 days, and at this dose has been shown to reverse 100 cells/μL are typical. Attempts to tailor therapy to a spe-
established rejection with the same overall reversal rate as cific peripheral cell count have been made to limit the use of
OKT3.34 At least at high dose, nonspecific antibody infu- these costly preparations. Rejection can occur and persist
sion can modulate immune responses, perhaps through with very low T cell counts, however, and there is little evi-
complement sequestration and Fc receptor binding with dence that dose variation by cell count alters efficacy.
resultant downregulatory effects of Fc receptor-expressing As discussed earlier, polyclonal antibody preparations
antigen-presenting cells (APCs).105 evoke a humoral immune response to themselves.10–12 This
response can be detected by enzyme-linked immunosor-
bent assay for antirabbit or antihorse antibody, but these
ADMINISTRATION AND ADVERSE EFFECTS
tests are typically unavailable in clinical settings. Failure
The polyclonal preparations used in modern clinical prac- to achieve significant T cell depletion suggests the presence
tice are generally given through a large-caliber central of these antibodies. Serum sickness and anaphylaxis also
vein to avoid thrombophlebitis. In experienced hands, can occur.12 Preemptive skin testing is not often practiced
a dialysis fistula can be accessed for this purpose. Some because these tests have not correlated well with clini-
reports have suggested that polyclonal antibodies can cal outcome.113,114 Rather, slow infusion rates should be
be administered peripherally when diluted and formu- employed during initial exposure. Xenospecific antibodies
lated with heparin, hydrocortisone, or bicarbonate solu- are most likely to occur in individuals with prior exposure
tions.106,107 An in-line filter is recommended to prevent to the preparation involved, but can also exist in individuals
infusion of precipitates that may develop during storage. with significant prior exposure to the animals themselves.
The protein content should not exceed 4 mg/mL, and dex- The most common adverse symptoms related to poly-
trose-containing solutions should be avoided because they clonal antibodies are fever, urticaria, rash, and headache.
induce protein precipitation. These are most likely related to the release of pyrogenic
Given the weeks-long half-lives of polyclonal antibod- cytokines, such as tumor necrosis factor (TNF)-α, IL-1, and
ies, divided doses are not required for steady-state levels. IL-6, which result from activating antibody binding to tar-
The tolerability of these compounds is markedly improved, geted cell surface receptors and subsequent cell lysis.115–117
however, by spaced dosing. The rate of infusion is associ- Infrequently, pulmonary edema and severe hypertension or
ated with the severity of side effects, and the course of ther- hypotension can result in death. As the number of target
apy is generally over several days, with individual doses cells decreases with repeated dosing, this response typically
given over 4 to 6 hours. This time course depends on the abates. The most concerning response is within the first 24
dose used and is most applicable to the standard doses of hours of the first dose, and patients should be monitored
ATG-R and ATG-F (1.5 mg/kg/dose for a total of 7.5–10 closely during this period. The response is limited consid-
mg/kg) or ATGAM (15 mg/kg/dose for a total of 75–100 erably by methylprednisolone premedication. The rash
mg/kg). More recent investigational induction studies have associated with polyclonal antibody administration, con-
employed substantially higher doses given over 12 to 24 versely, tends to occur late in treatment or, at times, after
hours or, alternatively, while the patient is anesthetized the last dose. It is generally self-limiting and requires only
with comparable safety profiles.82,95,96,108 With a grow- symptomatic treatment for urticaria. Antiendothelial anti-
ing emphasis being placed on reduced length of stay after bodies in polyclonal antibodies have been suggested to bind
transplantation, larger infusions over fewer days are being to donor endothelia and activate complement, inducing
employed. humoral rejection in some patients.118
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 291

The resultant hybrid antibody gene can be transfected into


Monoclonal Antibody and Fusion a high-expressing eukaryotic cell line and grown in vitro to
Protein Preparations produce antibodies that are predominantly human antibody,
yet still bind to a specific human epitope (Fig. 19.4). These
MAb preparations differ from polyclonal preparations in hybrid antibodies can be considered chimeric, if the entirety
that all antibody molecules are derived from a single genetic of the murine antibody-binding site is used in the construct,
template and are identical. Batch-to-batch variation is elim- or humanized, if the only murine portion is the specific com-
inated, allowing the mechanism of action and half-life to be plementary determining regions of the parent antibody.20
extrapolated based on a single ligand receptor interaction Generally, chimeric antibodies preserve the specificity of
(although this still can be influenced by many individual- the original antibody better, whereas humanized antibodies
ized circumstances). This preparation narrows the scope of have less chance of evoking a neutralizing response.124 Prac-
effect, however, making the use of these drugs more depen- tically speaking, both are effective strategies that help avoid
dent on precise knowledge of the pathology involved. the problem of antibody clearance.
Historically, MAbs are the product of clonally immor- The entire IgG gene has been transgenically expressed in
talized B cell hybridomas. More recently, genetically engi- a mouse.22 This animal, when immunized, makes a human,
neered mammalian cells have been the source. Alternative not mouse, antibody, which can be prepared for monoclo-
production methods, including other eukaryotic cells such nal production. This method is likely to be more efficient for
as yeast, prokaryotic bacteria, viruses, or even plant cells, producing truly human antihuman antibodies without the
are being utilized.119,120 As the production cell becomes need to engineer each antibody individually.
increasingly distant from human, the resultant antibodies When approved for clinical use, MAbs must be named
have increasingly aberrant glycosylation, which can radi- based on their structural characteristics (Table 19.1). The
cally alter their efficacy.33 Regardless of the production cell, generic name of a MAb gives the practitioner a reasonable
the resultant antibody can be purified of any extraneous understanding of the origins and specificity of the MAb.
proteins or other antibodies and used as an infused drug.
The most common method for deriving a MAb is typically
to immunize a mouse with a cell or cell fraction containing Monoclonal Antibodies and
the antigen desired. Splenocytes are isolated from the immu-
nized animal and fused with an immortalized cell, producing
Fusion Proteins in Clinical
many diverse antibody-producing cells. These cells are cloned Transplantation Practice
(grown from single-cell suspensions), and the supernatant
from each clone is tested for reactivity against the desired Because each MAb has a singular specificity, each agent
antigen. A single robust clone with the desired antibody pro- available for general clinical use is considered individually
duction characteristics is chosen and grown either in vitro or (see Fig. 19.3). Most MAbs are defined based on their tar-
in a carrier animal. The supernatant from the clone is puri- geted cell surface protein, and these are generally classified
fied for therapeutic use. Because many MAbs are made by based on the CD nomenclature. A numerical CD designa-
mouse B cells, they are mouse antibodies. Similar to animal- tion does not define an antigen, but rather defines a mol-
derived polyclonal antibodies, they can be cleared from the ecule or group of molecules. MAbs that bind to the same
circulation by an antibody-directed immune response.121 CD molecule can bind to the same or different epitopes and
This immune response can cause anaphylaxis and neutralize have similar or different effects.
the effect of the MAb in subsequent administrations.122
To improve the efficiency of antibody production and elim-
MUROMONAB (OKT3; MURINE ANTI-CD3)
inate animal-derived protein epitopes, the gene fragment
encoding the binding site of murine antibodies can be iso- OKT3 (muromonab) is no longer available for clinical use.
lated and engineered onto the gene that encodes for nonpoly- However, it is considered here given its unique place in the
morphic regions of a human antibody, such as IgG1.19,21,123 historical continuum of MAb therapy. OKT3 is CD3-specific;

Mouse MAb Chimeric MAb Humanized MAb Fully human MAb Fusion protein
Fig. 19.4 Types of monoclonal antibodies (MAbs) and fusion proteins. Dark areas represent portions of the molecule of nonhuman origin, and light
areas represent human proteins.
292 Kidney Transplantation: Principles and Practice

TABLE 19.1 Nomenclature for Monoclonal Antibodiesa


Target Source Suffix
Varies based on preference -v(i)- Viral -u- Human -mab
of developer -b(a)- Bacterial -o- Mouse
-l(i)- Immune -a- Rat
-les- Inflammatory lesions -e- Hamster
-c(i)- Cardiovascular -i- Primate
-t(u)- Melanoma, colonic, testicular, ovarian, mam- -xi- Chimeric
mary, prostate, miscellaneous tumors
-f(u)- Fungus -zu- Humanized
-gr(o)- Growth factor -axo- Rat/mouse
-tox(a)- Toxin
-k(i)- Interleukin
-s(o)- Bone
-ne(u)(r)- Nervous system
aThe naming of antibodies follows the general guideline with the target and source preceding the suffix -mab.

CD3 is a transmembrane complex of proteins that links to internalization, cytokine-mediated regulatory changes, dis-
the TCR and conveys its activating signal to the nucleus via rupted trafficking, and cell depletion. OKT3 has proven effi-
a calcineurin-dependent pathway, thus serving as the fun- cacy as an induction and rescue agent. Its immunogenicity
damental signal in antigen-specific T cell activation. CD3 is has prevented its use as a maintenance agent, and the drug
present on essentially all T cells, defining the cell type. The is effective only in combination with other immunosuppres-
TCR signal is generally known as signal 1 because it is pri- sive compounds.127
marily required for T cell activation and defines the antigen
specificity of the T cell. Given that T cells are a crucial medi- Induction
ator of acute cellular rejection, CD3 was one of the first mol- Initial trials with OKT3 have shown that this MAb is an effi-
ecules to be targeted with MAbs, and OKT3 (muromonab) cacious induction agent in kidney transplantation,128–131
was the first MAb to gain clinical approval for therapeutic but only when combined with otherwise effective main-
use in humans.15 tenance immunosuppression.127 OKT3 cannot prevent
Although the molecular target of OKT3 is singular and rejection beyond the period of its actual infusion without
precise, its effects are many. The mechanism by which additional maintenance therapy. Its usefulness as an induc-
OKT3 mediates its immunosuppressive effect remains ill tion agent is most pronounced in sensitized patients132 and
defined. OKT3 is an IgG2a mouse antibody that binds to patients with delayed graft function, in whom it facilitates
the ε component of human CD3. On binding, the antibody the delay of CNI administration and the resultant nephro-
mediates complement-dependent cell lysis and ADCC and, toxicity.133,134 It reduces the number of acute rejection epi-
in doing so, rapidly clears T cells from the peripheral circu- sodes and the time to first rejection episode. In more recent
lation.117 This binding event also leads to pan-T cell activa- literature, OKT3 has been shown to reduce acute rejection
tion before their elimination, resulting in systemic cytokine episodes compared with cyclosporine, azathioprine, or
release. The result is a marked cytokine release syndrome mycophenolate mofetil (MMF) and steroids without chang-
responsible for most of the adverse effects associated with ing patient or graft survival,135–137 but to be equivalent to
the drug (see later). intravenous cyclosporine induction in children.138 Despite
When antigen binds to the TCR, TCR-CD3 internalization its early prominence, the use of OKT3 as an induction agent
occurs; physiologically, this ensures that antigen binding is dramatically declined subsequently, primarily as a result of
reflective of antigen burden and avoids activation mediated its side effect profile, and this led to its voluntary withdrawal
by continuous binding of a low-prevalence antigen. Simi- from the market in 2009.
larly, OKT3 binding to CD3 leads to TCR-CD3 internaliza- Because OKT3 is an entirely mouse-derived antibody,
tion.115 Uncleared T cells are often rendered void of surface its use leads to the development of an antibody response
TCR, are incapable of receiving a primary antigen signal, directed against OKT3 in a significant percentage of
and are immunologically inert. patients. The development of antimouse antibodies varies
Bulk T cell clearance likely is not the primary mechanism based on the concomitant immunosuppression given, but is
of action of OKT3. Clinical rejection can occur with excep- seen in at least 30% of patients.
tionally low T cell counts achieved by other means, and
stable graft function can occur with large T cell infiltrates Rescue
within the graft itself.125,126 Although the peripheral circu- The primary indication for OKT3 was for the treatment of
lation is rapidly cleared by OKT3, many T cells can be found biopsy-proven, steroid-refractory, acute cellular rejection.
in the periphery and in the allograft itself.32 A substantial In this indication, the side effect profile was deemed justifi-
amount of the rapid T cell clearance from the circulation able, and the efficacy of OKT3 was undeniable.4,15,139–142
is likely related to lymphocyte marginalization, perhaps OKT3 was successful in providing sustained reversal of
induced by the cytokines released and by the methylpred- approximately 80% of these vigorous rejections. It was
nisolone that is given with OKT3. The overall effect of OKT3 effective even in the presence of prior aggressive lympho-
is likely an aggregate effect of interrupted TCR binding, TCR cyte depletion, suggesting that its mechanism of action was
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 293

not primarily a result of bulk T cell depletion.16,126,143 The Induction


incidence of steroid-refractory rejection, defined as failure to Many anti-CD25 antibodies, including anti-Tac,148
respond to 3 consecutive days of bolus methylprednisolone 33B3.1,151 LO-Tact-1,152 and BT563,153 have been tested
(e.g., 500 mg daily), declined considerably with improved in humans and been shown to delay modestly or reduce
maintenance immunosuppressive agents, as did the inci- the onset of acute rejection when used with conventional
dence of rejection in general. Thus the need for OKT3 was maintenance immunosuppression. Experimental rodent
reduced considerably. That, combined with its unfavorable antibodies have been generally abandoned in favor of the
side effect profile relative to newer agents, led to its with- humanized/chimerized antibodies.
drawal from the US market in 2009. Sporadic use based on Daclizumab and basiliximab have been shown to mod-
existing stocks of the drug continued into 2010. estly reduce the incidence of acute cellular rejection com-
pared with methylprednisolone induction when used in
INTERLEUKIN-2 RECEPTOR (CD25)-SPECIFIC triple or double immunosuppressive regimens, with excep-
MONOCLONAL ANTIBODIES tional patient tolerability in kidney and extrarenal trans-
plantation.154–161 Studies comparing basiliximab with
The receptor for IL-2 is composed of three chains (α, β, and polyclonal antibodies in regimens using cyclosporine, MMF,
γ), of which the α and γ chains are constitutively expressed, and steroids have shown comparable outcomes.162–164 The
and the β chain is induced with activation. The presence of use of depletional agents appears to be preferable in high-
the β chain, now designated as CD25, indicates prior T cell risk situations.89,94,165 The magnitude of the antirejection
activation and identifies cells that have undergone some effect seen with anti-CD25 therapy depends to some extent
degree of effector maturation. CD25 has been targeted to on the intensity of the maintenance regimen, with earlier
suppress activated cells, while sparing resting cells. trials using cyclosporine-based and azathioprine-based
Two commercially available anti-CD25 antibodies have regimens showing a 25% reduction and later trials in the
been developed, both of which have been engineered to tacrolimus/MMF era showing a more modest 10% improve-
avoid antimurine antibody responses. Daclizumab is a ment. Anti-CD25 induction also has been used successfully
humanized anti-CD25 IgG1, and basiliximab is a chimeric in steroid-free regimens in kidney transplantation and asso-
mouse–human anti-CD25 IgG1. Both agents avoid immune ciated with sustained improvements in growth for pedi-
clearance and can be used for prolonged periods without atric recipients.166–170 The use of anti-CD25 has not been
inducing a neutralizing antibody.144–147 CD25 was the first shown, however, to facilitate more aggressive maintenance
molecule to be targeted successfully with a humanized MAb reduction regimens, such as monotherapy or calcineurin
in transplantation.148 These agents also avoid the serum avoidance.171–173
sickness associated with mouse-, rabbit-, or horse-derived
proteins. Daclizumab was voluntarily withdrawn in 2009, Administration and Adverse Effects
largely based on market rather than biologic considerations. Although the efficacy of anti-CD25 therapies is modest, the
A subcutaneous form of daclizumab that may have future safety profile is highly favorable.154–161,164 The binding of
implications in transplantation was reintroduced and US anti-CD25 antibodies does not mediate T cell activation,
Food and Drug Administration (FDA)-approved in 2016 for and no perceptible cytokine release occurs. Clinical trials
relapsing multiple sclerosis. Currently, basiliximab remains have generally shown no increase in infectious complica-
the only CD25-specific MAb available for use in transplan- tions or delayed wound healing. The risk of PTLD with anti-
tation. Studies for both of these agents are considered as the CD25 induction is similar to that when no induction agent
efficacy and mechanisms of action of these agents appear to is employed.49,54
be practically interchangeable.
Anti-CD25 antibodies are thought to work primar-
ALEMTUZUMAB (HUMANIZED ANTI-CD52)
ily through steric hindrance of IL-2 binding to CD25 and
deprive T cells of this cytokine during early activation. There Given the reduction in rejection achieved with prolonged
is little evidence for a depletional effect, or if there is one, it polyclonal antibody-mediated T cell depletion, the ease of
is limited to a few cells. More recently, it has become clear administration and consistency of MAbs, and the benefits
that CD25 induction is involved not only in the activation of humanization, clinicians have sought agents with a
of cytotoxic T cells but also in the activation of cells with combination of these traits. The CD52-specific humanized
potentially salutary effects on the allograft, such as T regu- MAb alemtuzumab has emerged as a promising depletional
latory cells.149 Previously activated T cells that are respond- MAb.165,174–177
ing in an anamnestic response are less dependent on IL-2 Alemtuzumab (Campath-1H) is a humanized IgG1 deriv-
for proliferation. Heterologous responses (cross-reactive ative of a rat antihuman CD52.176 CD52 is a nonmodulat-
responses between a previously encountered pathogen and ing, glycosylphosphatidylinositol-anchored membrane
an alloantigen) or memory alloimmune responses seem not protein of unknown function found in high density on
to be affected significantly by CD25 interruption. Given this most T cells, B cells, and monocytes.178 CD52 is not found
biology, primarily focused on naïve T cell early activation, on hematopoietic precursor cells and does not seem to be
CD25-directed antibodies have found a role in induction, an adhesion molecule; it is not necessary for T cell activa-
but have no role in the treatment of established rejection. tion. Several versions of the nonhumanized anti-CD52 pre-
Although there has been anecdotal experience using these decessors of alemtuzumab have been shown to be effective
antibodies for maintenance immunosuppression in the set- in mediating rapid T cell depletion and reversing steroid-
ting of CNI toxicity with recurrent rejection,150 no study resistant rejection. The humanized form has been studied
has formally evaluated this approach. in several indications and is currently approved for the
294 Kidney Transplantation: Principles and Practice

treatment of multiple sclerosis and marketed as Lemtrada. (HLA)-specific antibodies remains to be determined. There
Initially approved in 2001 to treat lymphogenous malig- appears to be a homeostatic response to the B cell depleting
nancies, it was withdrawn from the market in 2014. It effect, leading to high levels of B cell activating factor BAFF
remains available for this indication through the worldwide and concomitant increases in activated B cells emerging
Campath Distribution Program. Although not approved for after alemtuzumab administration that is a potential mech-
use in solid-organ transplantation, alemtuzumab has been anistic explanation for a rise in alloantibody production.174
used off-label as an induction agent.179,180 Its mechanism
of action seems to be predominantly related to bulk T cell Rescue
depletion, with lesser depletion of B cells and monocytes. The rodent antihuman CD52 predecessors of alemtu-
It rapidly depletes CD52-expressing lymphocytes centrally zumab, Campath-1M, and Campath-1G, were originally
and peripherally in renal transplant recipients.126 The use tested as rescue agents.195–198 In the original studies using
of alemtuzumab as a rescue drug is burgeoning, and there anti-CD52 for steroid-resistant rejection, the antibodies
has been anecdotal investigation in this drug as a mainte- were used with triple immunosuppression and steroid bolus
nance therapy. Alemtuzumab is currently unavailable for therapy, leading to a prohibitively immunosuppressive reg-
commercial use in transplantation, but is being supplied for imen with excess infectious morbidity and mortality. With
transplant use by its maker (Sanofi). the success of alemtuzumab as an induction agent, interest
in using it as a rescue agent has resurged. Several anecdotal
Induction reports have emerged in both children and adults.199–203
In preliminary, uncontrolled studies, alemtuzumab has Additional study is required to define its role in this setting,
been shown to facilitate reduced-maintenance immuno- although its predilection for naïve cells may limit its efficacy
suppressive requirements without an apparent increase after sensitization.
in infectious or malignant complications in kidney and
extrarenal transplantation compared with historical con- Administration and Adverse Effects
trols.126,179,181–192 Specifically, alemtuzumab has been Alemtuzumab can be administered through a peripheral
used to achieve perioperative depletion in combination intravenous catheter and can be dosed as a 30-mg flat dose
with triple immunosuppression and early steroid weaning; or at 0.3 mg/kg dose over 3 hours. Almost total elimination
steroid-free regimens with CNIs and MMF maintenance; of peripheral CD3+ T cells can be expected within 1 hour
and with monotherapy regimens of cyclosporine, tacro- of the first infusion, although secondary lymphoid deple-
limus, or sirolimus. Graft and patient survival has been tion requires 48 hours and at least two doses.36,126 Higher
comparable to contemporaneously reported registry data, doses have not been shown to be of additional benefit in
although the incidence of reversible rejection has predict- transplantation.
ably increased with decreases in concomitant maintenance The rapid depletion characteristic of alemtuzumab is
therapy. Prospective comparison of alemtuzumab has been associated with a cytokine release phenomenon similar
shown to provide similar outcomes in low-risk patients com- to, but typically less severe than, that seen with polyclonal
pared with basiliximab induction, and similar outcomes antibodies or OKT3. Administration should be preceded by
in high-risk patients compared with ATG-R.165 Addition- a bolus of methylprednisolone, diphenhydramine, and acet-
ally, compared with standard basiliximab-based therapy, aminophen. The first dose should be given in a setting capa-
alemtuzumab induction followed by early CNI reduction, ble of dealing with hypotension, anaphylaxis, and other
mycophenolate exposure, and steroid avoidance reduced sequelae of cytokine release. Neutralizing antibodies have
the risk of biopsy-proven acute rejection at 6 months with not been described for alemtuzumab.
no increased risk of serious infections or death.193 Long- Early trials investigating alemtuzumab as a therapy for
term follow-up is ongoing to assess the sustained effects on multiple sclerosis suggested an association between its use
allograft function and survival. and the development of autoimmune thyroiditis.204 Specifi-
Mechanistic studies investigating alemtuzumab induc- cally, patients with multiple sclerosis receiving high-dose
tion have shown that, although it depletes all T cell sub- investigational therapy with alemtuzumab had a signifi-
sets to some degree, it has modest selectivity for naïve cell cantly increased risk of hyperthyroidism developing 1 to
types.36 Nondepleted T cells exhibit a memory phenotype 3 years after therapy and declining thereafter.205 It has
and seem to be most susceptible to CNIs. Maintenance regi- been hypothesized that T cell depletion, particularly deple-
mens including CNIs seem to do best in alemtuzumab-based tion that selectively spares activated cells, could disrupt T
maintenance reduction strategies. The rapid and profound cell regulation and unmask autoreactive clones. This effect
depletion allows for a delay in the initiation of therapeutic could be most evident in individuals with low-level adju-
CNI levels and makes this an attractive option for patients vant maintenance immunosuppression, as was the case in
with delayed graft function.188 the multiple sclerosis trials. There has been a case report of
Although alemtuzumab depletes B cells, its effect on T autoimmune thyroiditis in an alemtuzumab-treated renal
cells is more profound and lasting. It does not clear plasma transplant patient, leaving the potential for autoimmune
cells. Some investigators have associated alemtuzumab disease as an unresolved matter of concern.206,207
administration with an increase in antibody-mediated
rejection or at least posttransplant development of donor- RITUXIMAB (HUMANIZED ANTI-CD20)
specific alloantibody.194 Whether this association is related
to the effects of the antibody, the reductionist maintenance Rituximab is a chimeric MAb-specific for CD20. CD20 is a
regimens used with alemtuzumab, or specifics of patient cell surface glycoprotein involved in B cell activation and
selection and screening for human leukocyte antigen maturation whose natural ligand is unknown.208 Similar to
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 295

alemtuzumab, it has been developed and approved for use immunosuppression reduction is the primary therapeutic
in lymphogenous malignancies, particularly CD20+ B cell maneuver in PTLD, rituximab has emerged as an effec-
lymphomas and PTLD.209 Given its specificity for B cells (and tive and well-tolerated maneuver to be interjected between
despite its lack of specificity for antibody-producing plasma immunosuppressive withdrawal and more aggressive
cells), rituximab has been suggested to be a therapy for anti- chemotherapy.234
body-mediated rejection and rejections involving vasculi-
tis.210,211 Rituximab also has been used in regimens designed Administration and Adverse Effects
to facilitate transplantation in sensitized individuals, such Rituximab can be administered through a peripheral vein
as ABO-incompatible donor–recipient pairs or transplants and is associated with few overt side effects. As with all pro-
across a positive crossmatch after antibody removal.212–218 teins, anaphylaxis can occur, and initial doses should be
Lastly, rituximab has been used with varying efficacy to treat given in a monitored environment. Additionally, rituximab
recurrent diseases posttransplantation, including mem- has been linked to serious infections including Pneumocys-
branous nephropathy, focal segmental glomerulosclerosis tis jiroveci (PJP) pneumonia, reactivation of hepatitis B virus
(FSGS), antineutrophil cytoplasmic autoantibody mediated (HBV), and tuberculosis (TB). Multiple factors, including
vasculitis, and IgA nephropathy.207,219–225 At present, the prolonged B cell depletion, B cell–T cell crosstalk, panhy-
role of rituximab in transplantation is largely investigational; pogammaglobinemia, and blunted immune response after
however, similar to alemtuzumab, its off-label use is increas- immunizations, contribute to the increased infectious risk
ing considerably, despite a randomized trial adding ritux- with rituximab. Therefore updated vaccinations, screening
imab to standard immunosuppression that showed a marked for HBV and TB before treatment, and PJP antimicrobial
increase in rejection in the rituximab arm.226 prophylaxis are recommended.235 When used as a treat-
The mechanism of action of rituximab is presumed to be ment for PTLD, it is typically given at a dose of 375 mg/
depletional, primarily through induced apoptosis.227,228 m2. Dosing as an immunosuppressant has empirically fol-
Treatment with this antibody rapidly and specifically clears lowed this regimen. Rituximab persists in the circulation
CD20+ cells from the circulation. The role of CD20+ cells in for weeks to months, and a single dose effectively eliminates
alloimmune responses is currently incompletely defined. CD20+ cells for a similarly prolonged period. The presence of
Although these cells are precursors to antibody-producing rituximab in the serum artificially produces a pan-positive
plasma cells, they do not produce antibody without further B cell crossmatch by complement-dependent cytotoxicity
maturation. Their role in acute antibody production is not and flow techniques. Characterization of alloantibody after
well established, and it is unlikely that they have a direct the use of rituximab requires alloantigen-specific methods,
effector cell role in rejection. Several authors have docu- such as solid-phase bead array assays.
mented CD20+ infiltrates as a marker for particularly recal- Numerous other humanized and fully human CD20-spe-
citrant acute rejection.229,230 These cells are known to have cific MAbs are under development for a host of immune and
APC function and it has been postulated that they serve oncologic indications.117,236 Their use in transplantation
to facilitate intragraft antigen presentation. Although the remains expectant.
mechanism of action for rituximab and recurrent disease
remains unclear, rituximab has been shown to stabilize the BELATACEPT
renal podocyte by binding to SMPDL-3b protein, thereby
decreasing proteinuria in FSGS.231 Currently, rituximab is Maintenance
being used in induction and rescue indications. Belatacept is the first fusion protein to achieve FDA approval
for use in kidney transplantation. It is a fusion of a recombi-
Induction nant, high-affinity form of the T cell coinhibitory molecule
The use of rituximab as an induction agent has been limited CTLA4 (CD152) fused to a human IgG Fc portion. Com-
to patients with known donor-specific sensitization. In par- posed of two human proteins, it evokes little heterologous
ticular, rituximab has been suggested to be a surrogate for antibody response and can be given indefinitely. As such,
recipient splenectomy in patients undergoing donor desen- belatacept is the first antibody or fusion protein in trans-
sitization with plasmapheresis or intravenous immunoglob- plantation given as a maintenance agent rather than for
ulin infusion, or both.215,216 It has not been prospectively induction or rescue. Indeed, belatacept has no efficacy as an
studied, but rituximab seems to have some effect in reducing induction or rescue agent per se, and will thus be discussed
the rebound of alloantibody in these complex patients. here only for its role as a maintenance immunosuppressant.
Belatacept has higher binding affinity to CD86 and CD80
Rescue than its parent compound abatacept, which is mostly used
Several reports have emerged suggesting that rituximab for rheumatologic diseases. Significant interest in belata-
has a role in the treatment of vascular rejection (Banff clas- cept was driven by a pivotal phase II study conducted to
sification 2 and 3) and in reversing emerging alloantibody compare the efficacy of belatacept as a maintenance immu-
formation.210,211,232 This would be presumed to be rel- nosuppressant to the standard CNI cyclosporine in combi-
evant for allograft infiltrates shown to contain CD20+ cells, nation with steroids, MMF, and a basiliximab induction.46
although specific guidelines for the use of rituximab remain The study demonstrated that belatacept-based immuno-
forthcoming. As with its use as an induction therapy, use of therapy did not increase rates of acute rejection at 6 months
rituximab as a rescue agent remains investigational. and improved renal function at 1 year compared with cyclo-
Rituximab’s most important indication in organ trans- sporine. This laid the basis for a large, international phase
plantation is not as a rescue agent for rejection, but III trial, the Belatacept Evaluation of Nephroprotection and
rather as a primary treatment for PTLD.233 Although Efficacy as First-line Immunosuppression Trial (BENEFIT),
296 Kidney Transplantation: Principles and Practice

1.0

Probability of patient and


0.9
0.8

graft survival
0.7
0.6
0.5 Belatacept MI vs. cyclosporine: hazard ratio for
0.4 death or graft loss, 0.57 (95% Cl, 0.35–0.95);
0.3 Belatacept MI P = 0.02
0.2 Belatacept LI Belatacept LI vs. cyclosporine: hazard ratio for
0.1 Cyclosporine death or graft loss, 0.57 (95% Cl, 0.35–0.94);
0.0
0 6 12 18 24 30 36 42 48 54 60 66 72 78 84
Months since transplantation
No. at Risk
Belatacept MI 219 212 208 206 204 202 199 153 151 149 146 142 135 131 128
Belatacept LI 226 220 218 216 213 209 204 165 161 159 152 151 142 139 137
Cyclosporine 221 208 206 202 199 197 186 137 123 117 112 107 102 100 92

Fig. 19.5 Long-term patient and graft survival for belatacept- versus cyclosporine-based regimens from the BENEFIT trial. (From Vincenti F, Larsen C,
Durrbach A, et al. Costimulation blockade with belatacept in renal transplantation. N Engl J Med 2005;353:770–81. Used with permission.)

80

70
Mean eGFR (ml/min/1.73 m2)

60

50

40

30

20 Belatacept MI
10 Belatacept LI
Cyclosporine
0
0 6 12 18 24 30 36 42 48 54 60 66 72 78 84
Month
Fig. 19.6 Patients on belatacept-based regimens had a significant increase in mean estimated glomerular filtration rate compared with cyclosporine
over an 84-month study period in the BENEFIT trial. (From Vincenti F, Rostaing L, Grinyo J, et al. Belatacept and long-term outcomes in kidney transplantation.
N Engl J Med 2016;374:333–43. Used with permission.)

designed to evaluate less intensive and more intensive belatacept-based more intensive (MI) and less intensive (LI)
belatacept- versus cyclosporine-based regimens in approxi- regimens (22% and 17%, respectively) versus the cyclo-
mately 650 adult patients receiving kidney transplants from sporine group (7%), and CD57+ CD4 T cells have been
living or standard criteria deceased donors.45,237 Overall, implicated (Fig. 19.7).243 The majority of acute rejection
the trial was a success. Belatacept immunotherapy resulted episodes occurred early (within the first 3 months), showed
in significantly higher patient and graft survival, and mean no sign of recurrence, and resolved with treatment. More
estimated glomerular filtration rate over the 7-year period importantly, the use of depletional therapy in conjunction
(Figs. 19.5 and 19.6). Patients who received expanded cri- with belatacept has eliminated the risk of acute rejection in
teria donor kidneys (i.e., older age and more comorbid con- some exploratory studies.243
ditions than standard criteria donors, BENEFIT-EXT) and Other exploratory open-label phase II trials244,245 have
were treated with belatacept experienced similar long-term examined whether the use of belatacept-based immuno-
benefits for renal allograft function, although there was no suppression can entirely preclude CNIs and corticoste-
difference in patient or allograft survival compared with roids.144,200 One study randomized recipients of living and
cyclosporine at 7 years.238 Additionally, patients treated standard criteria deceased donors to receive belatacept-
with belatacept had lower rates for developing de novo MMF, belatacept-sirolimus, or tacrolimus (TAC)-MMF, a
donor-specific antibodies, likely due to suppression of fol- standard steroid avoidance regimen.144 The belatacept-
licular helper T cells and B cell clonal expansion in germinal MMF group had a higher rate of acute rejection at 6 months
centers of the lymph nodes and spleen.239 (12%) versus the belatacept-sirolimus (4%) and TAC-MMF
Belatacept was generally well tolerated by patients. The (3%) groups, although 12% is much lower than reported in
long-term safety profile of belatacept was comparable to other CNI- or steroid-avoiding regimens. Belatacept-based
cyclosporine with similar rates of adverse events, including therapy also appeared to confer superior renal function,
serious infections and cancers. As there was an increased compared with TAC. Another study investigated a com-
risk of posttransplant lymphoproliferative disorder in EBV- bined therapy with alemtuzumab, sirolimus, and belata-
seronegative patients, belatacept is only approved for EBV- cept, indicating that this base regimen facilitates a low rate
seropositive recipients.45,240–242 Surprisingly, the rates of steroid-sensitive acute rejection and preserves excellent
and grades of acute allograft rejection were higher in the 3-year allograft function. It also facilitates a long-term
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 297

psoriasis and rheumatoid arthritis, but their immunomodu-


lating effects have clear potential in transplant indications.
The following agents have been studied in clinical transplant
trials or have received approval for clinical use in nontrans-
plant indications and have preclinical trials suggesting effi-
cacy in transplantation. These agents are discussed based on
their targeted ligand. All new antibodies under clinical devel-
opment are now humanized or fully human.1 Numerous
agents have been tested and have shown promise in preclini-
cal studies, but will not be specifically addressed here.

CD2-SPECIFIC APPROACHES
CD2, also known as LFA-2, is an adhesion molecule
expressed on T cells and natural killer cells that binds to
CD58 (LFA-3) on APCs and facilitates TCR binding and
signal transduction. It has been targeted by the rat IgG2b
Fig. 19.7 Increased rate and grade of rejection in patients treated with anti-CD2 MAb BTI-322, and more recently by siplizumab
belatacept from the BENEFIT trial. Shown are rejection rates and grades (also known as MEDI-507), a humanized IgG1 version of
for belatacept LI (less intensive)-treated patients compared with cyclo- BTI-322. BTI-322 was investigated initially as an induc-
sporine-treated patients. Belatacept is associated with more rejection
and with higher grades of rejection.
tion and rescue agent for deceased donor renal and hepatic
allografts, and for graft-versus-host disease, and was shown
therapy of belatacept monotherapy in selected recipients to have biologic activity and to give results consistent with
of living donor kidneys.200 Belatacept monotherapy is par- standard therapies available at the time.168,248–250
ticularly attractive for nonadherent EBV-seropositive ado- Clinical trials in psoriasis using siplizumab began in
lescents. Furthermore, anecdotal reports have shown that 1999 and were met with an unexpected propensity toward
adolescents who are nonadherent with their oral immu- agent immunogenicity.145 This agent has been used in
nosuppression will comply with a monthly belatacept infu- nonhuman primate transplant tolerance trials with suc-
sion.246 A pharmacokinetic, safety, and tolerability study of cess in mixed chimerism-directed approaches and human
belatacept in adolescents has recently been completed2 and trials are ongoing.2,251 It has been used clinically as part of
a conversion study is imminent. a nonmyeloablative conditioning regimen to achieve mixed
hematopoietic chimerism.252 Siplizumab has also been
Administration and Adverse Effects investigated in phase I and II trials for T lymphocytic malig-
At present, belatacept is approved for use in combination nancies, graft-versus-host disease, and psoriasis.2
with steroid and MMF maintenance immunosuppression. It Alefacept is a human fusion protein of the CD2 ligand
is given intravenously via peripheral vein, initially at a dose (CD58, LFA-3) with IgG1 shown to inhibit T cell prolif-
of 10 mg/kg at increasingly spaced intervals, eventually eration. Its administration has also been shown to have
leading to monthly dosing. Maintenance doses are typically a relative selective depleting effect on effector memory T
5 mg/kg monthly. Belatacept is generally well-tolerated. cells, the same cells that have been relatively spared by
Infusion reactions are mild and rare, which can include fever other depleting MAbs and polyclonal preparations.17,253
and headache. This improves with acetaminophen premedi- It gained increased attention more recently in experimen-
cation. Belatacept effects are primarily related to its immuno- tal transplantation. Alefacept is currently approved for the
suppressive effect, which is most evident in its ability to inhibit treatment of plaque-like psoriasis. Preclinical trials in non-
de novo infectious responses. Bacterial urinary tract infec- human primate transplantation have shown that alefacept
tions, pneumonias, and pyelonephritis are most common, has minimal effect on graft survival when used alone, but
but occur infrequently. Additionally, viral infections against that it does extend graft survival when used with adjuvant
EBV and CMV can develop. For this reason, use of belatacept therapies.254–256 In particular, its pairing with belatacept
is contraindicated in EBV-negative recipients, as the risk of was shown to eliminate belatacept-resistant memory T cells
PTLD increases considerably in association with primary and facilitate belatacept-based maintenance therapy in
EBV infection. CMV prophylaxis is also recommended. nonhuman primates and in vitro in humans.255,257 More-
over, it has been effective in promoting immunosuppres-
sion-free renal allograft survival in nonhuman primates.258
Monoclonal Antibodies and Alefacept was studied in a phase II trial pairing it with
TAC-based maintenance immunosuppression.259 As the
Fusion Proteins in Clinical addition of alefacept was associated with an increased risk
Transplantation Investigation of malignancy and no clear efficacy benefit, its development
in transplantation was halted.
The promise of MAb therapy has led to the development of a
rapidly expanding number of antibodies and fusion proteins CD3-SPECIFIC ANTIBODIES
targeting a wide variety of surface molecules.247 Several of
these agents have shown efficacy in large-animal transplant Targeting CD3 is a proven strategy, as shown by the suc-
models and in early clinical transplant trials. Even more cess of OKT3. Significant effort has been directed toward
have been developed for autoimmune indications, such as modernizing the anti-CD3 approach to avoid the many side
298 Kidney Transplantation: Principles and Practice

effects associated with CD3 activation. Several CD3-specific of cell–cell communication, or signal transduction through
antibodies, including huOKT3γ1 (MGA031, teplizumab), CD4 may be mechanistically relevant. Two humanized
aglycosyl CD3 (otelixizumab), visilizumab (HuM291), and anti-CD4 preparations have shown significant prolonga-
foralumab (28F11-AE, NI-0401), have been humanized tion of nonhuman primate renal allograft survival.276,279
and otherwise engineered to eliminate their undesirable Initial clinical transplantation trials using anti-CD4 MAbs
activating properties and immunogenicity.260–263 Phase employed murine-derived antibodies, including OKT4A,
I studies have indicated that modified versions of a CD3- BL4, MT151, and B-F5.275,284,285 Predictably, these agents
specific antibody can achieve T cell depletion without the were subject to immune clearance, but nevertheless were
confounding problems of cytokine release or an antibody shown to lead to CD4+ T cell clearance. Regardless, patients
neutralization. Phase II trials using visilizumab in mar- experienced rejection rates of 50%, and the agents were
row transplantation have shown initial efficacy against not sufficiently efficacious to warrant further development.
graft-versus-host disease,264 and teplizumab has shown Subsequent trials investigating the humanized OKT4A286
promise as a prophylactic agent in new-onset diabetes mel- and the chimeric cM-T412287 have been evaluated in con-
litus.265,266 These studies have shown that the side effects junction with cyclosporine-based maintenance therapy in
related to OKT3 use are not inherent in CD3-directed ther- kidney and heart transplant recipients.288 In both cases, the
apies, opening the door for more refined targeting of this antibody was well tolerated, and treated patients had low
receptor complex. Currently, teplizumab has completed a rates of rejection, suggesting that this approach is promis-
single clinical study in islet transplantation, the results of ing. Antibody responses toward the remnant murine por-
which have yet to be reported.2 Visilizumab exhibited ini- tions of the MAb, however, were surprisingly frequent.
tial potential in phase III clinic trials for ulcerative colitis. CD4+ T cell depletion was not achieved using OKT4A, but
Unfortunately, it was associated with cytokine release syn- was common with cM-T412.
drome, infections, and vascular/cardiac symptoms, and The mouse antihuman CD4 MAb, Max.16H5, has
further development was terminated. Foralumab, a fully been tested in pilot fashion as a clinical rescue agent.289
humanized, oral anti-CD3 antibody, has been shown to Max.16H5 depletes CD4+ T cells and was associated with
increase regulatory T cells and decrease disease activity reversal of rejection in most treated patients. Neutraliz-
in patients with resistant chronic hepatitis C infection and ing antibodies were not detected. No trials investigating
nonalcoholic steatohepatitis (NASH).267,268 A phase III humanized anti-CD4 MAbs have been reported for rescue
clinical trial will soon begin enrollment for NASH and type therapy.
2 diabetes mellitus.2 Its use to treat inflammatory and auto- Many human or humanized CD4-specific MAbs, includ-
immune disease shows promise and may have applications ing HuMax-CD4 (zanolimumab), TNX355, MT412, and
in transplantation. 4162 W94,290–292 have been evaluated in phase I, II, and
III trials for nontransplant indications, such as psoriasis
and rheumatoid arthritis, as well as oncologic indications.
CD4-SPECIFIC ANTIBODIES
These studies have shown that CD4-specific antibodies can
CD4 is a cell surface glycoprotein that binds to a mono- influence immune responses and that their use is relatively
morphic region of MHC class II molecules and in doing so safe in humans. Currently, there are no active anti-CD4
stabilizes the interaction between the TCR and MHC class MAb trials registered in transplantation.2
II. It is expressed on approximately two-thirds of periph-
eral T cells and has partially defined several functional COSTIMULATION-BASED THERAPIES
T cell subsets, including helper T cells and T regulatory
cells. CD4 is also expressed by peripheral monocytes and Interest in the costimulation pathways as targets for
other APCs, where its function is poorly characterized. It immune manipulation has exploded in recent years.293
likely plays a crucial role in facilitating cell-to-cell com- Generally, these agents interfere with pathways that influ-
munication among lymphoid cells, and it has lesser effects ence the outcome of antigen binding to the TCR. Costimula-
on physiologic effector functions. Given its central role in tory molecules can exert positive or negative influences on
cellular immune responses, CD4 has long been a target for the efficiency of antigen presentation and recognition and
immune manipulation, and several antibodies have been alter the threshold for activation of naive T lymphocytes
tested in transplantation. Generally, the efficacy has been without having a primary activating or inhibitory function.
exceptional in defined rodent models and modest in more Costimulatory molecule manipulation influences only cells
clinically relevant settings; this may relate to the growing with ongoing TCR activation and should affect only cells
recognition that CD4+ T cells have a potential role in tem- actively undergoing antigen recognition; this may allow for
pering immune responses.149,269,270 antigen-specific immune manipulation.
Many studies have shown that anti-CD4 antibody The most studied costimulatory receptor on T cells is
induction dramatically inhibits the development of acute CD28. It has two known ligands, CD80 (B7-1) and CD86
rejection in rodents, particularly when combined with (B7-2), both of which are expressed on APCs. CD28 is con-
a supplementary donor antigen, such as donor-specific stitutively expressed on most T cells and on ligation reduces
transfusion.271–274 Given that the distribution of MHC the threshold for TCR activation.294 CD152 (cytotoxic T
class II molecules differs substantially between rodents and lymphocyte associated antigen 4 [CTLA-4]) is an induced
humans, however, these studies have not been predictive of molecule expressed on T cell activation that is structurally
the anticipated effect in humans. Depleting275–277 and non- similar to CD28 and competitively binds CD80 and CD86,
depleting278–283 antibodies have shown an effect in experi- transmitting an inhibitory signal that acts to terminate the
mental models, suggesting that cell elimination, disruption immune response.295 CD28 and CD152 serve reciprocal
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 299

roles, both stimulated by the B7 molecules and facilitating been shown in numerous nonhuman primate studies to
(CD28) or quelling (CD152) a T cell response. prevent kidney and liver allograft rejection, particularly
An additional receptor–ligand pair that has gained con- when used in combination with tacrolimus.315–319 Inves-
siderable attention involves CD40 and CD154. CD154, also tigational interest in CD154 manipulation also remains
known as CD40 ligand, is expressed on activated T cells and intense.
other cells, including platelets.296–299 CD40 is expressed on A cocktail of two humanized MAbs specific for the B7
APCs. Although the specific effect of CD154 on T cells is molecules CD80 and CD86 has been shown to facilitate pro-
incompletely defined, CD40 has a major influence on APC longed renal allograft survival in nonhuman primates.320
activation. CD40 ligation leads to marked APC activation, These antibodies reached clinical trials in organ transplan-
including increased expression of B7 molecules and MHC, tation and were shown to have initial safety in humans.
and stimulatory cytokine production, greatly facilitating Their development has not been pursued.
antigen presentation.300 CD154 is released from the alpha Many other costimulatory molecules have been inves-
granules of activated platelets and acts locally to greatly tigated successfully in animal models, but none has yet
augment alloimmune responses.301 It can serve as the sole been exploited as a target in the clinic.2,321 Costimulatory
source of CD154 responsible for rejection.302 CD154 exists molecules can be targeted with blocking MAbs to inhibit
as a large inducible reservoir that can be triggered by plate- their stimulatory effects. Because it is difficult to determine
let activation and augment antigen presentation at the time prospectively whether a MAb is stimulatory or inhibitory
of a traumatic injury, including a transplant procedure. Its in vivo, and because costimulatory molecules have stimu-
release is local and governed by alpha degranulation.301 latory and inhibitory effects, it has been challenging to
Seminal studies using small animals and nonhuman pri- find therapeutically reliable agents. For example, CD152
mates revealed that simultaneous blockade of the CD28 and and CD154 are upregulated on activated T cells and may
CD40 pathways with CTLA-4 Ig and anti-CD154 inhibited serve as targets for selective elimination of activated effector
T cell responses, provided durable protection from allograft cells.322 Selective targeting of CD152 with the MAb ipilim-
rejection and held promise for both induction and mainte- umab has been used to augment immunity in the setting of
nance therapy.303–305 Furthermore, costimulation block- metastatic melanoma, with salutary oncologic results bal-
ade with belatacept, a high-affinity CTLA4 fusion protein, anced by considerable induction of autoimmune side effects
combined with mammalian target of rapamycin (mTOR) including enteritis and vasculitis.323,324 This situation likely
inhibition promoted a sustained CNI-free, steroid-sparing relates to the fundamental role that costimulation mol-
immunomodulatory regimen for kidney transplantation in ecules have in general immunity and immune homeostasis.
nonhuman primates.306 However, long-term host allograft Similarly, severe septic-like responses have been reported
tolerance with costimulation blockade has remained a after the administration of TGN1412, a CD28-specific MAb
challenge. The combination of anti-B7-1 and B7-2 antibod- tested in phase I trials.39 A fundamental balance between
ies with either cyclosporine A or sirolimus was not sufficient the two B7-specific T cell molecules CD28 and CD152
to induce long-term tolerance.128,166,296,307–309 Moreover, seems to be required to avoid dysregulated autoimmunity.
a regimen consisting of donor antigen-pulsed autologous However, the conceptual promise of CD28-specific blockade
regulatory dendritic cell infusions, CTLA 4 IgG, and tapered has led to the development of numerous agents, including
sirolimus did not show a beneficial effect on graft survival in single-chain domain-specific antibodies that have proven
nonhuman primates.310 Although most experimental use themselves efficacious in nonhuman primate transplanta-
of MAbs directed against costimulatory molecules in trans- tion.325,326 Their clinical translation is anticipated.
plantation has focused on tolerance induction (elimination
of a need for any maintenance therapy), the clinical focus CYTOKINE-BASED APPROACHES
has been on pairing costimulation-directed biologics with
maintenance minimization strategies, particularly calci- Sequestration of cytokines using MAbs has long been con-
neurin-sparing approaches. Agents interfering with the templated as a therapeutic strategy in many inflammatory
CD28/B7 and the CD40/CD154 pathways have reached diseases. Although many cytokine-specific agents have
clinical trials. been developed, only TNF-α-specific agents have gained
Two humanized MAbs specific for CD154, hu5c8, and widespread clinical use. TNF-α is a cytokine produced by
IDEC-131, have been shown in nonhuman primates to many immune cells that is ubiquitously present in most
prevent acute rejection for months to years without addi- inflammatory responses and has numerous general pro-
tional immunosuppression and have been paired with siro- inflammatory effects, including increased chemotaxis,
limus monotherapy and donor-specific transfusion to lead vascular permeability, and fever. It has been considered
to operational tolerance in some cases.303,304,311,312 Early as an attractive target for many inflammatory aspects of
human trials with hu5c8 were hindered by unimpressive transplantation, including depletion-associated cytokine
efficacy and concerns for thromboembolic risk.313 release syndrome, ischemia-reperfusion injury, and rejec-
CD154-specific therapies have not been studied clinically tion. Three TNF-α-specific agents are currently approved
in recent years, and most preclinical attention has turned for nontransplant conditions, and their use in transplanta-
toward intervention with CD40 as opposed to CD154.314 tion is emerging, particularly in islet transplantation.
Pursuant to CD40 blockade, ASKP1240 (also known Infliximab is a chimeric IgG1 MAb that binds to cell-
as 4d11) and CFZ533 have reached the clinical arena. bound and circulating TNF-α, sequestering it from the TNF
ASKP1240 has completed phase I and II trials, whereas receptor and inhibiting TNF-dependent proinflammatory
CFZ533 is undergoing phase II trials.2 ASKP1240 and effects. It has been developed for the treatment of numerous
CFZ533 are fully human CD40-specific MAbs that have autoimmune disorders, including rheumatoid arthritis (its
300 Kidney Transplantation: Principles and Practice

primary approved clinical use), psoriasis, Crohn’s disease, pathway greatly facilitates initial lymphocyte recruitment
and ulcerative colitis.327,328 It has been used in pilot studies at sites of injury and inflammation.345 Adhesion pathways
of many transplant indications, including renal, bone mar- have been studied in several preclinical settings, including
row, intestinal, and islet transplantation, with suggestive rodents346,347 and nonhuman primates,40,348,349 with sur-
success. Its predominant therapeutic effect in transplanta- vival being markedly prolonged in rodents and prolonged
tion seems to be to limit paracrine cytokine-mediated acti- 30 days in primates. Data pairing the LFA-1-specific MAb
vation within the graft and to mute the clinical sequelae of TS1/22 with belatacept has led to exceptionally prolonged
rejection without altering the overall infiltrate of inciting survival in nonhuman primate islet transplantation.350
allosensitization.329–331 Similarly, adalimumab is a TNF- Enlimomab, a murine anti-CD54 MAb, was successfully
α-specific MAb approved for the treatment of psoriatic tested in a phase I trial involving high-risk deceased donor
arthritis.328 kidneys351 and subsequently evaluated in a placebo-con-
Etanercept is a soluble recombinant TNF receptor–IgG trolled phase II study combined with conventional triple-
fusion protein that acts to absorb soluble TNF-α and limit drug maintenance therapy.352 No significant difference
its availability in the circulation. It is approved for the treat- was detected between the treated and the placebo groups,
ment of rheumatoid arthritis and has been increasingly and further development was not pursued. Similarly, odu-
evaluated for a role in the treatment of graft-versus-host limomab, a murine anti-LFA-1 MAb, was studied as an
disease.332 The use of etanercept in islet transplantation induction agent compared with ATG-R in renal transplan-
has been reported.326,333,334 tation, with no significant difference being found between
Golimumab is a fully human TNF-α-specific MAb that is the groups.353 A single rescue trial using the anti-LFA-1
in phase II trials for rheumatoid arthritis. No reports have murine MAb 25-3 failed to show efficacy.354
been made of this agent in transplantation, although there The most promising and well-studied LFA-1-specific
are numerous trials in autoimmune indications.2 MAb is efalizumab. Efalizumab (Raptiva) is a recombinant
Several other cytokine-directed MAbs are now approved humanized MAb that binds to human CD11a and inhib-
for immune indications outside of transplantation and are its the LFA-1/ICAM-1 interaction.355,356 Efalizumab has
likely to have relevance to transplantation. Ustekinumab been tested in phase I/II studies for renal transplantation
is a humanized MAb specific for the p40 subunit common where it was paired with conventional triple immunosup-
to the receptors for both IL-12 and IL-23.335 It is approved pression. In that setting, the combined regimen was overly
for clinical use in the treatment of psoriasis,336 particu- immunosuppressive and its development waned.357 It
larly in patients who have failed therapy with etanercept. reemerged recently in two clinical islet trials as the center-
The IL-12/23 axis is increasingly recognized as relevant piece maintenance immunosuppressant. In these trials, the
in transplantation, particularly in the setting of costimu- efalizumab-based regimen was shown to facilitate initial
lation blockade resistant rejection, and it is likely that engraftment and function, and to prevent islet rejection suc-
ustekinumab will be evaluated as an adjuvant therapy in cessfully.358,359 Trials based on efalizumab as a calcineurin
combination with costimulation blockade based strate- alternative were initiated in kidney transplantation but had
gies in the foreseeable future. Similarly, tocilizumab is an to be halted when the maker of efalizumab withdrew the
IL-6 receptor MAb approved for use in rheumatoid arthri- drug from the market.2 It was used clinically and approved
tis.337 Given the role of IL-6 in alloantibody production, by the FDA for treatment of mild-to-moderate psoriasis.
tocilizumab has been used to treat chronic antibody-medi- However, its use was associated with a low incidence of pro-
ated rejection and as part of a desensitization regimen in gressive multifocal leukoencephalopathy (approximately 1
patients awaiting kidney transplantation with promising in 10,000 exposures), which was cited as an undue risk for
results.2,338,339 Additionally, belimumab, a MAb that inhib- patients with psoriasis. In general, this risk profile, although
its BAFF, a cytokine critical to B cell activation, prolifera- perhaps adverse for psoriasis, is acceptable in kidney trans-
tion, and differentiation, mitigated autoantibody responses plantation, and there is genuine hope that this agent will
in systemic lupus erythematous.340 Both intravenous and resurface for use in transplant indications in the future.
subcutaneous formulations are currently FDA-approved, Other integrin-based adhesion molecules have been
and current trials in both sensitized and nonsensitized kid- shown to be clearly involved in peripheral immune
ney transplant recipients are either completed with results responses, the most studied of which is VLA4 (α4-integrin).
pending or ongoing.2 The MAb natalizumab targets this molecule and has gained
FDA approval for the treatment of multiple sclerosis.360 It
was subsequently withdrawn from the market due to an
TARGETING CELL ADHESION
elevated risk of progressive multifocal encephalopathy, but
Given the fundamental requirement for adhesion mol- returned with a more robust warning relative to this side
ecules in most inflammatory responses, there has been effect. VLA-4-specific MAbs have been shown to attenuate
long-standing interest in blocking adhesion interactions to costimulation blockade resistant rejection in mice,361 and
prevent platelet and leukocyte adherence and infiltration. promising preliminary results in nonhuman primates have
As discussed previously, polyclonal antibodies are thought been observed. Its availability for off-label use in the clinic
to bind to and inhibit some adhesion molecules. Several may facilitate clinical transplant trials.
MAbs have been developed to target adhesion pathways.341 PSGL1-Ig (YSPSL) is a fusion protein combining the
Among the most prominent is the LFA-1/ICAM-1 path- extracellular domains of P-selectin glycoprotein ligand-1
way. LFA-1 (the heterodimer integrin molecule CD11a/ (CD162) with the Fc portion of IgG1. CD162 is a ligand
CD18) is expressed on mature T cells and binds to ICAM-1 for P-selectin, E-selectin, and L-selectin, all of which have
(CD54) expressed on APCs and endothelial cells.342–344 The been shown to facilitate leukocyte and platelet adhesion.
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 301

Because cell adhesion has been implicated as a primary


TARGETING COMPLEMENT
event in reperfusion injury and in allorecognition, this
drug has been contemplated as a therapy to limit the effect Proteins of the complement cascade have long been known
of events occurring during initial implantation. Treatment to be crucial in mediating antibody-associated cytotox-
with PSGL1-Ig has been shown to attenuate ischemia- icity.377 Many approaches have been contemplated to
reperfusion injury, most prominently in rodent models of achieve complement elimination in the setting of antibody
hepatic warm ischemia.362–364 Recent testing in a phase I/ presensitization, including plasmapheresis and intravenous
II evaluation in liver transplantation has shown that this immunoglobulin administration. More recently, it has been
agent attenuates the clinical and biochemical syndrome shown that complement, specifically that produced locally
of hepatic ischemia-reperfusion injury.365 In kidney trans- within the kidney itself, is a contributing factor facilitat-
plantation, PSGL-1Ig was well tolerated and safe, though it ing peripheral T cell maturation and rejection.378 Poly-
did not decrease the rate of delayed graft function requiring morphisms in complement expression have been shown to
dialysis.366 influence the incidence of rejection and renal allograft sur-
vival in ways not previously recognized.379
TARGETING THE T CELL RECEPTOR Three complement-specific agents have been used clini-
cally and have been shown to be biologically active with
T cells bind their cognate antigen through their heterodi- promise for application in transplantation. Eculizumab is a
meric glycoprotein TCR. There are two general forms, humanized MAb specific for C5a, a key initiation factor in
an α/β form, expressed on 95% of peripheral T cells and complement membrane attack complex formation. It has
responsible for specifying most alloimmune responses, been shown to be effective therapy for paroxysmal nocturnal
and a γ/δ form, involved in innate immune responses and hemoglobinuria and atypical hemolytic uremic syndrome,
appearing late in allograft rejection.367 The TCR is a result and it is FDA-approved for both indications in children and
of somatic gene rearrangement similar to that seen in anti- adults.2,380–385 Numerous single-center studies and case
body formation, and the specificity of each T cell can be reports have previously shown that eculizumab, when used
defined by its individual TCR. Rejections based on specific as part of a multimodal treatment strategy, can facilitate the
TCR/MHC interactions select for specific TCR types, show- prevention or reversal of numerous complement-mediated
ing that each MHC mismatch is recognized by a few clones, maladies in transplantation, including hemolytic uremic
rather than by the entire T cell repertoire.368 Although this syndrome (HUS) and antibody-mediated rejection.386–392
finding fostered initial enthusiasm for targeting antigen- Although the results with eculizumab for desensitization
specific T cells through custom MAbs specific for a given and chronic antibody-mediated rejection have been disap-
TCR, this approach has been deemed impractical given pointing,393,394 its use for selected cases of acute antibody-
the vast number of TCRs generated during T cell matura- mediated rejection and long-term recurrent atypical HUS
tion and their variable cross-reactivity with variable MHC remains promising.390,392,395,396 Accordingly, it is likely
polymorphisms. Nevertheless, the success of targeting TCR- that this agent will become an established part of the trans-
associated proteins such as CD3 has generated some inter- plant armamentarium in the future for these indications.
est in targeting monomorphic portions of the TCR directly. TP-10 (soluble complement receptor type 3) is a recombi-
The realization that TCR signaling is required for T cell nant soluble protein that binds and inactivates the central
apoptosis and regulation has made preservation of the TCR activating component of the complement cascade, C3. It
a competing strategy. has been used in numerous preclinical settings and shown
T10B9, also known as Medi-500, is a murine IgM specific to be effective in preventing humoral xenograft rejection in
for a monomorphic determinant on α/β and γ/δ TCRs. It is a pig-to-nonhuman-primate model.397 It was investigated
effective in mediating T cell depletion in vitro and in vivo369 in a clinical trial for its role in preventing cardiopulmonary
and has been studied as a rescue and induction agent in bypass-related complications, with disappointing results.2
renal and cardiac transplantation.370,371 In both trials, Despite its clear ability to arrest C3-mediated complement
the antibody-mediated T cell depletion was well tolerated. activation, it has not been systematically investigated in
Its efficacy as a rescue agent seemed to be similar to that of clinical transplantation.
OKT3, and the cardiac trial suggested efficacy as an induc- Inhibition at the level of C3 and C5 does not prevent the
tion agent. Nevertheless, the agent has not been developed generation of important upstream inflammatory media-
further in organ transplantation, likely as a result of com- tors. Therefore therapeutic strategies for antibody-mediated
parably effective humanized MAbs. T10B9 has been studied rejection have recently focused on upstream targets of the
as a conditioning agent and ex vivo depletional agent for complement pathway such as the C1 complex, which con-
bone marrow transplantation.372,373 sists of C1q, C1r, and C1s, and is the first to interact with
TOL101 is an IgM MAb specific for the α/β TCR. It donor-specific antibody on the endothelium. Preclinical
has been shown to have numerous depletional and non- in vitro studies have shown that blockade of C1 prevented
depletional effects on human T cells.374,375 In kidney antibody-mediated complement activation using sera from
transplantation, TOL101 used for induction therapy was sensitized patients.398,399 Pilot studies with C1 inhibitor and
well-tolerated overall and had an acceptable rate of acute anti-C1s monoclonal antibody have shown modest results
cellular rejections (13.9%) without development of donor- with decreasing delayed graft function, short-term preven-
specific antibodies. Moreover, TOL101 was not associated tion of chronic transplant glomerulopathy, and reduction of
with patient or allograft loss after 6 months.376 Long-term C4d deposition in renal allografts.400–403 Multicenter stud-
safety and efficacy studies are required to assess its clinical ies to confirm efficacy are ongoing.2 Inhibiting the interac-
utility. tion between complement and donor-specific antibody is
302 Kidney Transplantation: Principles and Practice

an alternative strategy for reducing complement-mediated expression is augmented on activated alloimmune-respon-


allograft damage. Recently a novel approach using Ig-G sive T cells. CD7 has been thought to be an attractive target
enzyme derived from Streptococcus pyogenes (IdeS) as an for MAbs, offering the possibility of alloimmune-activated T
adjunctive therapy for desensitization was associated with cell-specific depletion.
significant antibody reduction and allowed for HLA-incom- SDZCHH380 is a chimeric mouse antihuman CD7 IgG1
patible transplantation in 24 of 25 patients.404 IdeS is an that has been studied in initial clinical renal transplant tri-
endopeptidase which cleaves human IgG into F(ab′)2 and als.418 SDZCHH380 induction was prospectively compared
Fc fragments, inhibiting complement-dependent cytotox- with OKT3 induction, with comparable results. At 4 years,
icity and antibody-dependent cellular cytotoxicity. These SDZCHH380-treated patients had good allograft function
promising findings are being validated in an ongoing phase and did not develop neutralizing antibodies.419 Additional
II trial.2 development has not been reported.

Other Experimental Antibodies TARGETING CD8


and Fusion Proteins CD8 is a glycoprotein present on approximately one-third
of T cells in lieu of CD4. Similar to CD4, it binds to a mono-
Almost all surface molecules expressed by leukocytes have morphic region MHC, although it binds to class I rather
been considered for therapeutic targeting. Many have been than to class II antigens. CD8 defines cytotoxic effector
formally investigated in early clinical trials without suffi- cells and perhaps subsets of natural killer and regulatory
cient promise to warrant additional clinical development. cells. It facilitates binding between the TCR and class I
Others have significant promise in advanced preclinical set- molecules and is important in protective immune lysis of
tings but have yet to be tested in humans. This section will virally infected parenchymal cells. CD8+ T cells are known
call out additional agents in which there is some clinical to infiltrate allografts and to participate in allograft rejec-
experience. Knowledge of these agents is useful for a com- tion.420 Despite this demonstrated role in rejection, CD8 has
plete understanding of the field. not been successfully targeted in transplantation, perhaps
because CD8+ T cells are recruited late in an alloimmune
TARGETING CD5 response and have less regulatory control over immune
responses than CD4+ T cells. The CD8-specific MAb anti-
CD5 is an adhesion molecule constitutively expressed on Leu2a has been shown to deplete peripheral blood CD8+
T cells and a subset of B cells.405 It binds to CD72 and is cells in humans; however, when tested as a rescue agent, it
thought to regulate the intensity of antigen receptor sig- had limited effects in reversing renal allograft rejection.421
nal transduction. Its primary function may be costimula- More recently, 76-2-11, a mouse antiswine CD8-specific
tory or inhibitory, but mounting evidence suggests that it MAb, has been shown to delay the onset of cardiac allograft
has a role in self-tolerance. XomaZyme-CD5 Plus (Xoma- vasculopathy in a miniature swine model of cardiac trans-
Zyme H65) is a ricin-conjugated CD5-specific MAb that plantation, suggesting that there may be a limited role for
has been evaluated in clinical trials to prevent graft-versus- this approach.422 Additionally, ex vivo depletion of CD8+ T
host disease after bone marrow transplantation, rheuma- cells with anti-Leu2a has been investigated as a means of
toid arthritis, and systemic lupus erythematosus, without reducing graft-versus-host disease, with promising prelimi-
apparent efficacy.406–409 As the biology of this molecule is nary results.423 Anti-CD8 induction has not been investi-
better understood, its reevaluation as a therapeutic target gated clinically in solid-organ transplantation.
may be warranted.
TARGETING CD45
TARGETING CD6
CD45 is a transmembrane protein tyrosine phosphatase
The human CD6 is a cell surface glycoprotein expressed by expressed on T cells. It is physically associated with the
T cells and a subset of B cells. It has been shown to act as TCR and facilitates the signal transduction function of CD3
a costimulatory molecule and can stimulate T cells when through interactions with the zeta and zeta-associated pro-
crosslinked with CD28.410 Anti-CD6 MAbs inhibit the tein-70 components of CD3.424 CD45 exists in several iso-
interaction of CD6 with its ligand, activated leukocyte cell forms (CD45RA, CD45RB, and CD45RO) that result from
adhesion molecule.411 Anti-T12, an anti-CD6 MAb, has RNA spliced variants, and these are differentially expressed
been evaluated clinically, but has not shown consistent effi- on T cells with varying degrees of maturity and activation.
cacy.412 An anti-CD6 has been used ex vivo to T cell deplete Of these, CD45RB has been most aggressively targeted as T
bone marrow before its use in marrow transplantation, cells expressing high amounts of this isoform skew toward
with promising short- and long-term results.413,414 More an aggressive T helper type 1 phenotype. CD45RB-specific
recently, the anti-CD6 antibody itolizumab has exhibited MAbs have been shown to induce transplant tolerance in
short-term efficacy for rheumatoid arthritis.415 some rodent models and to prolong the survival of nonhu-
man primate renal allografts significantly.424,425 Several
TARGETING CD7 antibodies, including those linked to the yttrium isotope
90Y, have entered phase I trials for oncologic indications,2
CD7 is a cell surface costimulatory molecule expressed on and at least one humanized anti-CD45RB has been antici-
human T and natural killer cells and on cells in the early pated to enter early-phase clinical trials in renal transplan-
stages of T, B, and myeloid cell differentiation.416,417 Its tation.335 ChA6, a chimeric MAb binding CD45RB and
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 303

CD45RO (an isotype found on memory T cells), has been transplantation. The trials performed to date have shown,
shown to prevent islet allograft rejection in mice by deleting however, that antibodies produce a modest benefit over
memory T cells and being permissive for the persistence of regimens with CNIs, antiproliferative agents, and steroids,
protolerant regulatory T cells.426,427 or that they are associated with increased morbidity. Nev-
ertheless, it is appropriate to consider antibody induction
as emerging from an adolescence of sorts, and less morbid
IMMUNOTOXINS
target strategies and reduced maintenance regimens are
Antibodies that have been joined either chemically or genet- expected to improve the side effect profile of antibody induc-
ically with a specific cytotoxic agent (e.g., ricin or diphtheria tion schemes. The use of these agents to facilitate CNI avoid-
toxin) have been termed immunotoxins.428 These com- ance or allow weaning to monotherapy, particularly when
pounds have the specificity of MAbs but can exert a cyto- paired with costimulation blockade based therapies, such as
toxic effect beyond that related to complement or ADCC. belatacept or anti-CD40 blockade, is likely to be a focus of
Many immunotoxins are now being investigated as tumor- the coming several years.
specific cytotoxic agents for malignancies and have been The optimal use of antibody induction is still being deter-
shown to have potent antitumor effects. Two CD25-specific mined, but it is increasingly clear that the benefits derived
immunotoxins currently in clinical trials for lymphoblas- from antibodies will be determined by their appropriate
tic leukemia, LMB-2 and RFT5.dgA, have shown the abil- application. Modern immunosuppressive regimens should
ity to clear CD25+ cells effectively from circulation.2,429,430 be individualized, specifically pairing induction agents
These agents could be envisioned to perform in a means based on their mechanism of action to a specific clinical
analogous to the CD25-specific MAbs currently available, need, and combining them with complementary mainte-
with a more potent depletional effect rather than acting pre- nance therapies.
dominantly through steric inhibition of CD25. Similarly, a The future of transplantation continues to be cloaked by
CD22-specific immunotoxin for CD22+ lymphoblastic leu- a need for more specific therapies with broader therapeutic
kemia has completed phase I trials with promising results indices. Antibodies are highly specific and have proven to
and might be envisioned as an agent similar to other B-cell- be safe and effective drugs whose side effects are generally
specific MAbs such as rituximab.2,431,432 confined to the specific effects of the target antigen bound.
Although immunotoxins have not been clinically tested Although the early hopes of clinicians have been slow to
in transplantation, ample preclinical data suggest that they materialize, the technology associated with antibody design,
have great therapeutic potential. Specifically, a macaque construction, and production has consistently improved
CD3-specific diphtheria immunotoxin, FN18-CRM9, has to yield a diverse array of agents to be tested and added to
been used in nonhuman primate renal transplantation, with the transplant armamentarium. The future is likely to see
remarkable success.43,433 Treatment with FN18-CRM9 almost exclusive use of humanized or human antibodies
induces a rapid 3-log-fold depletion of T cells in the periph- and fusion proteins as opposed to xenogeneic protein con-
eral circulation and in the secondary lymphoid organs. structs. Past problems of antigenicity and severe cytokine
Rhesus monkeys so treated before transplantation experi- release effects are surmountable, and as the targeted anti-
ence markedly prolonged allograft survival with no other gens become more rationally selected based on the growing
maintenance immunosuppression, and a significant pro- understanding of biology, the antibodies and fusion proteins
portion survive for years after T cell repopulation. Although are expected to continue to establish themselves as crucial
most of these animals eventually develop chronic allograft agents not only for induction and rescue but also, impor-
nephropathy,434 the induction effect is impressive, and it tantly, for maintenance therapy. Trials are continuing to
has served as the conceptual inspiration for many clinical explore this facet of antibody and fusion protein administra-
trials using T cell depletion.126,182,189 Because most adults tion. Additionally, the use of antibody combinations may
have antibodies against diphtheria toxin, this approach become an attractive way of manipulating the immune
has not been successfully transferred to a human-specific response and ensuring more reliable drug delivery, espe-
MAb. More recently, an immunotoxin against donor MHC cially in the context of nonadherence. Transplant clinicians
class II molecule conjugated with plant-derived ribosomal will need to become increasingly aware of immune thera-
inactivating protein gelonin resulted in long-term allograft pies developed for autoimmune and malignant indications.
survival, increased T regulatory cells, and decreased donor-
specific antibody formation in a murine renal model.435 References
This provides a promising approach for future development.
1. Reichert J, Rosensweig C, Faden L, Dewitz M. Monoclonal antibody
successes in the clinic. Nat Biotechnol 2005;23:1073–8.
2. U.S. National Library of Medicine. Find a study. Available online at:
Conclusion https://clinicaltrials.gov. Accessed September 14, 2018.
3. Ecker D, Jones S, Levine H. The therapeutic monoclonal antibody
market. MAbs 2015;7:9–14.
Antibodies are now established as valuable agents for the 4. Cosimi A, Burton R, Colvin R, et al. Treatment of acute renal
treatment and prevention of allograft rejection. Currently, allograft rejection with OKT3 monoclonal antibody. Transplanta-
several polyclonal and monoclonal anti-T cell antibod- tion 1981;32:535–9.
ies have proven roles in the treatment of steroid-resistant 5. Bishop G, Cosimi A, Voynow N, et al. Effect of immunosuppressive
acute rejection. The past 15 years have seen increasing therapy for renal allografts on the number of circulating sheep red
blood cells rosetting cells. Transplantation 1975;20:123–9.
justification for the use of antibodies as induction agents. 6. Cosimi A, Wortis H, Delmonico F, Russell P. Randomized clinical
Antibody induction has been shown to be an effective trial of antithymocyte globulin in cadaver renal allograft recipients:
means of achieving very low rates of acute rejection in renal importance of T cell monitoring. Surgery 1976;80:155–63.
304 Kidney Transplantation: Principles and Practice

7. Davis R, Nabseth D, Olsson C, et al. Effect of rabbit ALG on cadaver 35. Raghavan M, Bjorkman P. Fc receptors and their interactions with
kidney transplant survival. Ann Surg 1972;176:521–8. immunoglobulins. Annu Rev Cell Dev Biol 1996;12:181–220.
8. Starzl T, Porter K, Iwasaki Y, et al. The use of heterologous anti- 36. Pearl J, Parris J, Hale D, et al. Immunocompetent T-cells with a mem-
lymphocyte globulins in human homotransplantation. In: Wol- ory-like phenotype are the dominant cell type following antibody-
stenholme G, O’Connor M, editors. Antilymphocyte serum. Boston: mediated T-cell depletion. Am J Transplant 2005;5:465–74.
Little, Brown; 1967. p. 1. 37. Greenwood J, Clark M, Waldmann H. Structural motifs involved
9. Cosimi A. The clinical value of antilymphocyte antibodies. Trans- in human IgG antibody effector functions. Eur J Immunol
plant Proc 1981;13:462–8. 1993;23:1098–104.
10. Niblack G, Johnson K, Williams T, et al. Antibody formation fol- 38. Wang X, An Z, Luo W, et al. Molecular and functional analysis
lowing administration of antilymphocyte serum. Transplant Proc of monoclonal antibodies in support of biologics development.
1987;19:1896–7. Protein Cell 2018;9(1):74–85. Available online at: https://doi.
11. Tatum A, Bollinger R, Sanfilippo F. Rapid serologic diagnosis of org/10.1007/s13238-017-0447-x.
serum sickness from antilymphocyte globulin therapy using enzyme 39. Suntharalingam G, Perry M, Ward S, et al. Cytokine storm in a phase
immunoassay. Transplantation 1984;38:582–6. 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J
12. Mathieu CP, Renoult E, March AKD, et al. Serum anti-rabbit and Med 2006;355:1018–28.
anti-horse IgG, IgA, and IgM in kidney transplant recipients. Nephrol 40. Kaplon R, Hochman P, Michler R, et al. Short course single agent
Dial Transplant 1997;12:2133–9. therapy with an LFA-3-IgG1 fusion protein prolongs primate cardiac
13. Kohler G, Milstein C. Continuous cultures of fused cells secreting allograft survival. Transplantation 1996;61:356–63.
antibody of predefined specificity. Nature 1975;256:495–7. 41. Larsen C, Pearson T, Adams A, et al. Rational development of
14. Farges O, Samuel D, Bismuth H. Orthoclone OKT3 in liver transplan- LEA29Y (belatacept), a high-affinity variant of CTLA4-Ig with potent
tation. Transplant Sci 1992;2:16–21. immunosuppressive properties. Am J Transplant 2005;5:443–53.
15. Group OMTS. A randomized clinical trial of OKT3 monoclonal anti- 42. Linsley P, Wallace P, Johnson J, et al. Immunosuppression in vivo
body for acute rejection of cadaveric renal transplants. N Engl J Med by a soluble form of the CTLA-4 T cell activation molecule. Science
1985;313:337–42. 1992;257:792–5.
16. Ponticelli C, Rivolta E, Tarantino A, et al. Treatment of severe rejec- 43. Knechtle S, Vargo D, Fechner J, et al. FN18-CRM9 immunotoxin
tion of kidney transplants with Orthoclone OKT-3. Clin Transplant promotes tolerance in primate renal allografts. Transplantation
1987;1:99–103. 1997;63:1–6.
17. Scheinfeld N. Alefacept: a safety profile. Expert Opin Drug Saf 44. Webster A, Wu S, Tallapragada K, et al. Polyclonal and monoclonal
2005;4:975–85. antibodies for treating acute rejection episodes in kidney transplant
18. Jaffers G, Fuller T, Cosimi A, et al. Monoclonal antibody therapy: anti- recipients. Cochrane Database Syst Rev 2017;1:CD004759.
idiotype and non-anti-idiotype antibodies to OKT3 arising despite 45. Vincenti F, Charpentier B, Vanrenterghem Y, et al. A phase III study
intense immunosuppression. Transplantation 1986;41:572–8. of belatacept-based immunosuppression regimens versus cyclospo-
19. Boulianne G, Hozumi N, Shulman M. Production of functional chi- rine in renal transplant recipients (BENEFIT study). Am J Transplant
maeric mouse/human antibody. Nature 1984;312:643–6. 2010;10:535–46.
20. Jones P, Dear P, Foote J, et al. Replacing the complementarity- 46. Vincenti F, Larsen C, Durrbach A, et al. Costimulation blockade
determining regions in a human antibody with those from a mouse. with belatacept in renal transplantation. N Engl J Med 2005;353:
Nature 1986;321:522–5. 770–81.
21. Morrison S, Johnson M, Herzenberg L, Oi V. Chimeric human anti- 47. Humar A, Ramcharan T, Denny R, et al. Are wound complications
body molecules: mouse antigen-binding domains with human con- after a kidney transplant more common with modern immunosup-
stant region domains. Proc Natl Acad Sci U S A 1984;81:6851–5. pression? Transplantation 2001;72:1920–3.
22. Weiner L. Fully human therapeutic monoclonal antibodies. J Immu- 48. Singh A, Stablein D, Tejani A. Risk factors for vascular thrombosis in
nother 2006;29:1–9. pediatric renal transplantation: a special report of the North Ameri-
23. SRTR/OPTN. 2015 annual data report. Am J Transplant 2017;17. can Pediatric Renal Transplant Cooperative Study. Transplantation
24. Szczech L, Berlin J, Aradhye S, et al. Effect of anti-lymphocyte induc- 1997;63:1263–7.
tion therapy on renal allograft survival: a meta-analysis. J Am Soc 49. Cherikh W, Kauffman H, McBri M, et al. Association of the type of
Nephrol 1997;8:1771–7. induction immunosuppression with posttransplant lymphoprolif-
25. Szczech L, Berlin J, Feldman H. The effect of antilymphocyte induc- erative disorder, graft survival, and patient survival after primary
tion therapy on renal allograft survival: a meta-analysis of individ- kidney transplantation. Transplantation 2003;76:1289–93.
ual patient-level data. anti-lymphocyte antibody induction therapy 50. Malatack J, Gartner JJ, Urbach A, Zitelli B. Orthotopic liver trans-
study group. Ann Intern Med 1998;128:817–26. plantation, Epstein-Barr virus, cyclosporine, and lymphoprolifera-
26. Hill P, Cross N, Barnett A, et al. Polyclonal and monoclonal antibod- tive disease: a growing concern. J Pediatr 1991;118:667–75.
ies for induction therapy in kidney transplant recipients. Cochrane 51. Meier-Kriesche H, Andorfer J, Kaplan B. Association of antibody
Database Syst Rev 2017;1:CD004759. induction with short- and long-term cause-specific mortality in renal
27. Gill J, Gulley M. Immunoglobulin and T cell receptor gene rearrange- transplant recipients. J Am Soc Nephrol 2002;13:769–72.
ment. Hematol Oncol Clin North Am 1994;8:751–70. 52. Penn I. Cancers complicating organ transplantation. N Engl J Med
28. Hozumi N, Tonegawa S. Evidence for somatic rearrangement of 1990;323:1767–9.
immunoglobulin genes coding for variable and constant regions. 53. Penn I. The problem of cancer in organ transplant recipients: an
Proc Natl Acad Sci U S A 1976;73:3628–32. overview. Transplant Sci 1994;4:23–32.
29. Tite J, Sloan A, Janeway CJ. The role of L3T4 in T cell activation: 54. Kirk A, Cherikh W, Ring M, et al. Dissociation of depletional induc-
L3T4 may be both an Ia-binding protein and a receptor that trans- tion and posttransplant lymphoproliferative disease in kidney recipi-
duces a negative signal. J Mol Cell Immunol 1986;2:179–90. ents treated with alemtuzumab. Am J Transplant 2007;7:2619–25.
30. Wong J, Eylath A, Ghobrial I, Colvin R. The mechanism of anti-CD3 55. Bunnapradist S, Daswani A, Takemoto S. Patterns of administration
monoclonal antibodies: mediation of cytolysis by inter-T cell bridg- of antibody induction therapy and their associated outcomes. In:
ing. Transplantation 1990;50:683–9. Cecka J, Terasaki P, editors. Clinical transplants 2002. Los Angles:
31. Jonker M, Malissen B, Mawas C. The effect of in vivo application of UCLA Immunogenetics Center; 2003. p. 351.
monoclonal antibodies specific for human cytotoxic T cells in rhesus 56. Batiuk T, Bodziak K, Goldman M. Infectious disease prophylaxis in
monkeys. Transplantation 1983;35:374–8. renal transplant patients: a survey of US transplant centers. Clin
32. Kerr P, Atkins R. The effects of OKT3 therapy on infiltrating lympho- Transplant 2002;16:1–8.
cytes in rejecting renal allografts. Transplantation 1989;48:33–6. 57. Hibberd P, Tolkoff-Rubin N, Cosimi A, et al. Symptomatic cyto-
33. Ferrant J, Benjamin C, Cutler A, et al. The contribution of Fc effec- megalovirus disease in the cytomegalovirus antibody seropositive
tor mechanisms in the efficacy of anti-CD154 immunotherapy renal transplant recipient treated with OKT3. Transplantation
depends on the nature of the immune challenge. Int Immunol 1992;53:68–72.
2004;16:1583–94. 58. Turgeon N, Fishman J, Basgoz N, et al. Effect of oral acyclovir or gan-
34. Casadei D, Rial MDC, Opelz G, et al. A randomized and prospective ciclovir therapy after preemptive intravenous ganciclovir therapy
study comparing treatment with high-dose intravenous immuno- to prevent cytomegalovirus disease in cytomegalovirus seropositive
globulin with monoclonal antibodies for rescue of kidney grafts with renal and liver transplant recipients receiving antilymphocyte anti-
steroid-resistant rejection. Transplantation 2001;71:53–8. body therapy. Transplantation 1998;66:1780–6.
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 305

59. Bonnefoy-Berard N, Vincent C, Revillard J. Antibodies against func- 83. Shield C, Edwards E, Davies D, Daily O. Antilymphocyte induc-
tional leukocyte surface molecules in polyclonal antilymphocyte and tion therapy in cadaver renal transplantation. Transplantation
antithymocyte globulins. Transplantation 1991;51:669–73. 1997;63:1257–63.
60. Bourdage J, Hamlin D. Comparative polyclonal antithymocyte glob- 84. Crowson C, Reed R, Shelton B, et al. Lymphocyte-depleting induc-
ulin and antilymphocyte/antilymphoblast globulin anti-CD antigen tion therapy lowers the risk of acute rejection in African Ameri-
analysis by flow cytometry. Transplantation 1995;59:1194–200. can pediatric kidney transplant recipients. Pediatr Transplant
61. Rebellato L, Gross U, Verbanac K, Thomas J. A comprehensive defini- 2017;21:e12823.
tion of the major antibody specificities in polyclonal rabbit antithy- 85. Pilch N, Taber D, Moussa O, et al. Prospective randomized controlled
mocyte globulin. Transplantation 1994;57:685–94. trial of rabbit antithymocyte globulin compared with IL-2 receptor
62. Simpson M, Monaco A. Clinical uses of polyclonal and monoclonal antagonist induction therapy in kidney transplantation. Ann Surg
antilymphoid sera. In: Chatenoud L, editor. Monoclonal antibodies 2014;259:888–93.
in transplantation. Austin, TX: RG Landes; 1995. p. 1–19. 86. Deierhoi M, Sollinger H, Kalayoglu M, Belzer F. Quadruple therapy for
63. Hoitsma A, Lier HV, Reekers P, Koene R. Improved patient and cadaver renal transplantation. Transplant Proc 1987;19:1917–9.
graft survival after treatment of acute rejections of cadaveric renal 87. Shaffer D, Langone A, Nylander W, et al. A pilot protocol of a cal-
allografts with rabbit antithymocyte globulin. Transplantation cineurin-inhibitor free regimen for kidney transplant recipients of
1985;39:274–9. marginal donor kidneys or with delayed graft function. Clin Trans-
64. Stratta R, D’Alessandro A, Armbrust M, et al. Sequential antilym- plant 2003;17:31–4.
phocyte globulin/cyclosporine immunosuppression in cadaveric 88. Sommer B, Henry M, Ferguson R. Sequential antilymphoblast glob-
renal transplantation. Effect of duration of ALG therapy. Transplan- ulin and cyclosporine for renal transplantation. Transplant Proc
tation 1989;47:96–102. 1987;19:1879–80.
65. Streem S, Novick A, Braun W, et al. Low-dose maintenance predni- 89. Brennan D, Daller J, Lake K, et al. Rabbit antithymocyte globu-
sone and antilymphoblast globulin for the treatment of acute rejec- lin versus basiliximab in renal transplantation. N Engl J Med
tion. A steroid-sparing approach to immunosuppressive therapy. 2006;355:1967–77.
Transplantation 1983;35:420–4. 90. Hellemans R, Hazzan M, Durand D, et al. Daclizumab versus rab-
66. Thomas F, Cunningham P, Thomas J, et al. Superior renal allograft bit antithymocyte globulin in high-risk renal transplants: five-
survival and decreased rejection with early high-dose and sequen- year follow-up of a randomized study. Am J Transplant 2015;15:
tial multi-species antilymphocyte globulin therapy. Transplant Proc 1923–32.
1987;19:1874–8. 91. Thomusch O, Wiesener M, Opgenoorth M, et al. Rabbit-ATG or
67. Mueller T. Mechanism of action of thymoglobulin. Transplantation basiliximab induction for rapid steroid withdrawal after renal trans-
2007;84:S5–10. plantation (Harmony): an open-label, multicentre, randomised con-
68. Beiras-Fernandez A, Chappell D, Hammer C, Thein E. Influence trolled trial. Lancet 2016;388:3006–16.
of polyclonal anti-thymocyte globulins upon ischemia-reperfu- 92. Tanriover B, Jaikaransingh V, MacConmara M, et al. Acute rejection
sion injury in a non-human primate model. Transpl Immunol rates and graft outcomes according to induction regimen among
2006;15:273–9. recipients of kidneys from deceased donors treated with tacrolimus
69. Chappell D, Beiras-Fernande A, Hammer C, Thein E. In vivo visu- and mycophenolate. Clin J Am Soc Nephrol 2016;11:1650–61.
alization of the effect of polyclonal antithymocyte globulins on the 93. Koyawala N, Silber J, Rosenbaum P, et al. Comparing outcomes
microcirculation after ischemia/reperfusion in a primate model. between antibody induction therapies in kidney transplantation.
Transplantation 2006;81:552–8. J Am Soc Nephrol 2017;28:2188–200.
70. Bunn D, Lea C, Bevan D, et al. The pharmacokinetics of anti-thy- 94. Brennan D, Schnitzler M. Long-term results of rabbit antithymocyte
mocyte globulin (ATG) following intravenous infusion in man. Clin globulin and basiliximab induction. N Engl J Med 2008;359:1736–8.
Nephrol 1996;45:29–32. 95. Starzl T, Murase N, Abu-Elmagd K, et al. Tolerogenic immunosup-
71. Regan J, Lyonnais C, Campbell K, et al. Total and active thymoglobu- pression for organ transplantation. Lancet 2003;361:1502–10.
lin levels: effects of dose and sensitization on serum concentrations. 96. Swanson S, Hale D, Mannon R, et al. Kidney transplantation with
Transpl Immunol 2001;9:29–36. rabbit antithymocyte globulin induction and sirolimus monother-
72. Muller T, Grebe S, Neumann M, et al. Persistent long-term changes apy. Lancet 2002;360:1662–4.
in lymphocyte subsets induced by polyclonal antibodies. Transplan- 97. Shield C, Cosimi A, Tolkoff-Rubin N, et al. Use of antithymocyte
tation 1997;64:1432–7. globulin for reversal of acute allograft rejection. Transplantation
73. Preville X, Flacher M, LeMauff B, et al. Mechanisms involved in anti- 1979;28:461–4.
thymocyte globulin immunosuppressive activity in a nonhuman 98. Nelson P, Cosimi A, Delmonico F, et al. Antithymocyte globulin as
primate model. Transplantation 2001;71:460–8. the primary treatment for renal allograft rejection. Transplantation
74. Howard R, Condie R, Sutherland D, et al. The use of antilymphoblast 1983;36:587–9.
globulin in the treatment of renal allograft rejection. Transplant 99. Benvenisty A, Tannenbaum G, Cohen D, et al. Use of antithymocyte
Proc 1981;13:473–4. globulin and cyclosporine to treat steroid-resistant episodes in renal
75. Wechter W, Morrell R, Bergan J, et al. Extended treatment with anti- transplant recipients. Transplant Proc 1987;19:1889–91.
thymocyte globulin (ATGAM) in renal allograft recipients. Trans- 100. Gaber A, First M, Tesi R, et al. Results of the double-blind, random-
plantation 1979;28:365–7. ized, multicenter, phase III clinical trial of Thymoglobulin versus
76. Merion R, White D, Thiru S, et al. Cyclosporine: five years experience Atgam in the treatment of acute graft rejection episodes after renal
in cadaveric renal transplantation. N Engl J Med 1984;310:148–54. transplantation. Transplantation 1998;66:29–37.
77. Oyer P, Stinson E, Jamieson S, et al. Cyclosporin-A in cardiac allograft- 101. Matas A, Tellis V, Quinn T, et al. ALG treatment of steroid-resis-
ing: a preliminary experience. Transplant Proc 1983;15:1247–52. tant rejection in patients receiving cyclosporine. Transplantation
78. Charpentier B, Rostaing L, Berthoux F, et al. A three-arm study com- 1986;41:579–83.
paring immediate tacrolimus therapy with antithymocyte globulin 102. Richardson A, Higgins R, Liddington M, et al. Antithymocyte globu-
induction therapy followed by tacrolimus or cyclosporine A in adult lin for steroid resistant rejection in renal transplant recipients immu-
renal transplant recipients. Transplantation 2003;75:844–51. nosuppressed with triple therapy. Transpl Int 1989;2:27–32.
79. Mourad G, Garrigue V, Squifflet J, et al. Induction versus noninduc- 103. Malinow L, Walker J, Klassen D, et al. Antilymphocyte induction
tion in renal transplant recipients with tacrolimus-based immuno- immunosuppression in the post-Minnesota antilymphocyte globu-
suppression. Transplantation 2001;72:1050–5. lin era: incidence of renal dysfunction and delayed graft function: a
80. Cecka J, Gjertson D, Terasaki P. Do prophylactic antilymphocyte single center experience. Clin Transplant 1996;10:237–42.
globulins (ALG and OKT3) improve renal transplant in recipient and 104. Bock H, Gallati H, Zurcher R, et al. A randomized prospective trial of
donor high-risk groups? Transplant Proc 1993;25:548–9. prophylactic immunosuppression with ATG-Fresenius versus OKT3
81. Cecka J, Terasaki P. The UNOS scientific renal transplant registry after renal transplantation. Transplantation 1995;59:830–40.
1991. In: Terasaki P, editor. Clinical transplants 1991. Los Angeles: 105. Kazatchkine M, Kaveri S. Immunomodulation of autoimmune and
UCLA Tissue Typing Laboratory; 1991. p. 1. inflammatory diseases with intravenous immune globulin. N Engl J
82. Goggins W, Pascual M, Powelson J, et al. A prospective, randomized, Med 2001;345:747–55.
clinical trial of intraoperative versus postoperative Thymoglobu- 106. Rahman G, Hardy M, Cohen D. Administration of equine anti-thy-
lin in adult cadaveric renal transplant recipients. Transplantation mocyte globulin via peripheral vein in renal transplant recipients.
2003;76:798–802. Transplantation 2000;69:1958–60.
306 Kidney Transplantation: Principles and Practice

107. Wiland A, Fink J, Philosophe B, et al. Peripheral administration of 131. Norman D, Shield Cr, Barry J, et al. Early use of OKT3 monoclonal
thymoglobulin for induction therapy in pancreas transplantation. antibody in renal transplantation to prevent rejection. Am J Kidney
Transplant Proc 2001;33:1910. Dis 1988;11:107–10.
108. Stevens R, Wrenshall L, Miles C, et al. A double-blind, double- 132. Opelz G. Efficacy of rejection prophylaxis with OKT3 in renal
dummy, flexible-design randomized multicenter trial: early safety of transplantation. collaborative transplant study. Transplantation
single- versus divided-dose rabbit anti-thymocyte globulin induction 1995;60:1220–4.
in renal transplantation. Am J Transplant 2016;16:1858–67. 133. Benvenisty A, Cohen D, Stegall M, Hardy M. Improved results using
109. Brennan D, Flavin K, Lowell J, et al. A randomized, double-blinded OKT3 as induction immunosuppression in renal allograft recipients
comparison of thymoglobulin versus ATGAM for induction immu- with delayed graft function. Transplantation 1990;49:321–7.
nosuppressive therapy in adult renal transplant recipients. Trans- 134. Kahana L, Ackermann J, Lefor W, et al. Uses of orthoclone OKT3 for
plantation 1999;67:1011–8. prophylaxis of rejection and induction in initial nonfunction in kid-
110. Hardinger K, Schnitzler M, Miller B, et al. Five-year follow up of thy- ney transplantation. Transplant Proc 1990;22:1755–8.
moglobulin versus ATGAM induction in adult renal transplantation. 135. Abramowicz D, Pradier O, Marchant A, et al. Induction of throm-
Transplantation 2004;78:136–41. boses within renal grafts by high-dose prophylactic OKT3. Lancet
111. Abbott K, Hypolite I, Viola R, et al. Hospitalizations for cytomega- 1992;339:777–8.
lovirus disease after renal transplantation in the united states. Ann 136. Henry M, Pelletier R, Elkhammas E, et al. A randomized prospective
Epidemiol 2002;12:402–9. trial of OKT3 induction in the current immunosuppression era. Clin
112. Gourishankar S, McDermid J, Jhangri G, Preiksaitis J. Herpes zos- Transplant 2001;15:410–4.
ter infection following solid organ transplantation: incidence, risk 137. Norman D, Kahana L, Stuart FJ, et al. A randomized clinical trial of
factors and outcomes in the current immunosuppressive era. Am J induction therapy with OKT3 in kidney transplantation. Transplan-
Transplant 2004;4:105–15. tation 1993;55:44–50.
113. Bielory L, Wright R, Nienhuis A, et al. Antithymocyte globu- 138. Benfield M, Tejani A, Harmon W, et al. A randomized multicenter
lin hypersensitivity in bone marrow failure patients. JAMA trial of OKT3 mAbs induction compared with intravenous cyclo-
1988;260:3164–7. sporine in pediatric renal transplantation. Pediatr Transplant
114. Brooks C, Karl K, Francom S. ATGAM skin test standardization: com- 2005;9:282–92.
parison of skin testing techniques in horse-sensitive and unselected 139. Delmonico F, Cosimi A. Monoclonal antibody treatment of human
human volunteers. Transplantation 1994;58:1135–7. allograft recipients. Surg Gynecol Obstet 1988;166:89–98.
115. Chatenoud L, Ferran C, Legendre C, et al. In vivo cell activation fol- 140. Tesi R, Elkhammas E, Henry M, Ferguson R. OKT3 for primary ther-
lowing OKT3 administration: systemic cytokine release and modula- apy of the first rejection episode in kidney transplants. Transplanta-
tion by corticosteroids. Transplantation 1990;49:697–702. tion 1993;55:1023–9.
116. Debets J, Leunissen K, Hooff HV, et al. Evidence of involvement of 141. Thistlethwaite JJ, Gaber A, Haag B, et al. OKT3 treatment of steroid-
tumor necrosis factor in adverse reactions during treatment of kid- resistant renal allograft rejection. Transplantation 1987;43:176–84.
ney allograft rejection with antithymocyte globulin. Transplanta- 142. Thistlethwaite JJ, Cosimi A, Delmonico F, et al. Evolving use of OKT3
tion 1989;47:487–92. monoclonal antibody for treatment of renal allograft rejection.
117. Vallhonrat H, Williams W, Cosimi A, et al. In vivo generation of C4b, Transplantation 1984;38:695–701.
Bb, iC3b, and SC5b-9 after OKT3 administration in kidney and lung 143. D’Alessandro A, Pirsch J, Stratta R, et al. OKT3 salvage therapy in
transplant recipients. Transplantation 1999;67:253–8. a quadruple immunosuppressive protocol in cadaveric renal trans-
118. Colovai A, Vasilescu E, Foca-Rodi A, et al. Acute and hyperacute plantation. Transplantation 1989;47:297–300.
humoral rejection in kidney allograft recipients treated with anti- 144. Amlot P, Rawlings E, Fernando O, et al. Prolonged action of a chi-
human thymocyte antibodies. Hum Immunol 2005;66:501–12. meric interleukin-2 receptor (CD25) monoclonal antibody used in
119. Andersen D, Reilly D. Production technologies for monoclonal anti- cadaveric renal transplantation. Transplantation 1995;60:748–56.
bodies and their fragments. Curr Opin Biotechnol 2004;15:456–62. 145. Kovarik J, Burtin P. Immunosuppressants in advanced clinical
120. Saeed A, Wang R, Ling S, Wang S. Antibody engineering for pursu- development for organ transplantation and selected autoimmune
ing a healthier future. Front Microbiol 2017;8:495. diseases. Expert Opin Emerg Drugs 2003;8:47–62.
121. Chatenoud L, Jonker M, Villemain F, et al. The human immune 146. Sarwal M, Ettenger R, Dharnidarka V, et al. Complete steroid avoid-
response to the OKT3 monoclonal antibody is oligoclonal. Science ance is effective and safe in children with renal transplants: a multi-
1986;232:1406–8. center randomized trial with three-year follow-up. Am J Transplant
122. Schroeder T, First M, Mansour M, et al. Antimurine antibody forma- 2012;12:2719–29.
tion following OKT3 therapy. Transplantation 1990;49:48–51. 147. Vincenti F, Lantz M, Birnbaum J, et al. A phase I trial of human-
123. Heinrich G, Gram H, Kocher H, et al. Characterization of a human ized anti-interleukin-2 receptor antibody in renal transplantation.
T cell-specific chimeric antibody (CD7) with human constant and Transplantation 1997;63:33–8.
mouse variable regions. J Immunol 1989;143:3589–97. 148. Kirkman R, Shapiro M, Carpenter C, et al. A randomized prospective
124. Delmonico F, Cosimi A, Kawai T, et al. Non-human primate trial of anti-Tac monoclonal antibody in human renal transplanta-
responses to murine and humanized OKT4A. Transplantation tion. Transplantation 1991;51:107–13.
1993;55:722–8. 149. Suri-Payer E, Amar A, Thornton A, Shevach E. CD4 CD25 T cells
125. Hoffmann S, Hale D, Kleiner D, et al. Functionally significant renal inhibit both the induction and effector function of autoreactive
allograft rejection is defined by transcriptional criteria. Am J Trans- T cells and represent a unique lineage of immunoregulatory cells.
plant 2005;5:573–81. J Immunol 1998;160:1212–8.
126. Kirk A, Hale D, Mannon R, et al. Results from a human renal 150. Gabardi S, Catella J, Martin S, et al. Maintenance immunosuppres-
allograft tolerance trial evaluating the humanized CD52-specific sion with intermittent intravenous IL-2 receptor antibody therapy
monoclonal antibody alemtuzumab (CAMPATH-1H). Transplanta- in renal transplant recipients. Ann Pharmacother 2011;45:e48.
tion 2003;76:120–9. 151. Soulillou J, Cantarovich D, Mauff BL, et al. Randomized controlled
127. Vigeral P, Chkoff N, Chatenoud L, et al. Prophylactic use of OKT3 trial of a monoclonal antibody against the interleukin-2 recep-
monoclonal antibody in cadaver kidney recipients: utilization tor (33B3.1) as compared with rabbit antithymocyte globulin
of OKT3 as the sole immunosuppressive agent. Transplantation for prophylaxis against rejection of renal allografts. N Engl J Med
1986;41:730–3. 1990;322:1175–82.
128. Abramowicz D, Goldman M, Pauw LD, et al. The long-term effects of 152. Hiesse C, Lantz O, Kriaa F, et al. Treatment with Lo-Tact-1, a mono-
prophylactic OKT3 monoclonal antibody in cadaver kidney trans- clonal antibody to the interleukin-2 receptor, in kidney transplanta-
plantation: a single-center, prospective, randomized study. Trans- tion. Presse Med 1991;40.
plantation 1992;54:433–7. 153. Gelder Tv, Zietse R, Mulder A, et al. A double-blind, placebo-con-
129. Debure A, Chkoff N, Chatenoud L, et al. One-month prophylactic use trolled study of monoclonal anti-interleukin-2 receptor antibody
of OKT3 in cadaver kidney transplant recipients. Transplantation (BT563) administration to prevent acute rejection after kidney
1988;45:546–53. transplantation. Transplantation 1995;60:248–52.
130. Millis J, McDiarmid S, Hiatt J, et al. Randomized prospective trial of 154. Beniaminovitz A, Itescu S, Leitz K, et al. Prevention of rejection in
OKT3 for early prophylaxis of rejection after liver transplantation. cardiac transplantation by blockade of the interleukin-2 receptor
Transplantation 1989;47:82–8. with a monoclonal antibody. N Engl J Med 2000;342:613–9.
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 307

155. Hershberger R, Starling R, Eisen H, et al. Daclizumab to prevent rejec- 179. Shapiro R, Basu A, Tan H, et al. Kidney transplantation under mini-
tion after cardiac transplantation. N Engl J Med 2005;352:2708–13. mal immunosuppression after pretransplant lymphoid depletion
156. Kahan B, Rajagopalan P, Hall M. Reduction of the occurrence of with thymoglobulin or campath. J Am Coll Surg 2005;200:505–15.
acute cellular rejection among renal allograft recipients treated with 180. Shapiro R, Young J, Milford E, et al. Immunosuppression: evolution
basiliximab, a chimeric anti-interleukin-2-receptor monoclonal in practice and trends, 1993–2003. Am J Transplant 2005;5:874–
antibody. Transplantation 1999;67:276–84. 86.
157. Nashan B, Light S, Hardie I, et al. Reduction of acute renal allograft 181. Bartosh S, Knechtle S, Sollinger H. Campath-1H use in pediatric
rejection by daclizumab. Transplantation 1999;67:110–5. renal transplantation. Am J Transplant 2005;5:1569–73.
158. Nashan B, Moore R, Amlot P, et al. Randomised trial of basilix- 182. Calne R, Friend P, Moffatt S, et al. Prope tolerance, periopera-
imab versus placebo for control of acute cellular rejection in renal tive campath 1H, and low-dose cyclosporin monotherapy in renal
allograft recipients. CHIB. 201 international study group. Lancet allograft recipients. Lancet 1998;351:1701–2.
1997;350:1193–8. 183. Calne R, Moffatt S, Friend P, et al. Campath IH allows low-dose
159. Neuhaus P, Clavien P, Kittur D, et al. Improved treatment response cyclosporine monotherapy in 31 cadaveric renal allograft recipients.
with basiliximab immunoprophylaxis after liver transplantation: Transplantation 1999;68:1613–6.
results from a double-blind randomized placebo-controlled trial. 184. Gruessner R, Kandaswamy R, Humar A, et al. Calcineurin inhibitor-
Liver Transpl 2002;8:132–42. and steroid-free immunosuppression in pancreas-kidney and soli-
160. Shapiro A, Lakey J, Ryan E, et al. Islet transplantation in seven tary pancreas transplantation. Transplantation 2005;79:1184–9.
patients with type 1 diabetes mellitus using a glucocorticoid-free 185. Kaufman D, Leventhal J, Axelrod D, et al. Alemtuzumab induc-
immunosuppressive regimen. N Engl J Med 2000;343:230–8. tion and prednisone-free maintenance immunotherapy in kidney
161. Vincenti F, Kirkman R, Light S, et al. Interleukin-2-receptor block- transplantation: comparison with basiliximab induction: long-term
ade with daclizumab to prevent acute rejection in renal trans- results. Am J Transplant 2005;5:2539–48.
plantation. daclizumab triple therapy study group. N Engl J Med 186. Kaufman D, Leventhal J, Gallon L, Parker M. Alemtuzumab induc-
1998;338:161–5. tion and prednisone-free maintenance immunotherapy in simul-
162. Lebranchu Y, Bridoux F, Buchler M, et al. Immunoprophylaxis with taneous pancreas-kidney transplantation comparison with rabbit
basiliximab compared with antithymocyte globulin in renal trans- antithymocyte globulin induction long-term results. Am J Trans-
plant patients receiving MMF-containing triple therapy. Am J Trans- plant 2006;6:331–9.
plant 2002;2:48–56. 187. Kirk A, Mannon R, Kleiner D, et al. Results from a human renal
163. Mourad G, Rostaing L, Legendre C, et al. Sequential protocols using allograft tolerance trial evaluating T-cell depletion with alem-
basiliximab versus antithymocyte globulins in renal-transplant tuzumab combined with deoxyspergualin. Transplantation
patients receiving mycophenolate mofetil and steroids. Transplanta- 2005;80:1051–9.
tion 2004;78:584–90. 188. Knechtle S, Fernandez L, Pirsch J, et al. Campath-1H in renal
164. Sollinger H, Kaplan B, Pescovitz M, et al. Basiliximab versus anti- transplantation: the University of Wisconsin experience. Surgery
thymocyte globulin for prevention of acute renal allograft rejection. 2004;136:754–60.
Transplantation 2001;72:1915–9. 189. Knechtle S, Pirsch J, Fechner JJ, et al. Campath-1H induction plus
165. Hanaway M, Woodle E, Mulgaonkar S, et al. Alemtuzumab induc- rapamycin monotherapy for renal transplantation: results of a pilot
tion in renal transplantation. N Engl J Med 2011;364:1909–19. study. Am J Transplant 2003;3:722–30.
166. Boillot O, Mayer D, Boudjema K, et al. Corticosteroid-free immuno- 190. McCurry K, Iacono A, Zeevi A, et al. Early outcomes in human lung
suppression with tacrolimus following induction with daclizumab: a transplantation with Thymoglobulin or Campath-1H for recipient
large randomized clinical study. Liver Transpl 2005;11:61–7. pretreatment followed by posttransplant tacrolimus near-monother-
167. Rostaing L, Cantarovich D, Mourad G, et al. Corticosteroid-free apy. J Thorac Cardiovasc Surg 2005;130:528–37.
immunosuppression with tacrolimus, mycophenolate mofetil, and 191. Tan H, Kaczorowski D, Basu A, et al. Living donor renal transplan-
daclizumab induction in renal transplantation. Transplantation tation using alemtuzumab induction and tacrolimus monotherapy.
2005;79:807–14. Am J Transplant 2006;6:2409–17.
168. Squifflet J, Besse T, Malaise J, et al. BTI-322 for induction therapy 192. Tzakis A, Tryphonopoulos P, Kato T, et al. Preliminary experience
after renal transplantation: a randomized study. Transplant Proc with alemtuzumab (Campath-1H) and low-dose tacrolimus immu-
1997;29:317–9. nosuppression in adult liver transplantation. Transplantation
169. Grenda R, Watson A, Trompeter R, et al. A randomized trial to assess 2004;77:1209–14.
the impact of early steroid withdrawal on growth in pediatric renal 193. Group CSC, Haynes R, Harden P, et al. Alemtuzumab-based induc-
transplantation: the TWIST study. Am J Transplant 2010;10:828– tion treatment versus basiliximab-based induction treatment in
36. kidney transplantation (the 3C Study): a randomised trial. Lancet
170. Webb N, Douglas S, Rajai A, et al. Corticosteroid-free kidney trans- 2014;384:1684–90.
plantation improves growth: 2-year follow-up of the TWIST ran- 194. Cai J, Terasaki P, Bloom D, et al. Correlation between human leuko-
domized controlled trial. Transplantation 2015;99:1178–85. cyte antigen antibody production and serum creatinine in patients
171. Parrott N, Hammad A, Watson C, et al. Multicenter, randomized receiving sirolimus monotherapy after Campath-1H induction.
study of the effectiveness of basiliximab in avoiding addition of ste- Transplantation 2004;78:919–24.
roids to cyclosporine a monotherapy in renal transplant recipients. 195. Friend P, Hale G, Waldmann H, et al. Campath-1M—prophylactic
Transplantation 2005;79:344–8. use after kidney transplantation: a randomized controlled clinical
172. Vincenti F, Ramos E, Brattstrom C, et al. Multicenter trial exploring trial. Transplantation 1989;48:248–53.
calcineurin inhibitors avoidance in renal transplantation. Trans- 196. Friend P, Rebello P, Oliveira D, et al. Successful treatment of renal
plantation 2001;71:1282–7. allograft rejection with a humanized antilymphocyte monoclonal
173. Asberg A, Midtvedt K, Line P, et al. Calcineurin inhibitor avoidance antibody. Transplant Proc 1995;27:869–70.
with daclizumab, mycophenolate mofetil, and prednisolone in DR- 197. Friend P, Waldmann H, Hale G, et al. Reversal of allograft rejec-
matched de novo kidney transplant recipients. Transplantation tion using the monoclonal antibody, Campath-1G. Transplant Proc
2006;82:62–8. 1991;23:2253–4.
174. Bloom D, Chang Z, Pauly K, et al. BAFF is increased in renal trans- 198. Hale G, Waldmann H, Friend P, Calne R. Pilot study of CAMPATH-1,
plant patients following treatment with alemtuzumab. Am J Trans- a rat monoclonal antibody that fixes human complement, as an
plant 2009;9:1835–45. immunosuppressant in organ transplantation. Transplantation
175. Magliocca J, Knechtle S. The evolving role of alemtuzumab (Cam- 1986;42:308–11.
path-1H) for immunosuppressive therapy in organ transplantation. 199. Clatworthy M, Friend P, Calne R, et al. Alemtuzumab (CAMPATH-
Transpl Int 2006;19:705–14. 1H) for the treatment of acute rejection in kidney transplant recipi-
176. Waldmann H, Hale G. CAMPATH: from concept to clinic. Philos ents: long-term follow-up. Transplantation 2009;87:1092–5.
Trans R Soc Lond B Biol Sci 2005;360:1707–11. 200. Basu A, Ramkumar M, Tan H, et al. Reversal of acute cellular rejec-
177. Weaver T, Kirk A. Alemtuzumab. Transplantation 2007;84:1545–7. tion after renal transplantation with Campath-1H. Transplant Proc
178. Hale G. The CD52 antigen and development of the CAMPATH anti- 2005;37:923–6.
bodies. Cytotherapy 2001;3:137–43.
308 Kidney Transplantation: Principles and Practice

201. Csapo Z, Benavides-Viveros C, Podder H, et al. Campath-1H as res- 225. Chancharoenthana W, Townamchai N, Leelahavanichkul A, et al.
cue therapy for the treatment of acute rejection in kidney transplant Rituximab for recurrent IgA nephropathy in kidney transplantation:
patients. Transplant Proc 2005;37:2032–6. A report of three cases and proposed mechanisms. Nephrology (Carl-
202. Upadhyay K, Midgley L, Moudgil A. Safety and efficacy of alemtu- ton) 2017;22:65–71.
zumab in the treatment of late acute renal allograft rejection. Pediatr 226. Clatworthy M, Watson C, Plotnek G, et al. B-cell-depleting induction
Transplant 2012;16:286–93. therapy and acute cellular rejection. N Engl J Med 2009;360:2683–5.
203. Mvd Hoogen, Hesselink D, Son Wv, et al. Treatment of steroid-resis- 227. Deans J, Li H, Polyak M. CD20-mediated apoptosis: signalling
tant acute renal allograft rejection with alemtuzumab. Am J Trans- through lipid rafts. Immunology 2002;107:176–82.
plant 2013;13:192–6. 228. Taylor R, Lindorfer M. Immunotherapeutic mechanisms of anti-
204. Coles A, Wing M, Smith S, et al. Pulsed monoclonal antibody treat- CD20 monoclonal antibodies. Curr Opin Immunol 2008;20:444–9.
ment and autoimmune thyroid disease in multiple sclerosis. Lancet 229. Hippen B, DeMattos A, Cook W, et al. Association of CD20 infiltrates
1999;354:1691–5. with poorer clinical outcomes in acute cellular rejection of renal
205. Coles A, Cohen J, Fox E, et al. Alemtuzumab CARE-MS II 5-year fol- allografts. Am J Transplant 2005;5:2248–52.
low-up: efficacy and safety findings. Neurology 2017;89:1117–26. 230. Sarwal M, Chua M, Kambham N, et al. Molecular heterogeneity in
206. Kirk A, Hale D, Swanson S, Mannon R. Autoimmune thyroid disease acute renal allograft rejection identified by DNA microarray profil-
after renal transplantation using depletional induction with alemtu- ing. N Engl J Med 2003;349:125–38.
zumab. Am J Transplant 2006;6:1084–5. 231. Fornoni A, Sageshima J, Wei C, et al. Rituximab targets podocytes
207. Sood P, Hariharan S. Anti-CD20 blocker rituximab in kidney trans- in recurrent focal segmental glomerulosclerosis. Sci Transl Med
plantation transplantation. 2018;102(1):44–58. Available online 2011;3. 85ra46.
at: https://doi.org/10.1097/TP.0000000000001849. 232. Faguer S, Kamar N, Guilbeaud-Frugier C, et al. Rituximab therapy
208. Deans J, Kalt L, Ledbetter J, et al. Association of 75/80-kDa phospho- for acute humoral rejection after kidney transplantation. Transplan-
proteins and the tyrosine kinases Lyn, Fyn, and Lck with the B cell tation 2007;83:1277–80.
molecule CD20. Evidence against involvement of the cytoplasmic 233. Svoboda J, Kotloff R, Tsai D. Management of patients with post-trans-
regions of CD20. J Biol Chem 1995;270:22632–8. plant lymphoproliferative disorder: the role of rituximab. Transpl Int
209. Grillo-Lopez A, White C, Varns C, et al. Overview of the clinical devel- 2006;19:259–69.
opment of rituximab: first monoclonal antibody approved for the 234. Trappe R, Dierickx D, Zimmermann H, et al. Response to rituximab
treatment of lymphoma. Semin Oncol 1999;26:66–73. induction is a predictive marker in B-cell post-transplant lymphopro-
210. Becker Y, Becker B, Pirsch J, Sollinger H. Rituximab as treat- liferative disorder and allows successful stratification into rituximab
ment for refractory kidney transplant rejection. Am J Transplant or R-CHOP consolidation in an international, prospective, multi-
2004;4:996–1001. center phase II trial. J Clin Oncol 2017;35:536–43.
211. Becker Y, Samaniego-Picota M, Sollinger H. The emerging role of 235. Nixon A, Ogden L, Woywodt A, Dhaygude A. Infectious com-
rituximab in organ transplantation. Transpl Int 2006;19:621–8. plications of rituximab therapy in renal disease. Clin Kidney J
212. Fuchinoue S, Ishii Y, Sawada T, et al. The 5-year outcome of ABO- 2017;10:455–60.
incompatible kidney transplantation with rituximab induction. 236. Bayes M, Rabasseda X, Prous J. Gateways to clinical trials. Methods
Transplantation 2011;91:853–7. Find Exp Clin Pharmacol 2005;27:49–77.
213. Genberg H, Kumlien G, Wennberg L, et al. ABO-incompatible kid- 237. Vincenti F, Rostaing L, Grinyo J, et al. Belatacept and long-term out-
ney transplantation using antigen-specific immunoadsorption and comes in kidney transplantation. N Engl J Med 2016;374:333–43.
rituximab: a 3-year follow-up. Transplantation 2008;85:1745–54. 238. Florman S, Becker T, Bresnahan B, et al. Efficacy and safety out-
214. Rehman S, Meier-Kriesche H, Scornik J. Use of intravenous immune comes of extended criteria donor kidneys by subtype: subgroup anal-
globulin and rituximab for desensitization of highly human leuko- ysis of BENEFIT-EXT at 7 years after transplant. Am J Transplant
cyte antigen-sensitized patients awaiting kidney transplantation. 2017;17:180–90.
Transplantation 2010;90:932. 239. Kim E, Kwun J, Gibby A, et al. Costimulation blockade alters germi-
215. Sonnenday C, Warren D, Cooper M, et al. Plasmapheresis, CMV nal center responses and prevents antibody-mediated rejection. Am
hyperimmune globulin, and anti-CD20 allow ABO-incompatible J Transplant 2014;14:59–69.
renal transplantation without splenectomy. Am J Transplant 240. Rostaing L, Vincenti F, Grinyo J, et al. Long-term belatacept exposure
2004;4:1315–22. maintains efficacy and safety at 5 years: results from the long-term
216. Tyden G, Kumlien G, Genberg H, et al. ABO incompatible kidney extension of the BENEFIT study. Am J Transplant 2013;13:2875–83.
transplantations without splenectomy, using antigen-specific 241. Vincenti F, Larsen C, Alberu J, et al. Three-year outcomes from BEN-
immunoadsorption and rituximab. Am J Transplant 2005;5:145–8. EFIT, a randomized, active-controlled, parallel-group study in adult
217. Vo A, Peng A, Toyoda M, et al. Use of intravenous immune globulin and kidney transplant recipient. Am J Transplant 2012;12:210–7.
rituximab for desensitization of highly HLA-sensitized patients await- 242. Archdeacon P, Dixon C, Belen O, et al. Summary of the US FDA
ing kidney transplantation. Transplantation 2010;89:1095–102. approval of belatacept. Am J Transplant 2012;12:554–62.
218. Kahwaji J, Jordan S, Najjar R, et al. Six-year outcomes in broadly 243. Espinosa J, Herr F, Tharp G, et al. CD57+ CD4 T cells underlie belata-
HLA-sensitized living donor transplant recipients desensitized cept-resistant allograft rejection. Am J Transplant 2016;16:1102–12.
with intravenous immunoglobulin and rituximab. Transpl Int 244. Ferguson R, Grinyo J, Vincenti F, et al. Immunosuppression with
2016;29:1276–85. belatacept-based, corticosteroid-avoiding regimens in de novo kid-
219. El-Zoghby Z, Grande J, Fraile M, et al. Recurrent idiopathic mem- ney transplant recipients. Am J Transplant 2011;11:66–76.
branous nephropathy: early diagnosis by protocol biopsies and 245. Kirk A, Guasch A, Xu H, et al. Renal transplantation using belata-
treatment with anti-CD20 monoclonal antibodies. Am J Transplant cept without maintenance steroids or calcineurin inhibitors. Am J
2009;9:2800–7. Transplant 2014;14:1142–51.
220. Pescovitz M, Book B, Sidner R. Resolution of recurrent focal segmen- 246. Lerch C, Kanzelmeyer N, Ahlenstiel-Grunow T, et al. Belatacept after
tal glomerulosclerosis proteinuria after rituximab treatment. N Engl kidney transplantation in adolescents: a retrospective study. Transpl
J Med 2006;354:1961–3. Int 2017;30:494–501.
221. Apeland T, Hartmann A. Rituximab therapy in early recurrent focal 247. Vincenti F. New monoclonal antibodies in renal transplantation.
segmental sclerosis after renal transplantation. Nephrol Dial Trans- Minerva Urol Nefrol 2003;55:57–66.
plant 2008;23:2091–4. 248. Lerut J, Thuyne VV, Mathjis J, et al. Anti-CD2 monoclonal anti-
222. Audard V, Kamar N, Sahali D, et al. Rituximab therapy prevents body and tacrolimus in adult liver transplantation. Transplantation
focal and segmental glomerulosclerosis recurrence after a second 2005;80:1186–93.
renal transplantation. Transpl Int 2012;25:e62–6. 249. Mourad M, Besse T, Malaise J, et al. BTI-322 for acute rejection after
223. Murakami C, Manoharan P, Carter-Monroe N, Geetha D. Rituximab renal transplantation. Transplant Proc 1997;29:2353.
for remission induction in recurrent ANCA-associated glomerulone- 250. Przepiorka D, Phillips G, Ratanatharathorn V, et al. A phase II study
phritis postkidney transplant. Transpl Int 2013;26:1225–31. of BTI-322, a monoclonal anti-CD2 antibody for treatment of ste-
224. Geetha D, Seo P, Specks U, Fervenza F. Successful induction of roid-resistant acute GVHD. Blood 1998;92:4066–71.
remission with rituximab for relapse of ANCA-associated vascu- 251. Kawai T, Wee S, Bazin H, et al. Association of natural killer cell
litis post-kidney transplant: report of two cases. Am J Transplant depletion with induction of mixed chimerism and allograft tolerance
2007;7:2821–5. in non-human primates. Transplantation 2000;70:368–74.
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 309

252. Spitzer T, McAfee S, Dey B, et al. Nonmyeloablative haploidenti- 275. Delmonico F, Cosimi A. Anti-CD4 monoclonal antibody therapy.
cal stem-cell transplantation using anti-CD2 monoclonal antibody Clin Transplant 1996;10:397–403.
(MEDI-507)-based conditioning for refractory hematologic malig- 276. Powelson J, Knowles R, Delmonico F, et al. CDR-grafted OKT4A
nancies. Transplantation 2003;75:1748–51. monoclonal antibody in cynomolgus renal allograft recipients.
253. Gordon K, Vaishnaw A, O’Gorman J, et al. Treatment of psoriasis Transplantation 1994;57:788–93.
with alefacept: correlation of clinical improvement with reductions 277. Sablinski T, Sayegh M, Kut J, et al. The importance of targeting
of memory T-cell counts. Arch Dermatol 2003;139:1563–70. the CD4+ T cell subset at the time of antigenic challenge for induc-
254. Dhanireddy K, Bruno D, Zhang X, et al. Alefacept (LFA3-Ig), tion of prolonged vascularized allograft survival. Transplantation
portal venous donor specific transfusion, and sirolimus prolong 1992;53:219–21.
renal allograft survival in non-human primates. J Am Coll Surg 278. Arima T, Lehmann M, Flye M. Induction of donor specific trans-
2006;203:S92. plantation tolerance to cardiac allografts following treatment with
255. Weaver T, Charafeddine A, Agarwal A, et al. Alefacept promotes nondepleting (RIB 5/2) or depleting (OX-38) anti-CD4 mAb plus
co-stimulation blockade based allograft survival in nonhuman pri- intrathymic or intravenous donor alloantigen. Transplantation
mates. Nat Med 2009;15:746–9. 1997;63:284–92.
256. Kwun J, Page E, Hong J, et al. Neutralizing BAFF/APRIL with ataci- 279. Cosimi A, Delmonico F, Wright J, et al. Prolonged survival of non-
cept prevents early DSA formation and AMR development in T cell human primate renal allograft recipients treated only with anti-
depletion induced nonhuman primate AMR model. Am J Transplant CD4 monoclonal antibody. Surgery 1990;108:406–13; discussion
2015;15:815–22. 413–4.
257. Lo D, Weaver T, Stempora L, et al. Selective targeting of human allo- 280. Darby C, Bushell A, Morris P, Wood K. Nondepleting anti-CD4
responsive CD8+ effector memory T cells based on CD2 expression. antibodies in transplantation: evidence that modulation is far
Am J Transplant 2011;11:22–33. less effective than prolonged CD4 blockade. Transplantation
258. Lee S, Yamada Y, Tonsho M, et al. Alefacept promotes immu- 1994;57:1419–26.
nosuppression-free renal allograft survival in nonhuman pri- 281. Lehmann M, Sternkopf F, Metz F, et al. Induction of long-term
mates via depletion of recipient memory T cells. Am J Transplant survival of rat skin allografts by a novel, highly effective anti-CD4
2013;13:3223–9. monoclonal antibody. Transplantation 1992;54:959–62.
259. Rostaing L, Charpentier B, Glyda M, et al. Alefacept combined with 282. Niimi M, Witzke O, Bushell A, et al. Nondepleting anti-CD4 mono-
tacrolimus, mycophenolate mofetil and steroids in de novo kidney clonal antibody enhances the ability of oral alloantigen delivery to
transplantation: a randomized controlled trial. Am J Transplant induce indefinite survival of cardiac allografts: oral tolerance to allo-
2013;13:1724–33. antigen. Transplantation 2000;70:1524–8.
260. Friend P, Hale G, Chatenoud L, et al. Phase I study of an engineered 283. Wee S, Stroka D, Preffer F, et al. The effects of OKT4A monoclonal
aglycosylated humanized CD3 antibody in renal transplant rejec- antibody on cellular immunity of nonhuman primate renal allograft
tion. Transplantation 1999;68:1632–7. recipients. Transplantation 1992;53:501–7.
261. Norman D, Vincenti F, Mattos Ad, et al. Phase I trial of HuM291, a 284. Dantal J, Ninin E, Hourmant M, et al. Anti-CD4 MoAb therapy in kid-
humanized anti-CD3 antibody, in patients receiving renal allografts ney transplantation: a pilot study in early prophylaxis of rejection.
from living donors. Transplantation 2000;70:1707–12. Transplantation 1996;62:1502–6.
262. Woodle E, Xu D, Zivin R, et al. Phase I trial of a humanized, Fc 285. Land W. Monoclonal antibodies in 1991: new potential options
receptor nonbinding OKT3 antibody, huOKT3gamma1(Ala-Ala) in clinical immunosuppressive therapy. Clin Transplant 1991;5:
in the treatment of acute renal allograft rejection. Transplantation 493–500.
1999;68:608–16. 286. Group CCTiTR. Murine OKT4A immunosuppression in cadaver
263. Xu D, Alegre M, Varga S, et al. In vitro characterization of five donor renal allograft recipients: a cooperative clinical trials in trans-
humanized OKT3 effector function variant antibodies. Cell Immunol plantation pilot study. Transplantation 1997;63:1087–95.
2000;200:16–26. 287. Meiser B, Reiter C, Reichenspurner H, et al. Chimeric monoclonal
264. Carpenter P, Lowder J, Johnston L, et al. A phase II multicenter study CD4 antibody: a novel immunosuppressant for clinical heart trans-
of visilizumab, humanized anti-CD3 antibody, to treat steroid-refrac- plantation. Transplantation 1994;58:419–23.
tory acute graft-versus-host disease. Biol Blood Marrow Transplant 288. Robbins R, Oyer P, Stinson E, et al. The use of monoclonal antibodies
2005;11:465–71. after heart transplantation. Transplant Sci 1992;2:22.
265. Herold K, Hagopian W, Auger J, et al. Anti-CD3 monoclonal 289. Reinke P, Kern F, Fietze E, et al. Anti-CD4 monoclonal antibody
antibody in new-onset type 1 diabetes mellitus. N Engl J Med therapy of late acute rejection in renal allograft recipients: CD4 T
2002;346:1692–8. cells play an essential role in the rejection process. Transplant Proc
266. Sherry N, Hagopian W, Ludvigsson J, et al. Teplizumab for treatment 1995;27:859–62.
of type 1 diabetes (Protégé study):1-year results from a randomised, 290. Choy E, Panayi G, Emery P, et al. Repeat-cycle study of high-dose
placebo-controlled trial. Lancet 2011;378:487–97. intravenous 4162W94 anti-CD4 humanized monoclonal antibody
267. Halota W, Ferenci P, Kozielewicz D, et al. Oral anti-CD3 immuno- in rheumatoid arthritis. A randomized placebo-controlled trial.
therapy for HCV-nonresponders is safe, promotes regulatory T cells Rheumatology (Oxford) 2002;41:1142–8.
and decreases viral load and liver enzyme levels: results of a phase-2a 291. Kuritzkes D, Jacobson J, Powderly W, et al. Antiretroviral activity
placebo-controlled trial. J Viral Hepat 2015;22:651–7. of the anti-CD4 monoclonal antibody TNX-355 in patients infected
268. Lalazar G, Mizrahi M, Turgeman I, et al. Oral administration of OKT3 with HIV type 1. J Infect Dis 2004;189:286–91.
MAb to patients with NASH, promotes regulatory T-cell induction, 292. Skov L, Kragballe K, Zachariae C, et al. HuMax-CD4: a fully human
and alleviates insulin resistance:results of a phase IIa blinded pla- monoclonal anti-CD4 antibody for the treatment of psoriasis vul-
cebo-controlled trial. J Clin Immunol 2015;35:399–407. garis. Arch Dermatol 2003;139:1433–9.
269. Akl A, Luo S, Wood K. Induction of transplantation tolerance: the 293. Harlan D, Kirk A. The future of organ and tissue transplantation:
potential of regulatory T cells. Transpl Immunol 2005;14:225–30. can T-cell costimulatory pathway modifiers revolutionize the pre-
270. Waldmann H. Therapeutic approaches for transplantation. Curr vention of graft rejection? JAMA 1999;282:1076–82.
Opin Immunol 2001;13:606–10. 294. June C, Ledbetter J, Gillespie M, et al. T-cell proliferation involving
271. Madsen J, Peugh W, Wood K, Morris P. The effect of anti-L3T4 the CD28 pathway is associated with cyclosporine-resistant interleu-
monoclonal antibody treatment on first set rejection of murine car- kin 2 gene expression. Mol Cell Biol 1987;7:4472–81.
diac allografts. Transplantation 1987;44:849–52. 295. Walunas T, Bakker C, Bluestone J. CTLA-4 ligation blocks CD28-
272. Sablinski T, Hancock W, Tilney N, Kupiec-Weglinski J. CD4 mono- dependent T cell activation. J Exp Med 1996;183:2541–50.
clonal antibodies in organ transplantation: a review of progress. 296. Armitage R, Fanslow W, Strockbine L, et al. Molecular and biologi-
Transplantation 1991;52:579–89. cal characterization of a murine ligand for CD40. Nature 1992;357:
273. Shizuru J, Seydel K, Flavin T, et al. Induction of donor-specific unre- 80–2.
sponsiveness to cardiac allografts in rats by pretransplant anti-CD4 297. Graf D, Korthauer U, Mages H, et al. Cloning of TRAP, a ligand for
monoclonal antibody therapy. Transplantation 1990;50:366–73. CD40 on human T cells. Eur J Immunol 1992;22:3191–4.
274. Wood K, Pearson T, Darby C, Morris P. CD4: a potential target 298. Henn V, Slupsky J, Grafe M, et al. CD40 ligand on activated plate-
molecule for immunosuppressive therapy and tolerance induction. lets triggers an inflammatory reaction of endothelial cells. Nature
Transplant Rev 1991;5:150–64. 1998;391:591–4.
310 Kidney Transplantation: Principles and Practice

299. Kirk A, Blair P, Tadaki D, et al. The role of CD154 in organ trans- 323. Hodi F, Mihm M, Soiffer R, et al. Biologic activity of cytotoxic T lym-
plant rejection and acceptance. Philos Trans R Soc Lond B Biol Sci phocyte-associated antigen 4 antibody blockade in previously vac-
2001;356:691–702. cinated metastatic melanoma and ovarian carcinoma patients. Proc
300. Cella M, Scheidegger D, Palmer-Lehmann K, et al. Ligation of CD40 Natl Acad Sci U S A 2003;100:4712–7.
on dendritic cells triggers production of high levels of interleukin-12 324. Hodi F, O’Day S, McDermott D, et al. Improved survival with ipi-
and enhances T cell stimulatory capacity: T-T help via APC activa- limumab in patients with metastatic melanoma. N Engl J Med
tion. J Exp Med 1996;184:747–52. 2010;363:711–23.
301. Charafeddine A, Kim E, Maynard D, et al. Platelet-derived CD154: 325. Poirier N, Blancho G, Vanhove B. CD28-specific immunomodulat-
ultrastructural localization and clinical correlation in organ trans- ing antibodies: what can be learned from experimental models? Am J
plantation. Am J Transplant 2012;12:3143–51. Transplant 2012;12:1682–90.
302. Xu H, Zhang X, Mannon R, Kirk A. Platelet-derived or soluble CD154 326. Poirier N, Mary C, Dilek N, et al. Preclinical efficacy and immuno-
induces vascularized allograft rejection independent of cell-bound logical safety of FR104, an antagonist anti-CD28 monovalent Fab’
CD154. J Clin Invest 2006;116:769–74. antibody. Am J Transplant 2012;12:2630–40.
303. Kirk A, Burkly L, Batty D, et al. Treatment with humanized monoclo- 327. Siddiqui M, Scott L. Infliximab: a review of its use in Crohn’s disease
nal antibody against CD154 prevents acute renal allograft rejection and rheumatoid arthritis. Drugs 2005;65:2179–208.
in nonhuman primates. Nat Med 1999;5:686–93. 328. Weinberg J, Bottino C, Lindholm J, Buchholz R. Biologic therapy for
304. Kirk A, Harlan D, Armstrong N, et al. CTLA4-Ig and anti-CD40 psoriasis: an update on the tumor necrosis factor inhibitors inflix-
ligand prevent renal allograft rejection in primates. Proc Natl Acad imab, etanercept, and adalimumab, and the T-cell-targeted thera-
Sci U S A 1997;94:8789–94. pies efalizumab and alefacept. J Drugs Dermatol 2005;4:544–55.
305. Larsen C, Elwood E, Alexander D, et al. Long-term acceptance of 329. Couriel D, Saliba R, Hicks K, et al. Tumor necrosis factor alpha
skin and cardiac allografts after blocking CD40 and CD28 pathways. blockade for the treatment of steroid-refractory acute GVHD. Blood
Nature 1996;381:434–8. 2004;104:649–54.
306. Lo D, Anderson D, Weaver T, et al. Belatacept and sirolimus prolong 330. Froud T, Ricordi C, Baidal D, et al. Islet transplantation in type 1 dia-
nonhuman primate renal allograft survival without a requirement betes mellitus using cultured islets and steroid-free immunosuppres-
for memory T cell depletion. Am J Transplant 2013;13:320–8. sion: Miami experience. Am J Transplant 2005;5:2037–46.
307. Birsan T, Hausen B, Higgins J, et al. Treatment with humanized 331. Pascher A, Radke C, Dignass A, et al. Successful infliximab treatment
monoclonal antibodies against CD80 and CD86 combined with of steroid and OKT3 refractory acute cellular rejection in two patients
sirolimus prolongs renal allograft survival in cynomolgus monkeys. after intestinal transplantation. Transplantation 2003;76:615–8.
Transplantation 2003;75:2106–13. 332. Jacobsohn D, Vogelsang G. Anti-cytokine therapy for the treatment
308. Hausen B, Klupp J, Christians U, et al. Coadministration of either of graft-versus-host disease. Curr Pharm Des 2004;10:1195–205.
cyclosporine or steroids with humanized monoclonal antibod- 333. Bellin M, Kandaswamy R, Parkey J, et al. Prolonged insulin indepen-
ies against CD80 and CD86 successfully prolong allograft survival dence after islet allotransplants in recipients with type 1 diabetes.
after life supporting renal transplantation in cynomolgus monkeys. Am J Transplant 2008;8:2463–70.
Transplantation 2001;72:1128–37. 334. Rickels M, Liu C, Shlansky-Goldberg R, et al. Improvement in β-cell
309. Ossevoort M, Ringers J, Kuhn E, et al. Prevention of renal allograft secretory capacity after human islet transplantation according to
rejection in primates by blocking the B7/CD28 pathway. Transplan- the CIT07 protocol. Diabetes 2013;62:2890–7.
tation 1999;68:1010–8. 335. Weber J, Keam S. Ustekinumab. BioDrugs 2009;23:53–61.
310. Ezzelarab M, Raich-Regue D, Lu L, et al. Renal allograft survival in 336. Leonardi C, Kimball A, Papp K, et al. Efficacy and safety of
nonhuman primates infused with donor antigen-pulsed autologous ustekinumab, a human interleukin-12/23 monoclonal antibody,
regulatory dendritic cells. Am J Transplant 2017;17:1476–89. in patients with psoriasis: 76-week results from a randomised,
311. Preston E, Xu H, Dhanireddy K, et al. IDEC-131 (anti-CD154), siro- double-blind, placebo-controlled trial (PHOENIX 1). Lancet
limus and donor-specific transfusion facilitate operational tolerance 2008;371:1665–74.
in non-human primates. Am J Transplant 2005;5:1032–41. 337. Venkiteshwaran A. Tocilizumab. MAbs 2009;1:432–8.
312. Xu H, Montgomery S, Preston E, et al. Studies investigating pretrans- 338. Choi J, Aubert O, Vo A, et al. Assessment of tocilizumab (anti-
plant donor-specific blood transfusion, rapamycin, and the CD154- interleukin-6 receptor monoclonal) as a potential treatment for
specific antibody IDEC-131 in a nonhuman primate model of skin chronic antibody-mediated rejection and transplant glomerulopa-
allotransplantation. J Immunol 2003;170:2776–82. thy in HLA-sensitized renal allograft recipients. Am J Transplant
313. Kirk A, Knechtle S, Sollinger H, et al. Preliminary results of the use 2017;17:2381–9.
of humanized anti-CD154 in human renal allotransplantation. Am J 339. Vo A, Choi J, Kim I, et al. A phase I/II trial of the interleukin-6 recep-
Transplant 2001;1:191. tor-specific humanized monoclonal (tocilizumab) + intravenous
314. Adams A, Shirasugi N, Jones T, et al. Development of a chimeric anti- immunoglobulin in difficult to desensitize patients. Transplantation
CD40 monoclonal antibody that synergizes with LEA29Y to prolong 2015;99:2356–63.
islet allograft survival. J Immunol 2005;174:542–50. 340. Navarra S, Guzman R, Gallacher A, et al. Efficacy and safety of
315. Aoyagi T, Yamashita K, Suzuki T, et al. A human anti-CD40 monoclonal belimumab in patients with active systemic lupus erythema-
antibody, 4D11, for kidney transplantation in cynomolgus monkeys: tosus: a randomised, placebo-controlled, phase 3 trial. Lancet
Induction and maintenance therapy. Am J Transplant 2009;2009:8. 2011;377:721–31.
316. Imai A, Suzuki T, Sugitani A, et al. A novel fully human anti-CD40 341. Berlin P, Bacher J, Sharrow S, et al. Monoclonal antibodies against
monoclonal antibody, 4D11, for kidney transplantation in cynomol- human T cell adhesion molecules: modulation of immune function
gus monkeys. Transplantation 2007;84:1020–8. in nonhuman primates. Transplantation 1992;53:840–9.
317. Kirk A. 4D11: the second mouse? Am J Transplant 2009;9:1701–12. 342. Marlin S, Springer T. Purified intercellular adhesion molecule-1
318. Oura T, Yamashita K, Suzuki T, et al. Long-term hepatic allograft (ICAM-1) is a ligand for lymphocyte function-associated antigen 1
acceptance based on CD40 blockade by ASKP1240 in nonhuman (LFA-1). Cell 1987;51:813–9.
primates. Am J Transplant 2012;12:1740–54. 343. Rothlein R, Dustin M, Marlin S, Springer T. A human intercellu-
319. Cordoba F, Wieczorek G, Audet M, et al. A novel, blocking, Fc-silent lar adhesion molecule (ICAM-1) distinct from LFA-1. J Immunol
anti-CD40 monoclonal antibody prolongs nonhuman primate renal 1986;137:1270–4.
allograft survival in the absence of B cell depletion. Am J Transplant 344. Springer T, Dustin M, Kishimoto T, Marlin S. The lymphocyte function-
2015;15:2825–36. associated LFA-1, CD2, and LFA-3 molecules: cell adhesion receptors
320. Kirk A, Tadaki D, Celniker A, et al. Induction therapy with monoclo- of the immune system. Annu Rev Immunol 1987;5:223–52.
nal antibodies specific for CD80 and CD86 delays the onset of acute 345. Nakajima H, Sano H, Nishimura T, et al. Role of vascular cell adhe-
renal allograft rejection in non-human primates. Transplantation sion molecule 1/very late activation antigen 4 and intercellular
2001;72:377–84. adhesion molecule 1/lymphocyte function-associated antigen 1
321. Clarkson M, Sayegh M. T-cell costimulatory pathways in allograft interactions in antigen-induced eosinophil and T cell recruitment
rejection and tolerance. Transplantation 2005;80:555–63. into the tissue. J Exp Med 1994;179:1145–54.
322. Monk N, Hargreaves R, Marsh J, et al. Fc-dependent depletion of acti- 346. Isobe M, Yagita H, Okumura K, Ihara A. Specific acceptance of car-
vated T cells occurs through CD40L-specific antibody rather than diac allograft after treatment with antibodies to ICAM-1 and LFA-1.
costimulation blockade. Nat Med 2003;9:1275–80. Science 1992;255:1125–7.
19 • Antilymphocyte Globulin, Monoclonal Antibodies, and Fusion Proteins 311

347. Qin L, Chavin K, Lin J, et al. Anti-CD2 receptor and anti-CD2 ligand 370. Waid T, Lucas B, Thompson J, et al. Treatment of renal allograft
(CD48) antibodies synergize to prolong allograft survival. J Exp Med rejection with T10B9.1A31 or OKT3: final analysis of a phase II
1994;179:341–6. clinical trial. Transplantation 1997;64:274–81.
348. Reichert J. Therapeutic monoclonal antibodies: trends in develop- 371. Waid T, Thompson J, McKeown J, et al. Induction immunotherapy
ment and approval in the US. Curr Opin Mol Ther 2002;4:110–8. in heart transplantation with T10B9.1A-31: a phase I study. J Heart
349. Cosimi A, Conti D, Delmonico F, et al. In vivo effects of monoclo- Lung Transplant 1997;16:913–6.
nal antibody to ICAM-1 (CD54) in nonhuman primates with renal 372. Godder K, Henslee-Downey P, Mehta J, et al. Long term disease-free
allografts. J Immunol 1990;144:4604–12. survival in acute leukemia patients recovering with increased gam-
350. Badell I, Russell M, Thompson P, et al. LFA-1-specific therapy pro- madelta T cells after partially mismatched related donor bone mar-
longs allograft survival in rhesus macaques. J Clin Invest 2010; row transplantation. Bone Marrow Transplant 2007;39:751–7.
120:4520–31. 373. Thompson J, Pomeroy C, Kryscio R, et al. Use of a T cell-specific
351. Haug C, Colvin R, Delmonico F, et al. A phase I trial of immunosup- monoclonal antibody, T10B9, in a novel allogeneic stem cell trans-
pression with anti-ICAM-1 (CD54) mAb in renal allograft recipients. plantation protocol for hematologic malignancy high-risk patients.
Transplantation 1993;55:766–72. Biol Blood Marrow Transplant 2004;10:858–66.
352. Salmela K, Wramner L, Ekberg H, et al. A randomized multicenter 374. Siemenow M, Brown S, Thompson J, et al. TOL101;a novel αβ TCR
trial of the anti-ICAM-1 monoclonal antibody (Enlimomab), for targeting monoclonal antibody. Am J Transplant 2010;10:36.
the prevention of acute rejection and delayed onset of graft func- 375. Getts D, Wiseman A, Mulgaonkar S, et al. Evaluating safety and effi-
tion in cadaveric renal transplantation: a report of the european cacy of TOL101 induction to prevent kidney transplant rejection,
anti-ICAM-1 renal transplant study group. Transplantation 1999; part a interim analysis. Transplant Int 2011;24:93.
67:729–36. 376. Flechner S, Mulgoankar S, Melton L, et al. Replace with First-in-
353. Hourmant M, Bedrossian J, Durand D, et al. A randomized mul- human study of the safety and efficacy of TOL101 induction to prevent
ticenter trial comparing leukocyte function-associated antigen-1 kidney transplant rejection. Am J Transplant 2014;14:1346–55.
monoclonal antibody with rabbit antithymocyte globulin as induc- 377. Colvin R, Smith R. Antibody-mediated organ-allograft rejection. Nat
tion treatment in first kidney transplantations. Transplantation Rev Immunol 2005;5:807–17.
1996;62:1565–70. 378. Pratt J, Basheer S, Sacks S. Local synthesis of complement com-
354. Mauff BL, Hourmant M, Rougier J, et al. Effect of anti-LFA1 (CD11a) ponent C3 regulates acute renal transplant rejection. Nat Med
monoclonal antibodies in acute rejection in human kidney trans- 2002;8:582–7.
plantation. Transplantation 1991;52:291–6. 379. Brown K, Kondeatis E, Vaughan R, et al. Influence of donor C3
355. Dedrick R, Walicke P, Garovoy M. Anti-adhesion antibodies efali- allotype on late renal-transplantation outcome. N Engl J Med
zumab, a humanized anti-CD11a monoclonal antibody. Transpl 2006;354:2014–23.
Immunol 2002;9:181–6. 380. Hill A, Hillmen P, Richards S, et al. Sustained response and long-
356. Nicolls M, Gill R. LFA-1 (CD11a) as a therapeutic target. Am J Trans- term safety of eculizumab in paroxysmal nocturnal hemoglobinuria.
plant 2006;6:27–36. Blood 2005;106:2559–65.
357. Vincenti F, Mendez R, Pescovitz M, et al. A phase I/II randomized 381. Woodle E, Baldwin Wr. Of mice and men: terminal complement
open-label multicenter trial of efalizumab, a humanized anti-CD11a, inhibition with anti-C5 monoclonal antibodies. Am J Transplant
anti-LFA-1 in renal transplantation. Am J Transplant 2007;7:1770–7. 2011;11:2277–8.
358. Turgeon N, Avila J, Cano J, et al. Experience with a novel efalizumab- 382. Reiss U, Schwartz J, Sakamoto K, et al. Efficacy and safety of ecu-
based immunosuppressive regimen to facilitate single donor islet cell lizumab in children and adolescents with paroxysmal nocturnal
transplantation. Am J Transplant 2010;10:2082–91. hemoglobinuria. Pediatr Blood Cancer 2014;61:1544–50.
359. Posselt A, Bellin M, Tavakol M, et al. Islet transplantation in type 1 383. Brodsky R, Young N, Antonioli E, et al. Multicenter phase 3
diabetics using an immunosuppressive protocol based on the anti- study of the complement inhibitor eculizumab for the treatment
LFA-1 antibody efalizumab. Am J Transplant 2010;10:1870–80. of patients with paroxysmal nocturnal hemoglobinuria. Blood
360. Polman C, O’Connor P, Havrdova E, et al. A randomized, placebo- 2008;111:1840–7.
controlled trial of natalizumab for relapsing multiple sclerosis. 384. Hillmen P, Young N, Schubert J, et al. The complement inhibitor
N Engl J Med 2006;354:899–910. eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med
361. Kitchens W, Haridas D, Wagener M, et al. Integrin antagonists 2006;355:1233–43.
prevent costimulatory blockade-resistant transplant rejection by 385. Legendre C, Licht C, Muus P, et al. Terminal complement inhibitor
CD8(+) memory T cells. Am J Transplant 2012;12:69–80. eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med
362. Carmody I, Meng L, Shen X, et al. P-selectin knockout mice have 2013;368:2169–81.
improved outcomes with both warm ischemia and small bowel 386. Krid S, Roumenina L, Beury D, et al. Renal transplantation under
transplantation. Transplant Proc 2004;36:263–4. prophylactic eculizumab in atypical hemolytic uremic syndrome with
363. Dulkanchainun T, Goss J, Imagawa D, et al. Reduction of hepatic CFH/CFHR1 hybrid protein. Am J Transplant 2012;12:1938–44.
ischemia/reperfusion injury by a soluble P-selectin glycoprotein 387. Locke J, Magro C, Singer A, et al. The use of antibody to complement
ligand-1. Ann Surg 1998;227:832–40. protein C5 for salvage treatment of severe antibody-mediated rejec-
364. Farmer D, Anselmo D, Shen XD, et al. Disruption of P-selectin signal- tion. Am J Transplant 2009;9:231–5.
ing modulates cell trafficking and results in improved outcomes after 388. Lonze B, Dagher N, Simpkins C, et al. Eculizumab, bortezomib and
mouse warm intestinal ischemia and reperfusion injury. Transplan- kidney paired donation facilitate transplantation of a highly sensitized
tation 2005;80:828–35. patient without vascular access. Am J Transplant 2010;10:2154–60.
365. Busuttil R, Lipshutz G, Kupiec-Weglinski J, et al. rPSGL-Ig for 389. Lonze B, Singer A, Montgomery R. Eculizumab and renal transplan-
improvement of early liver allograft function: a double-blind, tation in a patient with CAPS. N Engl J Med 2010;362:1744–5.
placebo-controlled, single-center phase II study. Am J Transplant 390. Stegall M, Diwan T, Raghavaiah S, et al. Terminal complement
2011;11:786–97. inhibition decreases antibody-mediated rejection in sensitized renal
366. Gaber A, Mulgaonkar S, Kahan B, et al. YSPSL (rPSGL-Ig) for transplant recipients. Am J Transplant 2011;11:2405–13.
improvement of early renal allograft function: a double-blind, 391. Zimmerhackl L, Hofer J, Cortina G, et al. Prophylactic eculizumab
placebo-controlled, multi-center phase IIa study. Clin Transplant after renal transplantation in atypical hemolytic-uremic syndrome.
2011;25:523–33. N Engl J Med 2010;362:1746–8.
367. Kirk A, Ibrahim S, Dawson D, et al. Characterization of T cells 392. Zuber J, Quintrec ML, Krid S, et al. Eculizumab for atypical hemolytic
expressing the antigen receptor in human renal allograft rejection. uremic syndrome recurrence in renal transplantation. Am J Trans-
Hum Immunol 1993;36:11–9. plant 2012;12:3337–54.
368. Hall B, Hand S, Alter M, et al. Variables affecting the T cell receptor 393. Cornell L, Schinstock C, Gandhi M, et al. Positive crossmatch kidney
V beta repertoire heterogeneity of T cells infiltrating human renal transplant recipients treated with eculizumab: outcomes beyond 1
allografts. Transpl Immunol 1993;1:217–27. year. Am J Transplant 2015;15:1293–302.
369. Brown S, Lucas B, Waid T, et al. T10B9 (MEDI-500) mediated immu- 394. Kulkarni S, Kirkiles-Smith N, Deng Y, et al. Eculizumab therapy for
nosuppression: studies on the mechanism of action. Clin Transplant chronic antibody-mediated injury in kidney transplant recipients: a
1996;10:607–13. pilot randomized controlled trial. Am J Transplant 2017;17:682–91.
312 Kidney Transplantation: Principles and Practice

395. Levi C, Fremeaux-Bacchi V, Zuber J, et al. Midterm outcomes of beads and its clinical application for prevention of acute graft-vs.-
12 renal transplant recipients treated with eculizumab to pre- host disease in allogeneic bone marrow transplantation: results of a
vent atypical hemolytic syndrome recurrence. Transplantation phase I–II trial. Int J Hematol 1999;69:27–35.
2017;101:2924–30. 415. Rodriguez P, Prada D, Moreno E, et al. The anti-CD6 antibody itoli-
396. Yelken B, Arpali E, Gorcin S, et al. Eculizumab for treatment of refrac- zumab provides clinical benefit without lymphopenia in rheumatoid
tory antibody-mediated rejection in kidney transplant patients: a arthritis patients: results from a 6-month, open-label phase I clinical
single-center experience. Transplant Proc 2015;47:1754–9. trial. Clin Exp Immunol 2018;191(2):229–39. Available online at:
397. Pruitt S, Kirk A, Bollinger R, et al. The effect of soluble complement https://doi.org/10.1111/cei.13061.
receptor type 1 on hyperacute rejection of porcine xenografts. Trans- 416. Sempowski G, Lee D, Kaufman R, Haynes B. Structure and function
plantation 1994;57:363–70. of the CD7 molecule. Crit Rev Immunol 1999;19:331–48.
398. Thomas K, Valenzuela N, Gjertson D, et al. An anti-C1s monoclo- 417. Stillwell R, Bierer B. T cell signal transduction and the role of CD7 in
nal, TNT003, inhibits complement activation induced by antibodies costimulation. Immunol Res 2001;24:31–52.
against HLA. Am J Transplant 2015;15:2037–49. 418. Lazarovits A, Rochon J, Banks L, et al. Human mouse chimeric CD7
399. Wahrmann M, Muhlbacher J, Marinova L, et al. Effect of the anti- monoclonal antibody for the prophylaxis of kidney transplant rejec-
C1s humanized antibody TNT009 and its parental mouse variant tion. J Immunol 1993;150:5163–74.
TNT003 on HLA antibody–induced complement activation: a pre- 419. Sharma L, Muirhead N, LAzarovits A. Human mouse chimeric CD7
clinical in vitro study. Am J Transplant 2017;17:2300–11. monoclonal antibody (SDZCHH380) for the prophylaxis of kid-
400. Eskandary F, Jilma B, Muhlbacher J, et al. Anti-C1s monoclonal ney transplant rejection: analysis beyond 4 years. Transplant Proc
antibody BIVV009 in late antibody-mediated kidney allograft rejec- 1997;29:323–4.
tion: results from a first-in-patient phase 1 trial. Am J Transplant 420. Rocha P, Plumb T, Crowley S, Coffman T. Effector mechanisms in
2018;18(4):916–26. Available online at: https://doi.org/10.1111/ transplant rejection. Immunol Rev 2003;196:51–64.
ajt.14528. 421. Wee S, Colvin R, Phelan J, et al. Fc-receptor for mouse IgG1 (Fc
401. Montgomery R, Orandi B, Racusen L, et al. Plasma-derived C1 ester- gamma RII) and antibody-mediated cell clearance in patients treated
ase inhibitor for acute antibody-mediated rejection following kidney with Leu2a antibody. Transplantation 1989;48:1012–7.
transplantation: results of a randomized double-blind placebo-con- 422. Allan J, Choo J, Vesga L, et al. Cardiac allograft vasculopathy is abro-
trolled pilot study. Am J Transplant 2016;16:3468–78. gated by anti-CD8 monoclonal antibody therapy. Ann Thorac Surg
402. Viglietti D, Gosset C, Loupy A, et al. C1 inhibitor in acute antibody- 1997;64:1019–25.
mediated rejection nonresponsive to conventional therapy in kid- 423. Nimer S, Giorgi J, Gajewski J, et al. Selective depletion of CD8 cells for
ney transplant recipients: a pilot study. Am J Transplant 2016;16: prevention of graft-versus-host disease after bone marrow transplan-
1596–603. tation: a randomized controlled trial. Transplantation 1994;57:82–7.
403. Vo A, Zeevi A, Choi J, et al. A phase I/II placebo-controlled trial of 424. Luke P, O’Brien C, Jevnikar A, Zhong R. Anti-CD45RB monoclo-
C1-inhibitor for prevention of antibody-mediated rejection in HLA nal antibody-mediated transplantation tolerance. Curr Mol Med
sensitized patients. Transplantation 2015;99:299–308. 2001;1:533–43.
404. Jordan S, Lorant T, Choi J. IgG endopeptidase in highly sensitized 425. Chen G, Luke P, Yang H, et al. Anti-CD45RB monoclonal antibody
patients undergoing transplantation. N Engl J Med 2017;377:1693–4. prolongs renal allograft survival in cynomolgus monkeys. Am J
405. Raman C. CD5, an important regulator of lymphocyte selection and Transplant 2007;7:27–37.
immune tolerance. Immunol Res 2002;26:255–63. 426. Jian Y, Ye J, Qi H, et al. Anti-CD45RB and donor-specific spleen cells
406. Martin P, Nelson B, Appelbaum F, et al. Evaluation of a CD5-specific transfusion inhibition allograft skin rejection mediated by memory T
immunotoxin for treatment of acute graft-versus-host disease after cells. Immunol Cell Biol 2017;95:189–97.
allogeneic marrow transplantation. Blood 1996;88:824–30. 427. Gregori S, Mangia P, Bacchetta R, et al. An anti-CD45RO/RB mono-
407. Olsen N, Brooks R, Cush J, et al. A double-blind, placebo-controlled clonal antibody modulates T cell responses via induction of apoptosis
study of anti-CD5 immunoconjugate in patients with rheuma- and generation of regulatory T cells. J Exp Med 2005;201:1293–305.
toid arthritis. the xoma RA investigator group. Arthritis Rheum 428. Kreitman R. Toxin-labeled monoclonal antibodies. Curr Pharm Bio-
1996;39:1102–8. technol 2001;2:313–25.
408. Przepiorka D, LeMaistre C, Huh Y, et al. Evaluation of anti-CD5 ricin 429. Arons E, Sorbara L L, Raffeld M, et al. Characterization of T-cell
A chain immunoconjugate for prevention of acute graft-vs.-host repertoire in hairy cell leukemia patients before and after recombi-
disease after HLA-identical marrow transplantation. Ther Immunol nant immunotoxin BL22 therapy. Cancer Immunol Immunother
1994;1:77–82. 2005;55:1100–10.
409. Stafford F, Fleisher T, Lee G, et al. A pilot study of anti-CD5 ricin A 430. Kreitman R, Wilson W, White J, et al. Phase I trial of recombinant
chain immunoconjugate in systemic lupus erythematosus. J Rheu- immunotoxin anti-Tac(Fv)-PE38 (LMB-2) in patients with hemato-
matol 1994;21:2068–670. logic malignancies. J Clin Oncol 2000;18:1622–36.
410. Osorio L, Rottenberg M, Jondal M, Chow S. Simultaneous cross- 431. Schnell R, Vitetta E, Schindler J, et al. Treatment of refractory Hodg-
linking of CD6 and CD28 induces cell proliferation in resting T cells. kin’s lymphoma patients with an anti-CD25 ricin A-chain immuno-
Immunology 1998;93:358–65. toxin. Leukemia 2000;14:129–35.
411. Starling G, Whitney G, Siadak A, et al. Characterization of mouse 432. Wayne A, Shah N, Bhojwani D, et al. Phase 1 study of the anti-CD22
CD6 with novel monoclonal antibodies which enhance the alloge- immunotoxin moxetumomab pasudotox for childhood acute lym-
neic mixed leukocyte reaction. Eur J Immunol 1996;26:738–46. phoblastic leukemia. Blood 2017;130:1620–7.
412. Kirkman R, Araujo J, Busch G, et al. Treatment of acute renal 433. Thomas J, Neville D, Contreras J, et al. Preclinical studies of allograft
allograft rejection with monoclonal anti-T12 antibody. Transplan- tolerance in rhesus monkeys: a novel anti-CD3-immunotoxin given
tation 1983;36:620–6. peritransplant with donor bone marrow induces operational toler-
413. Patel N, Chinen J, Rosenblatt H, et al. Long-term outcomes of non- ance to kidney allografts. Transplantation 1997;64:124–35.
conditioned patients with severe combined immunodeficiency 434. Torrealba J, Fernandez L, Kanmaz T, et al. Immunotoxin-treated
transplanted with HLA-identical or haploidentical bone marrow rhesus monkeys: a model for renal allograft chronic rejection. Trans-
depleted of T cells with anti-CD6 mAb. J Allergy Clin Immunol plantation 2003;76:524–30.
2008;122:1185–93. 435. Brown K, Nowocin A, Meader L, et al. Immunotoxin against a donor
414. Sao H, Kitaori K, Kasai M, et al. A new marrow T cell depletion MHC class II molecule induces indefinite survival of murine kidney
method using anti-CD6 monoclonal antibody-conjugated magnetic allografts. Am J Transplant 2016;16:1129–38.
20 Other Forms of
Immunosuppression
BEN SPRANGERS, JACQUES PIRENNE, CHANTAL MATHIEU, and
MARK WAER

CHAPTER OUTLINE Small Molecules Conclusion


Leflunomide and Malononitrilamides AEB071 or Sotrastaurin
Chemical Structure and Pharmacology Chemical Structure and Pharmacology
Mechanism of Action Mechanism of Action
Experimental Experience Experimental Experience
Clinical Experience Clinical Experience
Toxicity Toxicity
Conclusion Conclusion
FTY720 or Fingolimod Bortezomib
Chemical Structure and Pharmacology Chemical Structure and Pharmacology
Mechanism of Action Mechanism of Action
Experimental Experience Experimental Experience
Clinical Experience Clinical Experience
Toxicity Toxicity
Conclusion Conclusion
Bredinin or Mizoribine Others
Chemical Structure and Pharmacology Total Lymphoid Irradiation
Mechanism of Action Procedure
Experimental and Clinical Experience Mechanisms of Action
Toxicity Experimental Experience
Conclusion Clinical Experience
JAK3 Inhibitors: Tofacitinib Conclusion
Chemical Structure and Pharmacology Photopheresis
Mechanism of Action Splenectomy
Experimental Experience Plasmapheresis
Clinical Experience
Toxicity

Small Molecules vs. 15–18 days) it was believed to represent an attrac-


tive alternative to leflunomide for application in organ
LEFLUNOMIDE AND MALONONITRILAMIDES
transplantation.2
Leflunomide, initially developed as an agriculture herbicide,
was explored as an immunosuppressant because of its abil- Chemical Structure and Pharmacology
ity to inhibit the enzyme dihydroorotate dehydrogenase.1 Leflunomide (N-(4)) trifluoro-methylphenyl-5-methyl-
The potential of leflunomide as an immunosuppressant in isoxawol-4-carboximide) is a prodrug and is rapidly con-
the field of transplantation was extensively demonstrated in verted to its biologically active metabolite teriflunomide
various experimental studies, but its long half-life (several (A771703). Serum levels of teriflunomide are referred to
days) poses the problem of potential overimmunosuppres- as leflunomide levels. The half-life of teriflunomide is long
sion in transplant patients. Analogs of the active metabolite in humans (approximately 15 days). The drug enters the
of leflunomide have been developed and are called malo- enterohepatic recirculation and is excreted by the intestinal
nonitrilamides (MNAs). FK778 (also known as MNA715 and urinary systems in equal proportions. Leflunomide is
or HMR1715) is the best studied synthetic MNA, and as it insoluble in water and is suspended in 1% carboxymethyl-
has a much shorter half-life than leflunomide (6–45 hours cellulose for oral administration.
313
314 Kidney Transplantation: Principles and Practice

The MNAs are designed to be structurally similar to Interestingly, FK778 and leflunomide have been shown
A771726. Oral bioavailability of FK778 is not substantially to possess antiviral effects, although the precise mechanism
affected by food, and no gender effect on pharmacokinetics is unclear: inhibition of viral replication of members of the
was observed in phase I studies. herpesvirus family by preventing tegument acquisition by
viral nucleocapsids during the late stage of virion assembly
Mechanism of Action has been implicated.17,18 Leflunomide is effective against
Leflunomide and its analogs have strong antiproliferative multidrug-resistant cytomegalovirus (CMV) in vitro,19
effects on both T lymphocytes and B lymphocytes, thus although this in vitro activity is modest and the selectiv-
limiting the formation of antibodies.3,4 Inhibition of pyrimi- ity index is low.20 This anti-CMV effect of leflunomide
dine synthesis is the most important mechanism of action and FK778 was confirmed in a rat model of heterotopic
as leflunomide directly inhibits the enzyme dihydrooro- heart transplantation.21,22 Another interesting feature is
tate dehydrogenase.5 Lymphocytes rely entirely on the that both leflunomide and FK778 have vasculoprotective
de novo pathway of pyrimidine biosynthesis and cannot effects, independent of the inhibition of dihydroorotate
use another, the so-called “pyrimidine salvage pathway.” dehydrogenase.23,24
Dihydroorotate dehydrogenase inhibition leads to depletion
of the nucleotide precursors uridine triphosphate and cyti- Experimental Experience
dine triphosphate, which are necessary for the synthesis of In various rodent transplantation studies, leflunomide was
RNA and DNA, and hence strongly suppress DNA and RNA shown to be at least equally potent as cyclosporine,1 and
synthesis. able to synergize with cyclosporine to induce tolerance.25
The in vivo mechanism of action of leflunomide may Specific characteristics of leflunomide-mediated immu-
depend on factors such as drug levels, disposable uridine nosuppression in rats were its ability to interrupt ongoing
pools, and the immune activation pathway involved. Studies acute rejections,26,27 and its efficacy in preventing and
have indicated that, in addition to inhibition of dihydrooro- treating chronic vascular rejection.28
tate dehydrogenase, leflunomide and the MNAs may act One of the most attractive characteristics of leflunomide
through inhibition of tyrosine kinases. Phosphorylation of and the MNAs is their strong capacity to delay xenograft
the epidermal growth factor receptor of human fibroblasts rejection and to induce partial xenograft tolerance.29 This
has been shown to be inhibited by leflunomide.6 It was may be related to the strong suppressive effects of lefluno-
shown that leflunomide directly inhibited the interleukin mide on T cell-independent xenoantibody formation, and
(IL)-2-stimulated protein tyrosine kinase activity of p56lck on its capacity to induce natural killer (NK) cell nonrespon-
and p59fyn,6 which is associated with activation through siveness and to modulate xenoantigen expression.30
the T cell receptor/CD3 complex. At higher concentrations, Monotherapy with FK778 in rats,31 and its combina-
A771726 also inhibited IL-2-induced tyrosine phosphory- tion with microemulsified cyclosporine in dogs32 or tacro-
lation of Janus kinase (JAK)1 and JAK3 protein tyrosine limus in nonhuman primates,33 reduced chronic allograft
kinases.7 Leflunomide analogs have also been shown to nephropathy31 and significantly prolonged renal allograft
possess strong inhibitory activity on the antiapoptotic survival.31–33
tyrosine kinase Bruton’s tyrosine kinase, a key factor for
T cell-independent antibody formation.8 The hypothesis that Clinical Experience
leflunomide may exhibit more than one mechanism of action The main role of leflunomide in renal transplantation now-
in vivo was further illustrated in mice where uridine restored adays is the treatment of BK virus nephropathy (BKVN),
proliferation and IgM production by lipopolysaccharide- although its efficacy has never been documented in tri-
stimulated B cells, whereas suppression of IgG production als.34–38 Based on the in vitro effective anti-BK concentra-
was not reversed. This phenomenon correlated in a dose- tion, an in vivo target level of 50 to 100 mg/mL has been
dependent manner with tyrosine phosphorylation of JAK3 proposed. In a prospective study, 26 renal transplant recipi-
and STAT6 proteins, known to be involved in IL-4-induced ents with biopsy-proven BKVN were treated with leflu-
signal transduction pathways.4 This double in vivo mecha- nomide in combination with discontinuation of MMF and
nism of action was confirmed in rats, in which xenoreac- reduction of tacrolimus to a 4 to 6 ng/mL range.38 Although
tivity was counteracted by the administration of uridine, the leflunomide levels were in the lower range (on average
whereas alloreactivity was not.9 50 mg/mL), a significant reduction in serum and urine BK
Inhibition of various macrophage functions by leflu- virus (BKV) titers was obtained, allograft function stabi-
nomide and MNAs has also been described; in particular, lized, and the overall graft loss rates because of BKV were
inhibition of the production of oxygen radicals,10–12 the only 15%.35 Less encouraging results were obtained in
inhibition of IgE-mediated hypersensitivity responses,13 the another prospective open-label study in which viral clear-
expression of IL-8 receptor type A, as well as tumor necrosis ance was only obtained in 40% of patients with significant
factor (TNF)-mediated nuclear factor kappa B (NF-κB) acti- toxicity, resulting in discontinuation of the drug in 17% of
vation.14 Tacrolimus also inhibits maturation of dendritic patients.39 The contribution of reduction of immunosup-
cells by preventing upregulation of activation markers and pressive agents and leflunomide to the efficacy of BKVN
IL-12 production, and this phenomenon was not reversible treatment is unclear at this moment.40–42 Based on recent
by exogenous uridine. FK778 has equivalent or stronger in vitro data, it has been suggested that the combination of
immunosuppressive activity than leflunomide, both in vitro mammalian target of rapamycin (mTOR) inhibition with
and in vivo.2 The immunosuppressive effect is synergistic leflunomide might be an effective treatment approach.43
with that of calcineurin inhibitors (CNI) and mycopheno- Treatment with leflunomide in kidney transplant recipients
late mofetil (MMF).15,16 with BK viremia was able to prevent the development of
20 • Other Forms of Immunosuppression 315

BKVN.44 A study from Jaw et al. reported on three kidney a promising class of immunosuppressants but results in
transplant recipients with BKVN who were treated with a randomized clinical trials have been disappointing, and the
combination of leflunomide and everolimus; all patients development of these agents in organ transplantation has
experienced significant reductions in viral loads (one been halted.
with complete resolution) and two patients had preserved
allograft function at the end of follow-up.45 In case reports, FTY720 OR FINGOLIMOD
patients with resistant/refractory CMV infections,46–50
extensive cutaneous warts,51 and a patient with Kaposi’s Chemical Structure and Pharmacology
sarcoma52 have been successfully treated with leflunomide. FTY720 or 2-amino-2-[2-(4-octylphenyl)ethyl]-1,3-pro-
FK778 has also been studied in the context of BKVN, but panediol hydrochloride is a synthetic structural analog of
although it was able to decrease BK viral load, FK778 treat- myriocin, a metabolite of the ascomycete Isaria sinclairii, a
ment was associated with more acute rejections, decreased type of vegetative wasp.62–64 Maximal concentration and
renal function, and more adverse events compared with area under the curve are proportional to the dose, indi-
reduction of immunosuppression.53 cating that the pharmacokinetic profile of FTY720 is lin-
In animal studies, leflunomide was able to reverse acute ear. The volume of distribution is largely superior to the
and chronic rejection. Two clinical studies reported that blood volume, indicating a widespread tissue penetration.
leflunomide was capable of stabilizing allograft function in FTY720 undergoes hepatic metabolism and has a long half-
patients with worsening allograft function due to chronic life (around 100 hours). ASP0028 is a newly developed
allograft dysfunction.54 S1P1/S1P5-selective agonist in Astellas Pharma Inc.
A phase II multicenter study was performed with FK778
involving 149 renal transplant patients,55 where FK778 Mechanism of Action
was combined with tacrolimus and corticosteroids. The FTY720 has a unique mechanism of action as it mainly
patients receiving FK778 experienced a reduced number of affects lymphocyte trafficking.30,65–67 FTY720 acts as
acute rejections, but there was no effect on graft survival at a high-affinity agonist of the sphingosine 1-phosphate
week 16.55 The reduction of acute rejection episodes was receptor-1 (S1PR1 or Edg1). Binding of its receptor
most pronounced in the subgroup in which target levels results in internalization of S1PR1, rendering lympho-
were obtained in the second week. Of note, mean total and cytes unable to respond to the naturally occurring gradi-
low-density lipoprotein cholesterol levels were 20% lower ent of S1P (low concentrations in thymus and secondary
in the FK778 group versus the placebo group.55 The valid- lymphoid organs, high concentrations in lymph and
ity of these results was hampered by the design of the study, plasma) retaining lymphocytes in the low-S1P environ-
and, at this time, the development of FK778 in the field of ment of lymphoid organs.67,68 After FTY720 admin-
organ transplantation has ceased. istration in mice, B and T cells immediately leave the
peripheral blood and migrate to the peripheral lymph
Toxicity nodes, mesenteric lymph nodes, and Peyer’s patches.
Although rats tolerate leflunomide well, dogs readily The cells return to the peripheral blood after withdrawal
develop anemia and gastrointestinal ulcerations. Report- of the drug without undergoing apoptotic death.69 This
edly, the most frequent side effects in arthritis patients altered cell trafficking is accompanied by a reduction of
receiving long-term leflunomide treatment were diarrhea lymphocyte infiltration into grafted organs.69–71 Inter-
(17%), nausea (10%), alopecia (8%), and rash (10%), estingly, lymphocytes treated ex vivo with FTY720 and
leading to a dropout rate of ± 5%.56 Recently, throm- reintroduced in vivo similarly migrate to the peripheral
botic microangiopathy attributed to leflunomide was lymphoid tissues, indicating that FTY720 acts directly
reported in patients treated for BKVN.54 In the phase II on lymphocytes. This process of accelerated homing
study mentioned previously, involving FK778, there was was completely blocked in vivo by coadministration of
a dose-dependent increase in side effects, including ane- anti-CD62L, anti-CD49d, and anti-CD11a monoclonal
mia, hypokalemia, symptomatic myocardial ischemia, and antibody.30 In vitro, FTY720 in the presence of TNF-α
esophagitis.55 Other reported side effects are pneumonitis increases the expression of certain intercellular adhesion
and peripheral neuropathy.57,58 Leflunomide has terato- molecules on human endothelial cells.72 Thus alteration
genic effects in both animals and humans, and a washout of cell trafficking by FTY720 may result not only from its
period with cholestyramine is advised for both women and direct action on lymphocytes, but also from an effect on
men before considering conception.59 Combining lefluno- endothelial cells.
mide with methotrexate might increase the risk for bone Interestingly, it has been suggested that CD4+CD25+
marrow suppression and liver toxicity.60,61 Furthermore, regulatory T cells are differently affected by FTY720 com-
rifampin accelerates the conversion of leflunomide to teri- pared with T-effector cells.73 CD4+CD25+ regulatory T cells
flunomide, and might increase the levels. Combination express lower levels of S1P1 and S1P4 receptors and, hence,
with warfarin potentially increases the international nor- show reduced response to FTY720. Furthermore, in vitro
malized ratio. FTY720-treated CD4+CD25+ T-regulatory cells possess an
increased suppressive activity in an antigen-specific prolif-
Conclusion eration assay.73,74
The role of leflunomide in renal transplantation is lim- Unlike CNI, FTY720 is a poor inhibitor of T cell func-
ited to the treatment of patients with BKVN and some tion in vitro.75 In particular, FTY720 does not influence
promising results have been reported in this respect. The antigen-induced IL-2 production. In vitro exposure to high
MNAs, because of their shorter half-life, were considered FTY720 concentrations (4 × 10–6) induces chromatin
316 Kidney Transplantation: Principles and Practice

condensation, typical DNA fragmentation, and formation In rats ASP0028 at a dose of 1.0 mg/kg level signifi-
of apoptotic bodies. Whether administration of FTY720 cantly decreased the number of peripheral lymphocytes.
in vivo is also associated with significant apoptosis is a mat- In addition, heart transplant studies in rats indicated that
ter of debate.30,76 ASP0028 combined with suboptimal-dose of tacrolimus
S1PR are also present on murine dendritic cells. Upon significantly prolonged allograft survival, comparable to
administration of FTY720, dendritic cells in lymph nodes that of FTY720 in combination with suboptimal-dose of
and spleen are reduced, the expression of CD11b, CD31/ tacrolimus with a wider margin of safety than FTY720, in
PECAM-1, CD54/ICAM-1, and CCR-7 is downregulated, terms of side effects of macular edema and bradycardia.101
and transendothelial migration to CCL19 is diminished.77 In a study using a cynomolgus monkey renal transplanta-
In a recent study it was demonstrated that FTY720 inhib- tion model, ASP0028 was evaluated in combination with
ited lymphangiogenesis and thus prolonged allogeneic islet suboptimal-dose of tacrolimus. In the animals receiving
survival in mice.78 ASP0028 allograft median survival time was significantly
prolonged.101
Experimental Experience
FTY720 given daily by oral gavage has marked antirejec- Clinical Experience
tion properties in mice, rats, dogs, and monkeys.75,76,79,80 Stable renal transplant patients maintained on cyclosporine
FTY720 (0.1–10 mg/kg) prolongs survival of corneal and tolerate well one oral dose of FTY720 (0.25–3.5 mg). Simi-
skin allografts in highly allogeneic rodent models.81,82 In larly to its effect in animals, single doses of FTY720 cause a
a DA to LEW rat combination, a short course of peritrans- lymphopenia that is dose-dependent in intensity and dura-
plant oral FTY720 (5 mg/kg; days −1 and 0) prolongs tion, and that equally affects CD4 cells, CD8 cells, memory
cardiac allograft survival and is as efficient as a 10-day post- T cells, naïve T cells, and B cells.102
transplant treatment with tacrolimus at 1 mg/kg.83 Car- In phase II and III studies in de novo renal transplanta-
diac and liver allograft survival is prolonged in the August tion, it was shown that 2.5 mg FTY720 in combination
and Copenhagen Irish (ACI) rat to Lew rat model by either with full-dose cyclosporine and steroids is as effective as
induction or maintenance treatment with FTY720.84 Even MMF in combination with full-dose cyclosporine and ste-
delayed administration of FTY720 interrupts an ongoing roids, although the FTY720-treated patients had lower
allograft rejection, suggesting a role for FTY720 as a res- creatinine clearance at 12 months.103,104 FTY720 5 mg did
cue agent.85,86 FTY720 blocks not only rejection but also not allow a 50% reduction in cyclosporine exposure.103,105
graft-versus-host disease after rat intestinal transplanta- FTY720 2.5 mg in combination with reduced-dose cyclo-
tion.87 FTY720 may also protect from ischemia-reperfusion sporine resulted in underimmunosuppression.106 Also in
injury, partially through its cytoprotective actions.88–91 combination with tacrolimus, FTY720 2.5 mg was not
Both small- and large-animal models provide evidence superior to MMF in a recent study in de novo renal trans-
that FTY720 acts in synergy with CNI, and that this benefit plant recipients.107 Recently, it was reported that FTY720
does not result from pharmacokinetic interactions.85 An in combination with everolimus was not beneficial with
induction course with FTY720 acts in synergy with post- regard to prevention of acute rejection and preservation of
transplant tacrolimus in prolonging cardiac allograft sur- allograft function in renal transplant recipients at high risk
vival in rats.86 A similar phenomenon has been observed for delayed graft function.108
when FTY720 is used posttransplant in combination with
cyclosporine in rat skin and heart allografts.85,92 FTY720 Toxicity
shows synergistic effect with CNI in heart and liver trans- The side effects of FTY720 are in general similar to those
plant in the ACI to Lew rat model.80 FTY720 shows syn- of other immunosuppressants, with hypertension, anemia,
ergy with cyclosporine in dog kidney (0.1–5 mg/kg/day) constipation, and nausea most commonly reported. Side
and monkey kidney (0.1–1 mg/kg/day) transplantation.75 effects specific for FTY720 are bradycardia, macular/reti-
FTY720 (0.1 mg/kg) synergizes with CNI in dog liver nal edema, dyspnea, and a transient rise in liver function
transplantation.93 Synergy between FTY720 and rapamy- tests.109,110 Although it is considered a main impediment
cin was also observed in rat cardiac transplantation.94 of further clinical development, reduction of heart rate after
In a murine lung transplant model, FTY720 attenuated the first dose of FTY720 is transient and does not persist
ischemia-reperfusion injury.95 In a sensitized murine in the maintenance phase.109,110 Importantly, typical side
cardiac transplant model, FTY720 in combination with effects of CNI, such as nephrotoxicity, neurotoxicity, and
CTLA4-Ig resulted in inhibition of alloantibody production, diabetogenicity, have not been observed with FTY720.
reduction of donor-specific IFN-γ-producing T cells and pro-
longed allograft survival.96 Conclusion
KRP-203 or 2-amino-2-(2-[4-3(-benzyloxyphenylthio)- FTY720 has a unique mechanism of action. The available
2-cholorophenyl]ethyl)-1,3-propanediol hydrochloride has clinical studies show that FTY720 is not superior to stan-
a similar molecular structure as FTY720. KRP-203 alone dard care and therefore its future in organ transplantation
or in combination with low-dose cyclosporine or MMF pro- is uncertain.
longed skin, heart, and renal allograft survival.97–99 A short
course of KRP203 in BALB/c mice receiving C57BL/10 islet BREDININ OR MIZORIBINE
allografts resulted in significantly prolonged islet allograft
survival.100 Additional injection of intragraft regulatory T Chemical Structure and Pharmacology
cells allowed for prolonged drug-free graft survival, suggest- Bredinin, 4-carbamoyl-1-β-d-ribofurano-syhmidazolium-
ing tolerogenic effects.100 5-olate, is a nucleoside analog that is structurally similar to
20 • Other Forms of Immunosuppression 317

ribavirin. It was first isolated from the culture media of the Given the lymphocyte-restricted role of JAK3, JAK3 inhibi-
ascomycetes Eupenicillium brefeldianum harvested from the tors are considered an interesting novel class of immuno-
soil of Hachijo Island (Japan, 1971). It has weak antibiotic suppressive drugs.
activity against Candida albicans.111
Chemical Structure and Pharmacology
Mechanism of Action Of the multiple potential candidate compounds, one has
Mizoribine exerts its immunosuppressive function through entered clinical trials: the ATP congener tofacitinib (Pfizer,
selective inhibition of the enzymes inosine monophosphate NJ), which binds to the ATP catalytic site on JAK mol-
dehydrogenase and guanosine monophosphate synthetase, ecules. It has been proposed that analysis of P-STAT5 at
both of which are required for the generation of guanosine the cellular level could be an adequate means to monitor
monophosphate from inosine monophosphate in the de the immunomodulatory effect of tofacitinib.131 The oral
novo pathway. In contrast to azathioprine, mizoribine is bioavailability of tofacitinib is above 70% and tofacitinib
not incorporated into nucleic acids in the cells, resulting has a volume of distribution of 87L. Tofacitinib is metabo-
in fewer side effects, such as myelosuppression and hepa- lized primarily by CYP3A4 and to a much lesser extent by
totoxicity. Mizoribine was found to inhibit both humoral CYP2C19, and its metabolites are largely inactive. Due to
and cellular immunity by selectively inhibiting lymphocyte its metabolism through the CYPA4 pathway, drug interac-
proliferation.112 tions in solid-organ transplant recipients can be expected
and dose adjustments are necessary in the presence of azole
Experimental and Clinical Experience antifungals and rifamycins. Dose adjustments are recom-
In a canine model of renal transplantation, mizoribine mended in the context of renal and hepatic failure.
prolonged graft survival. In humans, compared with aza-
thioprine, mizoribine showed equally potent immuno- Mechanism of Action
suppressive activity and fewer adverse effects.113–116 As JAK3 is a tyrosine kinase essential for the signal trans-
expected based on its similarity in structure to ribavirin, duction from the common gamma-chain of the cytokine
mizoribine exhibits in vitro antiviral activity against CMV, receptors for IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21 to the
respiratory syncytial virus, measles, hepatitis C, corona nucleus. As the expression of the receptors is restricted
virus, parainfluenza, and influenza virus.117–120 In a clini- to immune cells, this makes them an attractive target for
cal study, mizoribine was substituted for MMF in patients new immunosuppressants. Signal transduction mediated
with BKV in their urine. BKV DNA in the urine became by JAK3 is obligatory for lymphocyte activation, differen-
negative in five out of seven patients.121 In the remaining tiation, and homeostasis, as evidenced by the finding that
two patients, there was a significant decrease in urinary deficiency in JAK3 results in severe combined deficiency syn-
BKV DNA. No acute rejection or deterioration of graft drome.132–135 A possible detrimental effect of interference
function occurred during the study period.121 Mycophe- with IL-2 signaling relates to the fact that tolerance induc-
nolate substitution with mizoribine has also been shown tion is essentially dependent on the IL-2 pathway.136–138
to be able to prevent and reverse gastrointestinal symp- Thus the challenge for immunosuppressive drug design has
toms.122,123 Japanese studies have suggested different been to achieve selective inhibition of JAK3 versus JAK2
combinations including mizoribine (with cyclosporine/ activities. Tofacitinib was initially claimed to be a selective
basiliximab/corticosteroids or with everolimus/tacroli- JAK3 inhibitor that was found to have 20- to 100-fold less
mus) in living donor kidney transplantation.124,125 activity to both JAK1 and JAK2 based on inhibitory con-
centration 50% (IC50) values; however, numerous other
Toxicity laboratories published inconsistent IC50 results at the vari-
The principal adverse reactions associated with the use of ous JAK domains. Tofacitinib inhibits JAK1 and JAK2 to a
mizoribine were leukopenia, abnormal hepatic function, greater degree than was initially thought and also inhibits
rash, increased levels of uric acid, and vomiting. Hyperuri- Tyk2 marginally.139–144
cemia can result in acute renal failure in renal transplant
recipients treated with high-dose mizoribine.126 Experimental Experience
Tofacitinib is the most potent (inhibitory potency of 1
Conclusion nM) and selective JAK3 inhibitor developed to date. In
Mizoribine has mainly been used in Japan and is infre- both rodents and nonhuman primates, tofacitinib exerted
quently used elsewhere. As a consequence, experience with strong suppression of immune reactions and prolonged
mizoribine is limited, but results show that it is a safe and the survival of cardiac and renal allografts. In monother-
effective immunosuppressant in human kidney transplan- apy it significantly delayed the onset of rejection in kidney
tation. Because of its antiviral activity, mizoribine might be allografts.145–147 In nonhuman primates, tofacitinib signifi-
yet another drug to be evaluated in the setting of BKVN. cantly reduced IL-2-enhanced IFN-γ production and CD25
and CD71 expression by T cells. Furthermore, tofacitinib
inhibited cellular alloimmune responses in vitro.147,148
JAK3 INHIBITORS: TOFACITINIB
Administration in vivo resulted in a reduction of NK cell
Several JAK3 inhibitors have been developed (e.g., tyr- and T cell numbers, whereas CD8 effector memory T cell
phostin AG-490, PNU156804, dimethoxyquinazoline levels were unaltered.148,149 Recently, it was shown that
compounds [WHI-P131], CP-690,550 [tofacitinib], and tofacitinib selectively inhibits T cell effector function while
Mannich base NC1153), and several of them have been preserving the suppressive activity of CD4+CD25high regu-
shown to possess immunosuppressive properties.127–130 latory T cells.150
318 Kidney Transplantation: Principles and Practice

Clinical Experience binding of noncatalytic sites. Time-weighted 2 h postdose


Tofacitinib was evaluated as induction and maintenance concentration therapeutic drug monitoring in tofacitinib-
therapy in a phase IIA, multicenter, open-label, random- treated group is potentially interesting to relaunch tofaci-
ized controlled trial in kidney transplant recipients.151 All tinib as an immunosuppressive drug for kidney transplant
patients received induction therapy, MMF, and steroids. recipients, especially in patients with refractory acute rejec-
Patients in group 1 received tacrolimus, and patients in tion where blockade of IL-15 and IL-7 can prevent the
groups 2 and 3 were administered tofacitinib 15 mg and 30 immunologic skewing toward memory cells.155,156
mg twice per day, respectively.151 Because of the high inci-
dence of BKVN in the tofacitinib groups, the protocol was AEB071 OR SOTRASTAURIN
adjusted to stop MMF and decrease the steroid exposure.
Compared with tacrolimus, both tofacitinib groups showed Chemical Structure and Pharmacology
similar rates of acute rejection episodes and allograft func- Sotrastaurin (AEB071) is a low-molecular-weight, synthetic
tion, but experienced greater incidences of hyperlipidemia compound that potently and reversibly inhibits all 10 isoforms
and infections (suggestive of additional JAK2 inhibition).151 of protein kinase C (PKC), most importantly, PKC-θ PKC-α,
A phase IIB trial compared cyclosporine at standard dosage and PKC-β, with lesser activity on PKC-λ.157 Sotrastaurin is
versus tofacitinib 15 mg twice per day for 6 months then 10 primarily metabolized through hepatic CYP3A4 into inac-
mg twice per day, or tofacitinib 15 mg twice per day for 3 tive metabolites and N-desmethyl-sotrastaurin, which has
months then 10 mg twice per day. In addition, all patients similar potency as sotrastaurin and is present at low blood
received induction therapy, MMF, and steroids. Unexpect- concentrations (less than 5% of the parent exposure). Renal
edly, the lower tofacitinib dosage arm showed a trend toward excretion of sotrastaurin is negligible and only a small
the lowest rejection rate and a superior allograft function. amount is excreted in the bile (1% of the dose). The elimina-
However, in this study there were important adverse events, tion half-life of sotrastaurin averages 6 hours. In clinical tri-
including serious infections and posttransplant lymphopro- als, it is recommended that patients administer sotrastaurin
liferative disease in both tofacitinib arms.152 In a post hoc consistently either with or without food to avoid food-related
analysis of this study patients were reclassified based on fluctuations in drug exposure over time.157
median tofacitinib exposure (2 h postdose concentration Clinical drug interaction studies to date have demon-
more [above-median exposure] or less [below-median expo- strated that sotrastaurin increases the area under the
sure] than 122 mg/mL).153 The incidences of biopsy-proven concentration–time curve of everolimus (1.2-fold) and of
acute rejection were noninferior compared with the cyclo- tacrolimus (2-fold).158 Sotrastaurin increased tacrolimus
sporine A group. However, serious infections and CMV dis- concentration inasmuch as the tacrolimus dose needed to
ease were only significantly increased in the above-median achieve a given C0 was up to 47% lower when combined
exposure group, and posttransplant lymphoproliferative with sotrastaurin versus MMF.158 Conversely, sotrastaurin
disorder (PTLD) cases only occurred in the above-median area under the concentration–time curve is increased up to
exposure group.153 Serious adverse events due to leukope- 1.8-fold by cyclosporine and 4.6-fold by ketoconazole.159
nia and neutropenia remained significant in both the tofaci-
tinib groups compared with the cyclosporine group.153 Mechanism of Action
PKC is a family of serine/threonine-specific protein kinases
Toxicity involved in diverse signal transduction pathways that
In a dose escalation study, the most frequent adverse events modulate a whole range of cellular processes, including
were infection and gastrointestinal side effects. Tofacitinib activation, proliferation, differentiation, apoptosis, and
15 mg and 30 mg twice per day were associated with a autophagy.160 PKC-θ has been shown to be essential in
mean decrease in hemoglobin from baseline of 11%.154 the T cell receptor/CD3 signal transduction pathway. PKC-
There was in addition a decrease in NK cells and B cells. α modulates Th1 responses, including IFN-γ production.
There were no changes in the number of neutrophils, total PKC-β controls B cell receptor-induced NF-κB transacti-
lymphocytes, platelets, CD4 T cells, or CD8 T cells.154 vation and T-independent B cell responses. Finally, PKC-ε
influences macrophage function.161,162
Conclusion
In summary, combination of tofacitinib with MMF resulted Experimental Experience
in acceptable rates of acute rejection with evidence of over- Of note, sotrastaurin was demonstrated to be nontoxic
immunosuppression when tofacitinib 30 mg twice per day when added to human islet cultures.163 These results sug-
was combined with MMF. Tofacitinib 15 mg twice per day gest AEB071 to be an appropriate immunosuppressive can-
coadministered with MMF resulted in similar outcomes didate for clinical trials in islet transplantation. Recently
while associated with modest lipid elevations and a higher it was shown in nonhuman primates that sotrastaurin in
rate of viral infections. In our opinion, considering the combination with cyclosporine at subtherapeutic doses
known side effect profile of CNI, further evaluation of tofaci- resulted in markedly prolonged renal allograft survival,
tinib is warranted. Given the side effects reported with the use indicating synergistic immunosuppressive effects.164
of tofacitinib, the challenge remains to develop immunosup-
pressive drugs with truly selective inhibition of JAK3 versus Clinical Experience
JAK2 activities. The lack of success in developing a selective In a phase II randomized controlled trial, sotrastaurin with
antagonist of JAK3 probably relates to the focus on chemical standard or reduced tacrolimus was compared with stan-
analogs of ATP whereas more success is to be expected from dard tacrolimus alone, in addition to induction therapy and
developing molecules displaying allosteric inhibition via steroids.165 Three months posttransplant, stable patients on
20 • Other Forms of Immunosuppression 319

sotrastaurin were switched from tacrolimus to MMF. The from CNI. However, the efficacy results of this phase II study
three arms showed equal efficacy up to 3 months; at the do not support the combination of sotrastaurin 300 mg
end of the study there was no difference in allograft func- twice per day with MMF as a CNI-free regimen.167 In addi-
tion, although the incidence of acute rejection was signifi- tion, sotrastaurin should be evaluated in nonrenal trans-
cantly higher in the standard tacrolimus with sotrastaurin plant recipients as data suggest it is nonnephrotoxic.169,170
arm.165 Because of lack of efficacy, this study was prema-
turely discontinued. In another recent trial in de novo renal BORTEZOMIB
transplant recipients with immediate graft function, study
subjects were randomized to sotrastaurin or tacrolimus.166 Chemical Structure and Pharmacology
All patients received basiliximab, MMF, and steroids. This Bortezomib is a proteasomal inhibitor approved by the Food
study demonstrated a lower degree of efficacy but better and Drug Administration for the treatment of multiple
renal function with the CNI-free regimen of sotrastaurin myeloma. Bortezomib is increasingly used in the setting of
with MMF versus the tacrolimus-based control.166 The com- solid-organ transplantation. Bortezomib was administered
bination of sotrastaurin with everolimus was evaluated in a in four doses of 1.3 mg/m2 intravenously in 3- to 5-minute
two-stage Phase II study of de novo kidney transplant recipi- infusions over an 11-day period in kidney transplant recipi-
ents evaluating sotrastaurin, in phase 1, 131 patients were ents. Before bortezomib administration, patients need to be
randomized 2:1 to sotrastaurin 300 mg or cyclosporine A premedicated with methylprednisolone. Peak serum con-
(CsA) and in phase II, 180 patients were randomized 1:1:1 centration is reached at 30 minutes and the drug is cleared
to sotrastaurin 300 or 200 mg or CsA. All patients received within 1 hour.171
basiliximab, everolimus (EVR), and prednisone.167 Compos-
ite efficacy failure rates (treated biopsy-proven acute rejec- Mechanism of Action
tion, graft loss, death, or lost to follow-up) at 12 months were Bortezomib is a selective inhibitor of the 26S proteasome,
higher in sotrastaurin arms (Stage 1: 16.5% and 10.9% for preventing the activation of NF-κB.172 It induces apoptosis
sotrastaurin 300 mg and CsA; Stage 2: 27.2%, 34.5%, and of rapidly dividing, metabolically active cells with extensive
19.4% for sotrastaurin 200 mg, 300 mg, and CsA) and protein synthesis. The ability of bortezomib to target plasma
eGFR was significantly better in sotrastaurin groups versus cells spurred interest as a new therapeutic approach in the
CsA at most time points, except at 12 months.167 In another treatment or prevention of alloantibody formation in organ
randomized controlled trial 298 patients were randomized transplantation.
1:1:1:1 to receive sotrastaurin 100 mg or 200 mg twice
per day plus standard tacrolimus (sTAC; 5–12 ng/mL), or Experimental Experience
sotrastaurin 300 mg (n = 75) twice per day plus reduced In vitro, bortezomib was associated with a reduction in the
tacrolimus (rTAC; 2–5 ng/mL), or enteric-coated mycophe- number of bone marrow-derived plasma cells and attenu-
nolic acid (MPA) plus sTAC; all patients received basiliximab ated alloantibody production.173 Bortezomib primarily acts
and corticosteroids.168 The sotrastaurin 100 mg group was through plasma cell depletion, resulting in reduced anti-
prematurely discontinued because of higher composite effi- body production to T-dependent antigens. As marginal zone
cacy failure (treated biopsy-proven acute rejection ≥ grade B cells are resistant to bortezomib-mediated effects, T-inde-
IA, graft loss, death, or loss to follow-up) versus the MPA pendent type 2 responses are less affected.174 In a mouse
group. Sotrastaurin 200 and 300 mg had comparable effi- model of lupus, bortezomib depleted both short- and long-
cacy to MPA in prevention of rejection with no significant lived plasma cells, resulting in a reduction of antidouble-
difference in renal function between the groups.168 stranded DNA antibody production.175 Vogelbacher et al.
recently established that bortezomib alone or in combina-
Toxicity tion with sirolimus can also prevent alloantibody formation
In the clinical trial mentioned previously, there was a in a rat model of kidney transplantation.176
12% incidence of tachycardia and 18% incidence of seri-
ous infections.165,166 A dose-dependent chronotropic effect Clinical Experience
has been observed in preclinical and phase I sotrastaurin Several clinical trials have been reported and are ongoing to
studies; therefore the higher heart rate and tachycardia evaluate bortezomib in the prevention and treatment of acute
observed with the sotrastaurin in combination with MMF humoral rejection.177 However, mixed results have been
were not unexpected. All tachycardia adverse events were reported in this context.178–180 Everly et al. administered
mild, and the majority occurred soon after transplantation. bortezomib to six patients with antibody-mediated rejec-
In the study of Tedesco-Silva et al., gastrointestinal and tion with concomitant acute cellular rejection refractory
cardiac adverse events were more frequent with sotrastau- to currently available therapies.179 Bortezomib administra-
rin whereas higher treatment discontinuation, deaths, tion resulted in resolution of antibody-mediated rejection in
and graft losses occurred with sotrastaurin 300 mg.167 In all six patients, a decrease of donor-specific antibody levels,
the study of Russ et al., mean heart rates, gastrointestinal and improvement of allograft function.179 In another study,
adverse events, and discontinuation due to adverse events Walsh et al. reported the results of bortezomib treatment in
were more frequent with sotrastaurin, whereas leukopenia 28 patients with antibody-mediated rejection.181 Bortezo-
was more frequent with MPA.168 mib treatment was associated with variable results, with
better responses in early (occurring in the first 6 months
Conclusion after transplantation) versus late (occurring 6 months or
Sotrastaurin blocks PKC-mediated early T cell activation, more after transplantation) antibody-mediated rejection.181
providing a new approach for immunosuppression distinct Although some studies have reported bortezomib-based
320 Kidney Transplantation: Principles and Practice

desensitization induced long-term decrease in donor-specific cells190,191 the control of VDR expression in immune cells
antibodies,182 in the study of Sberro-Soussan et al., bortezo- by immune signals192 and the presence of vitamin D metab-
mib treatment did not result in a decrease of donor-specific olizing enzymes such as CYP27B1 in T and B lymphocytes,
antibody mean fluorescence intensity when used as sole and CYP2R1 in dendritic cells,193,194 which allows for the
desensitization therapy in four renal transplant recipients local conversion of 25(OH)D3 into 1,25(OH)2D3.195
with subacute antibody-mediated rejection with persistent 1,25(OH)2D3 results in more immature or tolero-
donor-specific antibodies.180 As the densitization procotols genic phenotype of dendritic cells,196–198 induction of
also include IV Ig, rituximab, and plasmapheresis, it is impos- CD4+CD25highCD127low regulatory T cells,199–201 and B
sible to attribute the reduction in donor-specific antibodies cell apoptosis.202 A clear additive and even synergistic effect
solely to bortezomib. Moreover, in a report from the Mayo was observed between 1,25(OH)2D3 or its analogs and
Clinic only a modest reduction in HLA antibody titers and no other more classical immunosuppressants such as cyclo-
effect on cPRA or flow crossmatch was observed.183 A recent sporine, sirolimus, or MMF, both in vitro and in several
report in nonhuman primates undergoing fully mismatched in vivo autoimmune disease models, such as autoimmune
allogeneic skin transplant reported that bortezomib treat- diabetes203,204 and experimental autoimmune encepha-
ment was followed by a rebound effect characterized by lomyelitis.205–207 In transplant models, monotherapy,
increased circulating IgG+ B cells, increased germinal center 1,25(OH)2D3 and its analogs provoke only a modest pro-
B cells, and follicular helper T cells in the lymph nodes.184 longation of graft function.196,204,208–216 Human data are
This reaction was termed humoral compensation by the still lacking that can sustain the proposed benefits of vita-
authors and shown to be driven by B cell activating factor of min D supplementation for optimal immune function. In a
TNF family (BAFF).184 These data suggest that future trials randomized trial involving lung transplant recipients, once
should combine bortezomib with agents inhibiting BAFF sig- monthly oral vitamin D supplementation (cholecalciferol;
naling such as belimumab and atacicept. Newer proteasome 100,000 IU) did result in increased levels of 25-hydroxy
inhibitors such as carfilzomib result in irreversible inhibition vitamin D, but clinical outcomes were unaffected (chronic
whereas bortezomib only induces temporal inhibition of the lung allograft dysfunction, overall survival, prevalence of
proteasome.185 Studies are ongoing evaluating carfilzomib acute rejection, lymphocytic bronchiolitis and infection,
in desensitization protocol in patients with preformed HLA lung function, pulmonary and systemic inflammation, and
antibodies (NCT02442648) and in the treatment of acute bone mineral density).217 A major concern remains the side
humoral rejection in lung transplantation (NCT02474927). effects of 1,25(OH)2D3 on calcium and bone metabolism.
Apoptosis plays an important in the immune system as
Toxicity it controls lymphocyte deletion in primary and secondary
In general bortezomib is administered intravenously twice lymphoid organs. Apoptosis also plays a critical role in the
weekly for 1 month at 1.3 mg/m2 per administration. maintenance of immune tolerance. Two pathways of apop-
The most common side effects associated with bortezomib tosis (intrinsic and extrinsic apoptosis pathway) have been
treatment are gastrointestinal toxicity (including paralytic described, and agents that specifically influence the intrin-
ileus), thrombocytopenia, and neuropathy.79,186 These can sic apoptosis pathway have shown promise as immuno-
be potentially severe and disabling. To date, no increase in suppressive agents. The BH3-mimetic ABT-737 has been
rate of opportunistic infections has been reported. Impor- reported to inhibit allogeneic immune responses through a
tantly, although bortezomib is associated with substantial combination of lymphopenia, deletion of alloreactive T cells,
reductions in donor-specific antibody levels, it does not and relative enrichment of regulatory T cells.218,219 In vitro,
result in a decrease in protective antibody levels.187 ABT-737 inhibited allogeneic T cell activation, prolifera-
tion, and cytotoxicity. In vivo, a combination of ABT-737
Conclusion with low-dose CsA resulted in long-term skin graft survival
Proteasome inhibitor therapy has potential in the treat- in a murine major histocompatibility complex I (MHC I)
ment of antibody-mediated rejection in kidney transplant mismatch model.218,220 Furthermore, ABT-737 in combi-
recipients both as primary and rescue therapy. Optimal nation with low-dose CsA and anti-CD154 (costimulation
responses with bortezomib are obtained when antibody- blockade) induced stable mixed chimerism (and tolerance)
mediated rejection occurs early posttransplant and bort- in a MHC-mismatched murine bone marrow transplant
ezomib treatment is initiated promptly. Recent data suggest model.221 Interestingly, ABT-737 also induced apoptosis
that bortezomib should be combined with agents inhibiting in memory T cells and prolonged skin allograft survival in
BAFF signaling such as belimumab and atacicept to prevent sensitized mice.222 Another Bcl-2 inhibitor, AB199, is cur-
rebound humoral immunity. rently being evaluated in a clinical trial concerning sys-
temic lupus erythematosus.
Novel inhibitors of the mTOR pathway have been
OTHERS
reported. mTOR is the kinase subunit of mTOR complex 1
1,25-Dihydroxyvitamin D3 (1,25(OH)2D3) and some of its and mTOR complex 2. Although rapamycin partially inhib-
new synthetic structural analogs were considered prom- its mTOR complex 1, it does not inhibit mTOR complex 2.
ising immunomodulators, in addition to its well-known Dual blockade of mTOR complex 1 and mTOR complex 2
role in mineral and bone homeostasis, nonclassical effects has shown potential to abrogate chronic rejection in a rat
of vitamin D such as immune regulation have increas- cardiac allograft model.223 ATP-competitive inhibitors
ingly been recognized.188,189 A role for 1,25(OH)2D3 in compete with ATP and inhibit both mTOR complexes.224
immune regulation was suggested by the presence of its This new class of mTOR inhibitors has mainly been evalu-
receptor (vitamin D receptor, or VDR) in almost all immune ated for anticancer properties and only AZD8055 has been
20 • Other Forms of Immunosuppression 321

evaluated as an immunosuppressant. AZD8055 is a pow- Brequinar sodium exerts its immunosuppressive effects
erful inhibitor of T cell proliferation in vitro and a 10-day through the inhibition of the enzyme dihydroorotate dehy-
course of AZD8055 induced a prolongation of heart allograft drogenase, resulting in inhibition of both T and B lympho-
survival in a murine MHC-mismatched heart transplant cytes. Although the characteristics of brequinar suggest
model.225 AZD8055 is rapidly metabolized by human that it would be an attractive immunosuppressant, the
hepatocytes (resulting in a short half-life), and therefore, a suboptimal pharmacologic profile jeopardizes its use in
novel compound has been developed with a longer half-life, transplant patients. The future use of this drug in the field
AZD2014. AZD2014 is currently being evaluated in phase of transplantation will require the development of analogs
I and II trials in oncology.226 AZD2014 is rapidly absorbed exhibiting a shorter half-life and reduced toxicity.
after oral intake with a median time to peak of 30 to 60 Spergualin was originally isolated from the culture
minutes. The elimination half-life is approximately 3 hours filtrate of Bacillus laterosporus, and explored as a new
with important interpatient variability. The currently rec- anticancer or antibiotic substance. Its analog 15-deoxys-
ommended dose of AZD2014 is 50 mg twice daily.227 pergualin subsequently became widely known as a prom-
Cladribine is an adenosine deaminase-resistant analog of ising new immunosuppressant. The precise mode of action
deoxyadenosine and is used in the treatment of leukemia and of 15-deoxyspergualin is largely unknown and, because
lymphoma. A number of studies have explored the immu- of its poor oral bioavailability, 15-deoxyspergualin must
nosuppressive capacity of cladribine. In vitro, cladribine be delivered parenterally, which hampers its widespread
inhibits both B and T cell proliferation.228 In vivo, cladribine clinical use.238 Its efficacy has been demonstrated in the
monotherapy was shown to prolong skin allograft survival in treatment of kidney allograft rejection, ABO-incompatible
mice,229 in combination with cyclosporine it prolonged liver kidney transplantation, and transplantation in sensitized
and heart allograft survival in rats,230 and was more effective patients.239–241 Until analogs are developed that allow
than cyclosporine monotherapy in small-bowel allografts.231 oral administration,242 the major clinical indication of
However, no clinical trials are published to date. 15-deoxyspergualin is limited to the treatment of rejection
The farnesyltransferase inhibitor A-228839 was devel- crises where 15-deoxyspergualin may be an interesting
oped as an anticancer compound that inhibits Ras GTPases. alternative to steroids or antilymphocyte agents. The fact
A-228839 inhibited lectin-induced proliferation and that it remains effective after recurrent administration is
antigen-presenting cell-induced T cell proliferation. The promising.
compound also inhibited lymphocyte Th1 cytokine produc- Upon cellular uptake, cyclophosphamide is extensively
tion and promoted apoptosis in lectin-activated lympho- metabolized into its active compounds phosphoramide mus-
cytes.232 Another farnesyltransferase inhibitor, ABT-100 tard and acrolein.243,244 The reaction of the phosphoramide
was shown in vitro to block the secretion of IFN-γ and IL-4 mustard with DNA results in cell death.245 Because of its
by naïve T cells and suppressed alloreactivity. In a rat hetero- limited efficacy and multiple side effects, the only standard
topic cardiac transplant model, ABT-100 alone or in com- indication for cyclophosphamide in transplantation today
bination with subtherapeutic dose of cyclosporine delayed is the desensitization of highly sensitized recipients before
the development of acute rejection.233 Given its combined kidney transplantation.246 Most of these protocols involve
antirejection and antioncogenic effects, farnesyltransferase repeated plasmapheresis, in combination with cyclophos-
inhibitors could represent an attractive new class of immu- phamide, either with or without continuation of steroids,
nosuppressants in malignancy-prone organ transplant until a kidney transplant becomes available.
recipients if future clinical trials confirm their efficacy.
When a T cell receptor recognizes its specific antigen the
lymphoid cell-specific kinase lck is phosphorylated and, Total Lymphoid Irradiation
together with the receptor-associated CD3 complex, phos-
phorylates zinc-associated phosphorylase 70. These events For several decades, total lymphoid irradiation (TLI) has
trigger the downstream cascade that increases intracellular been used for the treatment of Hodgkin’s disease. The pos-
calcium, activating calcineurin. Inhibitors of lck have become sibility of applying TLI as an immunosuppressive regimen
available recently. A-770041 and structurally similar mole- rather than an anticancer treatment was discovered by
cules have been shown to prolong the survival of heterotopic investigators at Stanford University.247
murine heart and renal subcapsular islet grafts as well as
to blunt the production of immunoglobulin IgG2a. Emodin PROCEDURE
(C15H10O5), the cyclic derivative from rhubarb root and rhi-
zomes, has been shown to prolong murine skin graft survival TLI is delivered through two ports. A first so-called man-
and to dampen IL-2 production.234 However, neither of these tle port includes the lymph nodes of the neck, axilla, and
archetypal compounds shows sufficient specificity for lck ver- mediastinum. The other port is called the “inverted Y” and
sus other kinases to warrant clinical development.233 encompasses aortic, iliac, and pelvic lymph nodes and the
FR 252921, an immunosuppressive agent isolated from spleen. Usually, a total dose of 40 to 50 Gy (1 Gy = 100 rad)
the culture of Pseudomonas fluorescens, inhibits AP-1 tran- is administered in daily fractions of 1.5 to 2.5 Gy.
scription activity, and acts predominantly against antigen-
presenting cells. FR 252921 demonstrated synergy with MECHANISMS OF ACTION
tacrolimus in vitro and in vivo. In murine models of skin
transplantation, compared with the optimal dose of tacroli- Much of the currently available experimental evidence on
mus alone, the combination of FR 252921 and tacrolimus the immunologic mechanisms underlying TLI-induced
prolonged graft survival.235–237 tolerance points to the importance of suppressor cells. The
322 Kidney Transplantation: Principles and Practice

group of Strober-identified post-TLI suppressor cells as with cyclosporine and methotrexate in a pig heart-into-
host-type NK T cells, as the protective effect of TLI against baboon model, resulted in a graft survival time of more
graft-versus-host disease was abrogated in mice with a than 2 weeks. This regimen was able to inhibit xenoreac-
CD1d-inactivated gene.248 These host-type NK T cells pro- tive natural antibody production but not the xenoreactivity
duced IL-4 and stimulated donor-type cells to produce IL-4 of macrophages. In a pig islet-into-rat xenograft model, TLI
also.248,249 Definitive evidence of the functional importance in combination with deoxyspergualin was extremely effec-
and activity of these suppressor cells was delivered by the tive,272 and even in a discordant lamb-into-pig model, TLI
demonstration that they could prevent graft-versus-host synergized with cyclosporine and azathioprine to provoke
disease in vivo.250 Post-TLI attenuation of effector T-lym- a 30-fold increase of the mean xenograft survival time.273
phocyte reactivity was equally proposed to be responsible The principal disadvantage for the clinical application of
for the observed immunosuppressed state after TLI.251–253 TLI is that the complete regimen of fractionated daily irra-
This intrinsic T cell defect was dependent on the irradiation diations needs to be administered and completed before,
of both thymus and extrathymic tissues.254 After TLI, aner- and sufficiently close to, the moment of transplantation,
gized T cells were shown to be incapable of proliferating and finding a suitable donor organ within such a restricted
even in the presence of exogenous IL-2.255 In other studies, time frame is problematic. Investigators have therefore
TLI was shown to lead to thymic clonal deletion of donor- explored the possibility of using TLI after transplantation.
or host-reactive lymphocytes.256 TLI-treated mice also In mouse and rat heart allograft models, posttransplant
exhibited decreased antidonor cytotoxic T cell precursor TLI significantly prolonged graft survival when combined
frequencies.257 Finally, Strober’s group showed that Th2 with monoclonal anti-CD4 antibodies274 or with infusion of
lymphocytes recover soon after TLI, whereas Th1 lympho- donor-type dendritic cell precursors.275
cytes remain deficient for several months,10 and showed
that this defect can also be prevented by thymic shielding CLINICAL EXPERIENCE
during irradiation.251 This Th2 dominance after TLI has
been confirmed by other groups in rodents258 and in large The first clinical kidney transplants utilizing TLI were per-
animals.259 Recently, Nador et al. demonstrated that tol- formed at the University of Minnesota in 20 patients who
erance induction after conditioning with TLI and antithy- had previously rejected a renal allograft.276 Because simi-
mocyte globulin (ATG) depends on the ability of naturally lar results (an increase of about 30% 1-year graft survival
occurring regulatory NK T cells and regulatory T cells to compared with historical control data) were achieved in
suppress the residual alloreactive T cells that are capable of this patient population using cyclosporine, and because of
rejecting the allograft.260 the ease of administration, the investigators concluded to
prefer cyclosporine over TLI.
In the 1980s, a controlled trial was performed at the Uni-
EXPERIMENTAL EXPERIENCE
versity of Leuven, in which end-stage diabetic nephropathy
TLI-treated BALB/c mice receiving fully allogeneic C57BL6 patients received cadaveric kidney allografts, investigating
bone marrow and skin graft on the first day after TLI became the effect of pretransplant TLI (20 daily fractions of 1 Gy,
stable hematopoietic chimeras without signs of graft-versus- followed by 1 weekly TLI dose until a suitable donor was
host disease, and developed permanent donor-specific tol- found), followed by low-dose posttransplant prednisone
erance with preserved anti-third-party reactivity.261 Toler- maintenance treatment.277 Long-term (8-year) follow-up
ance induction was critically dependent on the width of the revealed that rejection episodes were more frequent, and
irradiation field, the time of transplantation after TLI, the patient and graft survival were significantly inferior in
total dose of TLI, and the absence of presensitization.261–263 the TLI-treated group.278 The excess mortality in the TLI-
Although bone marrow chimerism could easily be treated patients was due to sepsis, resulting from high-dose
induced, tolerance to either heart264 or kidney allografts265 steroid therapy needed to treat rejection crises. This clinical
was not obtained, suggesting that TLI-induced bone mar- experience confirmed the animal data that also showed that
row chimerism does not necessarily create tolerance toward TLI by itself is insufficient to provoke long-term graft sur-
organ-specific antigens. vival or tolerance and that extra manipulations are needed.
The combination of TLI and low-dose cyclosporine was In a study at Stanford University, 24 patients received
found to be effective and clinically safe in rats,266 and TLI a first, and one patient a second, cadaveric renal allograft
with postoperative ATG-induced permanent and specific using TLI and ATG.279 The actuarial graft survival was
transplantation tolerance toward heart allografts in about 76% and 68% at 1 and 2 years, respectively. Ten of the 25
40% of transplanted dogs.267 These encouraging results patients never had a rejection crisis despite an overall poor
led to a similar trial in clinical kidney transplantation. HLA-matching between donor and recipient. In follow-
Myburgh et al. applied a modified TLI regimen in baboons, up studies, a specific antidonor mixed lymphocyte culture
with low dosage and wide-field exposure, and showed that hypo- or nonresponsiveness was demonstrated280 and, in
tolerance can be achieved in larger animals without con- some patients, all immunosuppressive drugs could be with-
comitant bone marrow transplantation.268 drawn.281 An evaluation in a larger group of 52 patients
Also, in heart or heart–lung transplantation experiments treated with the same protocol at the same center showed
between xenogeneic nonhuman primate species, preop- a 3-year graft survival of about 50%, which is less than in
erative TLI, when administered in combination with cyclo- cyclosporine-treated patients (around 75%).279
sporine and ATG,269 cyclosporine and splenectomy,270 or Posttransplant TLI in combination with anti-CD3 mono-
cyclosporine and medrol,271 was more efficient than any clonal antibodies, or with ATG and donor-specific blood
other treatment regimen. Pretransplant TLI, combined transfusions, seemed very effective in a rat heart allograft
20 • Other Forms of Immunosuppression 323

model. On the basis of these results, the efficacy of TLI number of patients included in these studies is limited, and
was evaluated in heart transplant patients with therapy- prospective randomized trials are needed. The safety and
resistant or early vascular rejection.282–284 This resulted efficacy of photopheresis in the prevention of acute rejec-
in a significant reduction of rejection recurrences, an effect tion of cardiac allografts have been evaluated in primary
which was maintained for at least 2 years. In the mean- cardiac allograft recipients, randomly assigned to standard
time, these favorable results have been confirmed by sev- triple-drug immunosuppressive therapy (cyclosporine, aza-
eral other groups. Also, TLI-treated patients develop less thioprine, and prednisone) alone or in conjunction with
coronary atherosclerosis than matched controls despite 24 photopheresis sessions performed during the first 6
multiple rejection episodes.285–289 months after transplantation. After 6 months of follow-up,
Scandling et al. have reported the use of TLI to induce photopheresis-treated patients developed significantly
tolerance in the setting of combined kidney/hematopoietic fewer multiple rejections, and there were no significant dif-
stem cell transplantation between HLA-matched donor/ ferences in the rates or types of infection. Although there
recipient pairs.290,291 Patients received a conditioning was no significant effect on graft survival rates at 6 or 12
regimen of 10 doses of TLI (80–120 cGy), five doses of rab- months, this study indicated that photopheresis may be an
bit ATG, MMF for 1 month, and cyclosporine for at least effective new immunosuppressive regimen in transplant
6 months. Donor hematopoietic stem cells were injected recipients.310 In patients with refractory bronchiolitis oblit-
intravenously on day 11 in the outpatient infusion cen- erans after lung transplantation, photopheresis resulted
ter.291 In a recent study, Scandling et al. reported on 38 in a stabilization of graft function and/or in some of these
patients undergoing HLA matched or mismatched com- patients in histologic reversal of rejection.311,312
bined kidney/hematopoietic transplantation.292 Sixteen
out of 22 matched patients had persistent chimerism for at
least 6 months and were successfully withdrawn from all Splenectomy
immunosuppression. Whether immunosuppression can be
withdrawn in mismatched patients with mixed chimerism Pretransplant splenectomy in the recipient before transplan-
remains to be established at this time.292 tation was first proposed by Starzl et al. in 1963 as a means
of improving graft survival.313 Although splenectomy is a
standard procedure for patients who develop hypersplen-
CONCLUSION
ism or azathioprine-associated leukopenia, evidence on
TLI has been shown to be a safe immunosuppressive regi- the role of splenectomy in enhancing graft survival is con-
men. It has been abandoned in clinical practice for organiza- troversial.313–316 A large prospective randomized trial in
tional reasons, except for the treatment of therapy-resistant Minneapolis showed splenectomy improved graft survival
rejection of heart or heart–lung transplant. However, its significantly,317 but longer-term follow-up showed loss of
ability to induce tolerance in HLA-matched patients, in beneficial effects because of an increased infection-related
combination with ATG and hematopoietic stem cell trans- mortality.318 Several other single-center studies have
plantation, might renew interest in this treatment modal- shown an alarming risk of sepsis and death, nullifying any
ity. To date, no evidence of radiation-related late effects has early benefits of splenectomy on graft survival,319,320 and
been documented with TLI.293 a multicenter analysis from the South-Eastern Organ Pro-
curement Foundation confirmed a modest improvement in
graft survival after splenectomy, but a relentless increase in
Photopheresis patient mortality.321
Splenectomy has a place in the preparation of a recipient
Extracorporeal photopheresis is a technique in which leuko- who is to receive an ABO-incompatible graft, a practice that
cytes, removed from patients by leukopheresis, are exposed is likely to become more widely employed in living related
to 8-methoxypsoralen and ultraviolet A light. It was devel- donor transplantation, where an ABO-incompatible but
oped as an immunoregulatory treatment for erythrodermic otherwise suitable donor is the only available donor. Alex-
cutaneous T cell lymphoma.294 Subsequently, the proce- andre et al. reported a series of 38 such ABO-incompatible
dure was shown to be safe as an alternative treatment for living donor transplants in which the recipient was prepared
various human immune and autoimmune diseases.295 by plasmapheresis, donor-specific platelet transfusion, and
Furthermore, in rats296 and monkeys,297 the regimen splenectomy.322–324 Although the authors believe that the
was shown to result in extended skin allograft and cardiac need for plasmapheresis and donor-specific platelet transfu-
allograft and xenograft survival. sion should be reevaluated, splenectomy was thought to be
Different mechanisms have been shown to contribute to important, because 3 of 38 recipients who did not have a
the immunomodulatory effect of photopheresis: selective splenectomy lost their grafts from acute vascular rejection,
inhibition of effector cells,296,298 induction of a high rate of in contrast to only 5 of 33 who did undergo splenectomy. A
apoptosis,299 increased capacity to phagocytose apoptotic T small-scale but successful experience with postsplenectomy
cells, resulting in the induction of anticlonotypic immune ABO-incompatible living donor kidney transplantation has
responses,300 shift toward Th2 immune activation,301 and also been reported by Ishikawa et al. in Japan.325 In the set-
induction of regulatory CD4 and CD8 cells.302,303 ting of ABO-incompatible kidney transplantation, antigen-
In clinical transplantation, photopheresis has been specific immunoadsorption, rituximab, and bortezomib
applied as both a therapeutic and prophylactic option. It has treatment have been developed as alternatives to plasma-
been applied in the treatment of recurrent or resistant acute pheresis and splenectomy. This will further reduce the indi-
rejection in renal transplant patients,301,304–309 but the cations for splenectomy in organ transplantation.326,327
324 Kidney Transplantation: Principles and Practice

Splenectomy has also been applied in combination with treatment of antibody-mediated acute renal allograft
eculizumab as salvage therapy in patients developing rejection by combining plasmapheresis with 15-deoxys-
severe antibody-mediated rejection after HLA-incompatible pergualin.343 Antibody-mediated rejection is increasingly
kidney transplantation.328 In a follow-up report, splenec- being recognized as a determinant of short-term and long-
tomy was substituted for splenic irradiation.329 term allograft outcome.344 The optimal treatment of anti-
body-mediated rejection is undetermined at this moment,
and possible therapeutic approaches include combinations
Plasmapheresis of plasmapheresis exchange, IV IG, and anti-CD20 anti-
body to remove donor-specific antibodies and inhibit anti-
Plasmapheresis is increasingly used to facilitate kidney body production. However, evidence on safety and efficacy
transplantation in patients with high levels of anti-HLA is weak, and the optimal treatment protocol has yet to be
antibodies or ABO incompatibility.330–332 Furthermore, determined.345
plasmapheresis has been used in the treatment of antibody- Immunoabsorption has been applied as an alternative
mediated allograft rejection.333 Plasmapheresis is a compo- to plasmapheresis and was found to be an equally efficient
nent of several desensitization protocols applied in patients method.326,346 Studies of this approach in highly sensitized
with living donors and an incompatible crossmatch because candidate transplant recipients are continuing.
of donor-specific antibodies. Different combinations of plas-
mapheresis with rituximab, corticosteroids, IV IG, and bort- References
ezomib have been reported.334 Plasmapheresis should be 1. Bartlett RR, Dimitrijevic M, Mattar T, et al. Leflunomide (HWA 486),
performed until crossmatch testing becomes negative typi- a novel immunomodulating compound for the treatment of autoim-
cally on a daily basis or on alternate days, and the number mune disorders and reactions leading to transplantation rejection.
of plasmapheresis sessions is dependent on the antibody Agents Actions 1991;32:10–21.
levels and degree of mismatch. Transplantation should 2. Jin MB, Nakayama M, Ogata T, et al. A novel leflunomide deriva-
tive, FK778, for immunosuppression after kidney transplantation in
be performed within days of the last desensitization before dogs. Surgery 2002;132:72–9.
rebound of anti-HLA antibody titers occurs. Encouraging 3. Chong AS, Gebel H, Finnegan A, et al. Leflunomide, a novel immu-
early results of this approach have been reported, although nomodulatory agent: in vitro analyses of the mechanism of immu-
they were associated with considerable morbidity.335–337 nosuppression. Transplant Proc 1993;25:747–9.
4. Siemasko K, Chong AS, Jack HM, Gong H, Williams JW, Finnegan
Recent studies provided evidence of an important survival A. Inhibition of JAK3 and STAT6 tyrosine phosphorylation by the
benefit for patients undergoing HLA-incompatible kidney immunosuppressive drug leflunomide leads to a block in IgG1 pro-
transplantation after desensitization.330,331 In an analy- duction. J Immunol 1998;160:1581–8.
sis by Segev and colleagues, 1025 HLA-sensitized patients 5. Williamson RA, Yea CM, Robson PA, et al. Dihydroorotate dehydro-
from 22 centers undergoing desensitization before kidney genase is a high affinity binding protein for A77 1726 and mediator
of a range of biological effects of the immunomodulatory compound.
transplant from a living donor between 1997 and 2011 J Biol Chem 1995;270:22467–72.
were included. Different desensitization protocols were 6. Mattar T, Kochhar K, Bartlett R, Bremer EG, Finnegan A. Inhibition
applied in this study. Compared with patients on the wait- of the epidermal growth factor receptor tyrosine kinase activity by
ing list or receiving a deceased donor transplant or patients leflunomide. FEBS Lett 1993;334:161–4.
7. Elder RT, Xu X, Williams JW, Gong H, Finnegan A, Chong AS. The
on the waiting list not receiving a transplant, the 8-year immunosuppressive metabolite of leflunomide, A77 1726, affects
survival rates of HLA-sensitized recipients was 13.6 and murine T cells through two biochemical mechanisms. J Immunol
32.6 percentage points higher, respectively. Also the risk of 1997;159:22–7.
death was significantly reduced after transplantation from 8. Mahajan S, Ghosh S, Sudbeck EA, et al. Rational design and synthe-
an incompatible live donor after desensitization treatment. sis of a novel anti-leukemic agent targeting Bruton’s tyrosine kinase
(BTK), LFM-A13 [alpha-cyano-beta-hydroxy-beta-methyl-N-(2,
The most important adverse events associated with desensi- 5-dibromophenyl)propenamide]. J Biol Chem 1999;274:9587–99.
tization were anaphylaxis, hypotension, and infections.331 9. Chong AS, Huang W, Liu Wet al. In vivo activity of leflunomide:
Current ABO-incompatible transplantation protocols pharmacokinetic analyses and mechanism of immunosuppression.
also include plasmapheresis.322–324 Brynger et al. have Transplantation 1999;68:100–9.
10. Bass H, Mosmann T, Strober S. Evidence for mouse Th1- and Th2-
reported some successful ABO-incompatible grafts without like helper T cells in vivo. Selective reduction of Th1-like cells after
prior plasmapheresis of the recipient,338 and it was origi- total lymphoid irradiation. J Exp Med 1989;170:1495–511.
nally felt that splenectomy was a prerequisite for success- 11. Karaman A, Fadillioglu E, Turkmen E, Tas E, Yilmaz Z. Protective
ful ABO-incompatible transplantation. However, it has effects of leflunomide against ischemia-reperfusion injury of the rat
been demonstrated that combining plasmapheresis and IV liver. Pediatr Surg Int 2006;22:428–34.
12. Manna SK, Mukhopadhyay A, Aggarwal BB. Leflunomide sup-
IG (without splenectomy) allows ABO-incompatible renal presses TNF-induced cellular responses: effects on NF-kappa B,
transplantation. Plasmapheresis aims to reduce ABO anti- activator protein-1, c-Jun N-terminal protein kinase, and apoptosis.
body titers below center-specific critical thresholds before J Immunol 2000;165:5962–9.
transplantation, and plasmapheresis might be combined 13. Jarman ER, Kuba A, Montermann E, Bartlett RR, Reske-Kunz AB.
Inhibition of murine IgE and immediate cutaneous hypersensitivity
with rituximab.339 Excellent outcomes have been reported responses to ovalbumin by the immunomodulatory agent lefluno-
in patients undergoing ABO-incompatible living kidney mide. Clin Exp Immunol 1999;115:221–8.
transplantation after desensitization with 5- and 10-year 14. Manna SK, Aggarwal BB. Immunosuppressive leflunomide metabo-
allograft survival of 95% and 90%, respectively.334,340,341 lite (A77 1726) blocks TNF-dependent nuclear factor-kappa B acti-
Plasmapheresis is also a component of the treatment of vation and gene expression. J Immunol 1999;162:2095–102.
15. Bilolo KK, Ouyang J, Wang X, et al. Synergistic effects of malononi-
antibody-mediated allograft rejection. Although some ini- trilamides (FK778, FK779) with tacrolimus (FK506) in prevention
tial reports suggested a beneficial effect,342 controlled trials of acute heart and kidney allograft rejection and reversal of ongoing
were unconvincing. Nojima et al. reported the successful heart allograft rejection in the rat. Transplantation 2003;75:1881–7.
20 • Other Forms of Immunosuppression 325

16. Deuse T, Schrepfer S, Reichenspurner H. Immunosuppression with 39. Faguer S, Hirsch HH, Kamar N, et al. Leflunomide treatment for
FK778 and mycophenolate mofetil in a rat cardiac transplantation polyomavirus BK-associated nephropathy after kidney transplanta-
model. Transplantation 2003;76:1627–9. tion. Transpl Int 2007;20:962–9.
17. Evers DL, Wang X, Huong SM, Huang DY, Huang ES. 3,4’,5- 40. Egli A, Kohli S, Dickenmann M, Hirsch HH. Inhibition of polyoma-
Trihydroxy-trans-stilbene (resveratrol) inhibits human cytomegalo- virus BK-specific T-Cell responses by immunosuppressive drugs.
virus replication and virus-induced cellular signaling. Antiviral Res Transplantation 2009;88:1161–8.
2004;63:85–95. 41. Johnston O, Jaswal D, Gill JS, Doucette S, Fergusson DA, Knoll GA.
18. Knight DA, Hejmanowski AQ, Dierksheide JE, Williams JW, Chong Treatment of polyomavirus infection in kidney transplant recipients:
AS, Waldman WJ. Inhibition of herpes simplex virus type 1 by the a systematic review. Transplantation 2010;89:1057–70.
experimental immunosuppressive agent leflunomide. Transplanta- 42. Topalis D, Lebeau I, Krecmerova M, Andrei G, Snoeck R. Activities
tion 2001;71:170–4. of different classes of acyclic nucleoside phosphonates against BK
19. Waldman WJ, Knight DA, Lurain NS, et al. Novel mechanism of virus in primary human renal cells. Antimicrob Agents Chemother
inhibition of cytomegalovirus by the experimental immunosuppres- 2011;55:1961–7.
sive agent leflunomide. Transplantation 1999;68:814–25. 43. Liacini A, Seamone ME, Muruve DA, Tibbles LA. Anti-BK virus
20. Farasati NA, Shapiro R, Vats A, Randhawa P. Effect of leflunomide mechanisms of sirolimus and leflunomide alone and in combina-
and cidofovir on replication of BK virus in an in vitro culture system. tion: toward a new therapy for BK virus infection. Transplantation
Transplantation 2005;79:116–8. 2010;90:1450–7.
21. Chong AS, Zeng H, Knight DA, et al. Concurrent antiviral and 44. Zaman RA, Ettenger RB, Cheam H, Malekzadeh MH, Tsai EW.
immunosuppressive activities of leflunomide in vivo. Am J Trans- A novel treatment regimen for BK viremia. Transplantation
plant 2006;6:69–75. 2014;97:1166–71.
22. Zeng H, Waldman WJ, Yin DP, et al. Mechanistic study of malono- 45. Jaw J, Hill P, Goodman D. Combination of leflunomide and everoli-
nitrileamide FK778 in cardiac transplantation and CMV infection in mus for treatment of BK virus nephropathy. Nephrology (Carlton).
rats. Transplantation 2005;79:17–22. 2017;22:326–9.
23. Deuse T, Schrepfer S, Schafer H, et al. FK778 attenuates 46. Ciszek M, Mucha K, Foroncewicz B, Chmura A, Paczek L. Lefluno-
lymphocyte-endothelium interaction after cardiac transplantation: mide as a rescue treatment in ganciclovir-resistant cytomegalovirus
in vivo and in vitro studies. Transplantation 2004;78:71–7. infection in a seronegative renal transplant recipient—a case report.
24. Savikko J, Von WE, Hayry P. Leflunomide analogue FK778 is vascu- Ann Transplant 2014;19:60–3.
loprotective independent of its immunosuppressive effect: potential 47. El Chaer F, Mori N, Shah D, et al. Adjuvant and salvage therapy with
applications for restenosis and chronic rejection. Transplantation leflunomide for recalcitrant cytomegalovirus infections in hemato-
2003;76:455–8. poietic cell transplantation recipients: a case series. Antiviral Res
25. Lin Y, Vandeputte M, Waer M. A short-term combination therapy 2016;135:91–6.
with cyclosporine and rapamycin or leflunomide induces long-term 48. Goldsmith PM, Husain MM, Carmichael A, Zhang H, Middleton SJ.
heart allograft survival in a strongly immunogenic strain combina- Case report: multidrug-resistant cytomegalovirus in a modified mul-
tion in rats. Transpl Int 1996;9(Suppl. 1):S328–30. tivisceral transplant recipient. Transplantation 2012;93:e30–2.
26. Sun Y, Chen X, Zhao J, et al. Combined use of rapamycin and leflu- 49. Miszewska-Szyszkowska D, Mikolajczyk N, Komuda-Leszek E,
nomide in prevention of acute cardiac allografts rejection in rats. et al. Severe cytomegalovirus infection in a second kidney trans-
Transpl Immunol 2012;27:19–24. plant recipient treated with ganciclovir, leflunomide, and immu-
27. Williams JW, Xiao F, Foster P, et al. Leflunomide in experimental noglobulins, with complications including seizures, acute HCV
transplantation. Control of rejection and alloantibody production, infection, drug-induced pancytopenia, diabetes, cholangitis, and
reversal of acute rejection, and interaction with cyclosporine. Trans- multi-organ failure with fatal outcome: a case report. Ann Trans-
plantation 1994;57:1223–31. plant 2015;20:169–74.
28. Xiao F, Shen J, Chong A, et al. Control and reversal of chronic xeno- 50. Morita S, Shinoda K, Tamaki S, et al. Successful low-dose lefluno-
graft rejection in hamster-to-rat cardiac transplantation. Transplant mide treatment for ganciclovir-resistant cytomegalovirus infection
Proc 1996;28:691–2. with high-level antigenemia in a kidney transplant: a case report
29. Lin Y, Goebels J, Xia G, Ji P, Vandeputte M, Waer M. Induction of and literature review. J Clin Virol 2016;82:133–8.
specific transplantation tolerance across xenogeneic barriers in the 51. Nguyen L, McClellan RB, Chaudhuri A, et al. Conversion from tacro-
T-independent immune compartment. Nat Med 1998;4:173–80. limus/mycophenolic acid to tacrolimus/leflunomide to treat cuta-
30. Chiba K, Yanagawa Y, Masubuchi Y, et al. FTY720, a novel neous warts in a series of four pediatric renal allograft recipients.
immunosuppressant, induces sequestration of circulating mature Transplantation 2012;94:450–5.
lymphocytes by acceleration of lymphocyte homing in rats. I. 52. Basu G, Mohapatra A, Manipadam MT, Mani SE, John GT. Leflu-
FTY720 selectively decreases the number of circulating mature nomide with low-dose everolimus for treatment of Kaposi’s sar-
lymphocytes by acceleration of lymphocyte homing. J Immunol coma in a renal allograft recipient. Nephrol Dial Transplant
1998;160:5037–44. 2011;26:3412–5.
31. Pan F, Ebbs A, Wynn C, et al. FK778, a powerful new immuno- 53. Guasch A, Roy-Chaudhury P, Woodle ES, Fitzsimmons W, Holman J,
suppressant, effectively reduces functional and histologic changes First MR. Assessment of efficacy and safety of FK778 in comparison
of chronic rejection in rat renal allografts. Transplantation with standard care in renal transplant recipients with untreated BK
2003;75:1110–4. nephropathy. Transplantation 2010;90:891–7.
32. Kyles AE, Gregory CR, Griffey SM, et al. Immunosuppression with a 54. Leca N. Leflunomide use in renal transplantation. Curr Opin Organ
combination of the leflunomide analog, FK778, and microemulsified Transplant 2009;14:370–4.
cyclosporine for renal transplantation in mongrel dogs. Transplan- 55. Vanrenterghem Y, van Hooff JP, Klinger M, et al. The effects of FK778
tation 2003;75:1128–33. in combination with tacrolimus and steroids: a phase II multicenter
33. Qi S, Zhu S, Xu D, et al. Significant prolongation of renal allograft study in renal transplant patients. Transplantation 2004;78:9–14.
survival by delayed combination therapy of FK778 with tacrolimus 56. Smolen JS, Kalden JR, Scott DL, et al. Efficacy and safety of leflu-
in nonhuman primates. Transplantation 2003;75:1124–8. nomide compared with placebo and sulphasalazine in active rheu-
34. Hirsch HH, Randhawa P. BK polyomavirus in solid organ transplan- matoid arthritis: a double-blind, randomised, multicentre trial.
tation. Am J Transplant 2013;13:179–88. European Leflunomide Study Group. Lancet 1999;353:259–66.
35. Josephson MA, Gillen D, Javaid B, et al. Treatment of renal allograft 57. Chikura B, Lane S, Dawson JK. Clinical expression of leflunomide-
polyoma BK virus infection with leflunomide. Transplantation induced pneumonitis. Rheumatology (Oxford) 2009;48:1065–8.
2006;81:704–10. 58. Kho LK, Kermode AG. Leflunomide-induced peripheral neuropathy.
36. Krisl JC, Taber DJ, Pilch N, et al. Leflunomide efficacy and pharma- J Clin Neurosci 2007;14:179–81.
codynamics for the treatment of BK viral infection. Clin J Am Soc 59. Ostensen M. Disease specific problems related to drug therapy in
Nephrol 2012;7:1003–9. pregnancy. Lupus 2004;13:746–50.
37. Kuypers DR. Management of polyomavirus-associated nephropathy 60. Curtis JR, Beukelman T, Onofrei A, et al. Elevated liver enzyme
in renal transplant recipients. Nat Rev Nephrol 2012;8:390–402. tests among patients with rheumatoid arthritis or psoriatic arthri-
38. Williams JW, Javaid B, Kadambi PV, et al. Leflunomide for polyoma- tis treated with methotrexate and/or leflunomide. Ann Rheum Dis
virus type BK nephropathy. N Engl J Med 2005;352:1157–8. 2010;69:43–7.
326 Kidney Transplantation: Principles and Practice

61. Suissa S, Ernst P, Hudson M, Bitton A, Kezouh A. Newer disease- 83. Xu M, Pirenne J, Antoniou EA, Afford SC, D’Silva M, McMaster P.
modifying antirheumatic drugs and the risk of serious hepatic Effect of peritransplant FTY720 alone or in combination with post-
adverse events in patients with rheumatoid arthritis. Am J Med transplant tacrolimus in a rat model of cardiac allotransplantation.
2004;117:87–92. Transpl Int 1998;11:288–94.
62. Fujita T, Inoue K, Yamamoto S, et al. Fungal metabolites. Part 11. A 84. Suzuki S, Enosawa S, Kakefuda T, Amemiya H, Hoshino Y, Chiba
potent immunosuppressive activity found in Isaria sinclairii metabo- K. Long-term graft acceptance in allografted rats and dogs by treat-
lite. J Antibiot (Tokyo) 1994;47:208–15. ment with a novel immunosuppressant, FTY720. Transplant Proc
63. Fujita T, Inoue K, Yamamoto S, et al. Fungal metabolites. Part 12. 1996;28:1375–6.
Potent immunosuppressant, 14-deoxomyriocin, (2S,3R,4R)-(E)- 85. Suzuki S, Enosawa S, Kakefuda T, et al. A novel immunosuppressant,
2-amino-3,4-dihydroxy-2-hydroxymethyleicos-6-enoic acid and FTY720, with a unique mechanism of action, induces long-term
structure-activity relationships of myriocin derivatives. J Antibiot graft acceptance in rat and dog allotransplantation. Transplantation
(Tokyo) 1994;47:216–24. 1996;61:200–5.
64. Sasaki S, Hashimoto R, Kiuchi M, et al. Fungal metabolites. Part 86. Xu M, Pirenne J, Antoniou S, Gunson B, D’Silva M, McMaster P.
14. Novel potent immunosuppressants, mycestericins, produced by FTY720 compares with FK 506 as rescue therapy in rat heterotopic
Mycelia sterilia. J Antibiot (Tokyo) 1994;47:420–33. cardiac transplantation. Transplant Proc 1998;30:2221–2.
65. Halin C, Scimone ML, Bonasio R, et al. The S1P-analog FTY720 dif- 87. Mitsusada M, Suzuki S, Kobayashi E, Enosawa S, Kakefuda T, Miyata
ferentially modulates T-cell homing via HEV: T-cell-expressed S1P1 M. Prevention of graft rejection and graft-versus-host reaction by a
amplifies integrin activation in peripheral lymph nodes but not in novel immunosuppressant, FTY720, in rat small bowel transplanta-
Peyer patches. Blood 2005;106:1314–22. tion. Transpl Int 1997;10:343–9.
66. Mandala S, Hajdu R, Bergstrom J, et al. Alteration of lymphocyte 88. Delbridge MS, Shrestha BM, Raftery AT, El Nahas AM, Haylor
trafficking by sphingosine-1-phosphate receptor agonists. Science JL. Reduction of ischemia-reperfusion injury in the rat kidney by
2002;296:346–9. FTY720, a synthetic derivative of sphingosine. Transplantation
67. Matloubian M, Lo CG, Cinamon G, et al. Lymphocyte egress from 2007;84:187–95.
thymus and peripheral lymphoid organs is dependent on S1P recep- 89. Fuller TF, Hoff U, Kong L, et al. Cytoprotective actions of FTY720
tor 1. Nature 2004;427:355–60. modulate severe preservation reperfusion injury in rat renal trans-
68. Pappu R, Schwab SR, Cornelissen I, et al. Promotion of lymphocyte plants. Transplantation 2010;89:402–8.
egress into blood and lymph by distinct sources of sphingosine- 90. Man K, Ng KT, Lee TK, et al. FTY720 attenuates hepatic ischemia-
1-phosphate. Science 2007;316:295–8. reperfusion injury in normal and cirrhotic livers. Am J Transplant
69. Yuzawa K, Stephkowski SM, Wang M, Kahan BD. FTY720 blocks 2005;5:40–9.
allograft rejection by homing of lymphocytes in vivo. Transplant 91. Suleiman M, Cury PM, Pestana JO, Burdmann EA, Bueno V. FTY720
Proc 2000;32:269. prevents renal T-cell infiltration after ischemia/reperfusion injury.
70. Habicht A, Clarkson MR, Yang J, et al. Novel insights into the Transplant Proc 2005;37:373–4.
mechanism of action of FTY720 in a transgenic model of allograft 92. Hoshino Y, Suzuki C, Ohtsuki M, Masubuchi Y, Amano Y, Chiba K.
rejection: implications for therapy of chronic rejection. J Immunol FTY720, a novel immunosuppressant possessing unique mecha-
2006;176:36–42. nisms. II. Long-term graft survival induction in rat heterotopic
71. Zhang Q, Chen Y, Fairchild RL, Heeger PS, Valujskikh A. Lymphoid cardiac allografts and synergistic effect in combination with cyclo-
sequestration of alloreactive memory CD4 T cells promotes cardiac sporine A. Transplant Proc 1996;28:1060–1.
allograft survival. J Immunol 2006;176:770–7. 93. Suzuki T, Jin MB, Shimamura T, et al. A new immunosuppressant,
72. Li XK, Enosawa S, Kakefuda T, Amemiya H, Suzuki S. FTY720, a FTY720, in canine kidney transplantation: effect of single-drug,
novel immunosuppressive agent, enhances upregulation of the cell induction and combination treatments. Transpl Int 2004;17:574–
adhesion molecular ICAM-1 in TNF-alpha treated human umbilical 84.
vein endothelial cells. Transplant Proc 1997;29:1265–6. 94. Wang ME, Tejpal N, Qu X, Yu J, Okamoto M, Stepkowski SM, et al.
73. Sawicka E, Dubois G, Jarai G, et al. The sphingosine 1-phosphate Immunosuppressive effects of FTY720 alone or in combination with
receptor agonist FTY720 differentially affects the sequestration of cyclosporine and/or sirolimus. Transplantation 1998;65:899–905.
CD4+/CD25+ T-regulatory cells and enhances their functional 95. Stone ML, Sharma AK, Zhao Y, et al. Sphingosine-1-phosphate
activity. J Immunol 2005;175:7973–80. receptor 1 agonism attenuates lung ischemia-reperfusion injury.
74. Zhou PJ, Wang H, Shi GH, Wang XH, Shen ZJ, Xu D. Immunomod- Am J Physiol Lung Cell Mol Physiol 2015;308:L1245–52.
ulatory drug FTY720 induces regulatory CD4(+)CD25(+) T cells 96. Khiew SH, Yang J, Young JS, Chen J, Wang Q, Yin D, et al. CTLA4-Ig
in vitro. Clin Exp Immunol 2009;157:40–7. in combination with FTY720 promotes allograft survival in sensi-
75. Troncoso P, Stepkowski SM, Wang ME, et al. Prophylaxis of acute tized recipients. JCI. Insight 2017;2:92033.
renal allograft rejection using FTY720 in combination with subther- 97. Fujishiro J, Kudou S, Iwai S, et al. Use of sphingosine-1-phosphate
apeutic doses of cyclosporine. Transplantation 1999;67:145–51. 1 receptor agonist, KRP-203, in combination with a subtherapeutic
76. Masubuchi Y, Kawaguchi T, Ohtsuki M, et al. FTY720, a novel dose of cyclosporine A for rat renal transplantation. Transplantation
immunosuppressant, possessing unique mechanisms. IV. Pre- 2006;82:804–12.
vention of graft versus host reactions in rats. Transplant Proc 98. Shimizu H, Takahashi M, Kaneko T, et al. KRP-203, a novel syn-
1996;28:1064–5. thetic immunosuppressant, prolongs graft survival and attenu-
77. Lan YY, De CA, Colvin BL, et al. The sphingosine-1-phosphate recep- ates chronic rejection in rat skin and heart allografts. Circulation
tor agonist FTY720 modulates dendritic cell trafficking in vivo. Am J 2005;111:222–9.
Transplant 2005;5:2649–59. 99. Suzuki C, Takahashi M, Morimoto H, et al. Efficacy of mycophe-
78. Yin N, Zhang N, Xu J, Shi Q, Ding Y, Bromberg JS. Targeting lym- nolic acid combined with KRP-203, a novel immunomodulator,
phangiogenesis after islet transplantation prolongs islet allograft in a rat heart transplantation model. J Heart Lung Transplant
survival. Transplantation 2011;92:25–30. 2006;25:302–9.
79. Suzuki S, Kakefuda T, Amemiya H, et al. An immunosuppressive 100. Khattar M, Deng R, Kahan BD, et al. Novel sphingosine-1-
regimen using FTY720 combined with cyclosporin in canine kidney phosphate receptor modulator KRP203 combined with locally deliv-
transplantation. Transpl Int 1998;11:95–101. ered regulatory T cells induces permanent acceptance of pancreatic
80. Yamashita K, Nomura M, Omura T, et al. Effect of a novel immu- islet allografts. Transplantation 2013;95:919–27.
nosuppressant, FTY720, on heart and liver transplantations in rats. 101. Dun H, Song L, Ma A, et al. ASP0028 in combination with
Transplant Proc 1999;31:1178–9. suboptimal-dose of tacrolimus in cynomolgus monkey renal trans-
81. Chiba K, Hoshino Y, Suzuki C, et al. FTY720, a novel immunosup- plantation model. Transpl Immunol 2017;40:57–65.
pressant possessing unique mechanisms. I. Prolongation of skin 102. Budde K, Schmouder L, Nashan B, et al. Pharmacodynamics of sin-
allograft survival and synergistic effect in combination with cyclo- gle doses of the novel immunosuppressant FTY720 in stable renal
sporine in rats. Transplant Proc 1996;28:1056–9. transplant patients. Am J Transplant 2003;3:846–54.
82. Gao M, Liu Y, Xiao Y, et al. Prolonging survival of corneal trans- 103. Tedesco-Silva H, Pescovitz MD, Cibrik D, et al. Randomized con-
plantation by selective sphingosine-1-phosphate receptor 1 agonist. trolled trial of FTY720 versus MMF in de novo renal transplantation.
PLoS ONE 2014;9e105693. Transplantation 2006;82:1689–97.
20 • Other Forms of Immunosuppression 327

104. Tedesco-Silva H, Szakaly P, Shoker A, et al. FTY720 versus myco- 125. Yoshimura N, Nakao T, Nakamura T, et al. Effectiveness of the com-
phenolate mofetil in de novo renal transplantation: six-month bination of everolimus and tacrolimus with high dosage of mizorib-
results of a double-blind study. Transplantation 2007;84:885–92. ine for living donor-related kidney transplantation. Transplant Proc
105. Salvadori M, Budde K, Charpentier B, et al. FTY720 versus MMF 2016;48:786–9.
with cyclosporine in de novo renal transplantation: a 1-year, ran- 126. Akioka K, Ishikawa T, Osaka M, et al. Hyperuricemia and acute
domized controlled trial in Europe and Australasia. Am J Transplant renal failure in renal transplant recipients treated with high-dose
2006;6:2912–21. mizoribine. Transplant Proc 2017;49:73–7.
106. Mulgaonkar S, Tedesco H, Oppenheimer F, Walker R, Kunzendorf 127. Behbod F, Erwin-Cohen RA, Wang ME, et al. Concomitant inhibition
U, Russ G, et al. FTY720/cyclosporine regimens in de novo renal of janus kinase 3 and calcineurin-dependent signaling pathways
transplantation: a 1-year dose-finding study. Am J Transplant synergistically prolongs the survival of rat heart allografts. J Immu-
2006;6:1848–57. nol 2001;166:3724–32.
107. Hoitsma AJ, Woodle ES, Abramowicz D, Proot P, Vanrenterghem 128. Kirken RA, Erwin RA, Taub D, et al. Tyrphostin AG-490 inhibits
Y. FTY720 combined with tacrolimus in de novo renal transplanta- cytokine-mediated JAK3/STAT5a/b signal transduction and cellu-
tion: 1-year, multicenter, open-label randomized study. Nephrol Dial lar proliferation of antigen-activated human T cells. J Leukoc Biol
Transplant 2011;26:3802–5. 1999;65:891–9.
108. Tedesco-Silva H, Lorber MI, Foster CE, et al. FTY720 and everolimus 129. Kudlacz E, Perry B, Sawyer P, et al. The novel JAK-3 inhibitor
in de novo renal transplant patients at risk for delayed graft function: CP-690550 is a potent immunosuppressive agent in various murine
results of an exploratory one-yr multicenter study. Clin Transplant models. Am J Transplant 2004;4:51–7.
2009;23:589–99. 130. Stepkowski SM, Kao J, Wang ME, et al. The Mannich base NC1153
109. Ettenger R, Schmouder R, Kovarik JM, Bastien MC, Hoyer PF. promotes long-term allograft survival and spares the recipient from
Pharmacokinetics, pharmacodynamics, safety, and tolerability of multiple toxicities. J Immunol 2005;175:4236–46.
single-dose fingolimod (FTY720) in adolescents with stable renal 131. Quaedackers ME, Mol W, Korevaar SS, et al. Monitoring of the
transplants. Pediatr Transplant 2011;15:406–13. immunomodulatory effect of CP-690,550 by analysis of the JAK/
110. Oppenheimer F, Mulgaonkar S, Ferguson R, et al. Impact of long- STAT pathway in kidney transplant patients. Transplantation
term therapy with FTY720 or mycophenolate mofetil on cardiac 2009;88:1002–9.
conduction and rhythm in stable adult renal transplant patients. 132. Macchi P, Villa A, Giliani S, et al. Mutations of Jak-3 gene in patients
Transplantation 2007;83:645–8. with autosomal severe combined immune deficiency (SCID). Nature
111. Mizuno K, Tsujino M, Takada M, Hayashi M, Atsumi K. Studies on 1995;377:65–8.
bredinin. I. Isolation, characterization and biological properties. 133. Roberts JL, Lengi A, Brown SM, et al. Janus kinase 3 (JAK3) deficiency:
J Antibiot (Tokyo) 1974;27:775–82. clinical, immunologic, and molecular analyses of 10 patients and out-
112. Ichikawa Y, Ihara H, Takahara S, et al. The immunosuppressive comes of stem cell transplantation. Blood 2004;103:2009–18.
mode of action of mizoribine. Transplantation 1984;38:262–7. 134. Russell SM, Johnston JA, Noguchi M, et al. Interaction of IL-2R beta
113. Aso K, Uchida H, Sato K, et al. Immunosuppression with low-dose and gamma c chains with Jak1 and Jak3:implications for XSCID and
cyclosporine combined with bredinin and prednisolone. Transplant XCID. Science 1994;266:1042–5.
Proc 1987;19:1955–8. 135. Russell SM, Tayebi N, Nakajima H, et al. Mutation of Jak3 in a
114. Kokado Y, Ishibashi M, Jiang H, Takahara S, Sonoda T. A new patient with SCID: essential role of Jak3 in lymphoid development.
triple-drug induction therapy with low dose cyclosporine, mizorib- Science 1995;270:797–800.
ine and prednisolone in renal transplantation. Transplant Proc 136. Kundig TM, Schorle H, Bachmann MF, Hengartner H, Zinkernagel
1989;21:1575–8. RM, Horak I. Immune responses in interleukin-2-deficient mice. Sci-
115. Takeuchi N, Ohshima S, Matsuura O, et al. Immunosuppression ence 1993;262:1059–61.
with low-dose cyclosporine, mizoribine, and steroids in living-related 137. Malek TR, Bayer AL. Tolerance, not immunity, crucially depends on
kidney transplantation. Transplant Proc 1994;26:1907–9. IL-2. Nat Rev Immunol 2004;4:665–74.
116. Tanabe K, Tokumoto T, Ishikawa N, et al. Long-term results in 138. Malek TR, Yu A, Vincek V, Scibelli P, Kong L. CD4 regulatory T
mizoribine-treated renal transplant recipients: a prospective, ran- cells prevent lethal autoimmunity in IL-2Rbeta-deficient mice.
domized trial of mizoribine and azathioprine under cyclosporine- Implications for the nonredundant function of IL-2. Immunity
based immunosuppression. Transplant Proc 1999;31:2877–9. 2002;17:167–78.
117. Hosoya M, Shigeta S, Ishii T, Suzuki H, De CE. Comparative inhibi- 139. Jiang JK, Ghoreschi K, Deflorian F, et al. Examining the chirality, con-
tory effects of various nucleoside and nonnucleoside analogues formation and selective kinase inhibition of 3-((3R,4R)-4-methyl-
on replication of influenza virus types A and B in vitro and in ovo. 3-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)piperidin-1-y
J Infect Dis 1993;168:641–6. l)-3-oxopropanenitrile (CP-690,550). J Med Chem 2008;51:8012–
118. Naka K, Ikeda M, Abe K, Dansako H, Kato N. Mizoribine inhibits hep- 8.
atitis C virus RNA replication: effect of combination with interferon- 140. Karaman MW, Herrgard S, Treiber DK, et al. A quantitative analysis
alpha. Biochem Biophys Res Commun 2005;330:871–9. of kinase inhibitor selectivity. Nat Biotechnol 2008;26:127–32.
119. Saijo M, Morikawa S, Fukushi S, et al. Inhibitory effect of mizoribine 141. Meyer DM, Jesson MI, Li X, et al. Anti-inflammatory activity and
and ribavirin on the replication of severe acute respiratory syndrome neutrophil reductions mediated by the JAK1/JAK3 inhibitor,
(SARS)-associated coronavirus. Antiviral Res 2005;66:159–63. CP-690,550, in rat adjuvant-induced arthritis. J Inflamm (Lond).
120. Shigeta S. Recent progress in antiviral chemotherapy for respiratory 2010;7:41–7.
syncytial virus infections. Expert Opin Investig Drugs 2000;9:221– 142. Soth M, Hermann JC, Yee C, et al. 3-Amido pyrrolopyrazine JAK
35. kinase inhibitors: development of a JAK3 vs JAK1 selective inhibi-
121. Funahashi Y, Hattori R, Kinukawa T, Kimura H, Nishiyama Y, tor and evaluation in cellular and in vivo models. J Med Chem
Gotoh M. Conversion from mycophenolate mofetil to mizoribine for 2013;56:345–56.
patients with positive polyomavirus type BK in urine. Transplant 143. Thoma G, Nuninger F, Falchetto R, et al. Identification of a potent
Proc 2008;40:2268–70. Janus kinase 3 inhibitor with high selectivity within the Janus kinase
122. Liu H, Wang Y, Fan B, et al. Improvement in severe mycophenolic family. J Med Chem 2011;54:284–8.
acid-associated gastrointestinal symptoms after changing enteric- 144. Williams NK, Bamert RS, Patel O, et al. Dissecting specificity in the
coated mycophenolate sodium to mizoribine in renal transplant Janus kinases: the structures of JAK-specific inhibitors complexed
recipients: two case reports. Intern Med 2016;55:2005–10. to the JAK1 and JAK2 protein tyrosine kinase domains. J Mol Biol
123. Shi Y, Liu H, Chen XG, Shen ZY. Comparison of mizoribine and 2009;387:219–32.
mycophenolate mofetil with a tacrolimus-based immunosuppressive 145. Borie DC, Larson MJ, Flores MG, et al. Combined use of the JAK3
regimen in living-donor kidney transplantation recipients: a retro- inhibitor CP-690,550 with mycophenolate mofetil to prevent kid-
spective study in China. Transplant Proc 2017;49:26–31. ney allograft rejection in nonhuman primates. Transplantation
124. Ushigome H, Uchida K, Nishimura K, et al. Efficacy and safety of 2005;80:1756–64.
high-dose mizoribine combined with cyclosporine, basiliximab, and 146. Borie DC, O’Shea JJ, Changelian PS. JAK3 inhibition, a viable new
corticosteroids in renal transplantation: a Japanese multicenter modality of immunosuppression for solid organ transplants. Trends
study. Transplant Proc 2016;48:794–8. Mol Med 2004;10:532–41.
328 Kidney Transplantation: Principles and Practice

147. Changelian PS, Flanagan ME, Ball DJ, et al. Prevention of organ 170. Trotter JF, Levy G. Sotrastaurin in liver transplantation: has it had a
allograft rejection by a specific Janus kinase 3 inhibitor. Science fair trial? Am J Transplant 2015;15:1137–8.
2003;302:875–8. 171. Everly JJ, Walsh RC, Alloway RR, Woodle ES. Proteasome inhibi-
148. Paniagua R, Si MS, Flores MG, et al. Effects of JAK3 inhibition with tion for antibody-mediated rejection. Curr Opin Organ Transplant
CP-690,550 on immune cell populations and their functions in 2009;14:662–6.
nonhuman primate recipients of kidney allografts. Transplantation 172. Sunwoo JB, Chen Z, Dong G, et al. Novel proteasome inhibitor
2005;80:1283–92. PS-341 inhibits activation of nuclear factor-kappa B, cell survival,
149. Conklyn M, Andresen C, Changelian P, Kudlacz E. The JAK3 inhibi- tumor growth, and angiogenesis in squamous cell carcinoma. Clin
tor CP-690550 selectively reduces NK and CD8+ cell numbers in Cancer Res 2001;7:1419–28.
cynomolgus monkey blood following chronic oral dosing. J Leukoc 173. Perry DK, Burns JM, Pollinger HS, Amiot BP, Gloor JM, Gores GJ, et al.
Biol 2004;76:1248–55. Proteasome inhibition causes apoptosis of normal human plasma cells
150. Sewgobind VD, Quaedackers ME, van der Laan LJ, et al. The Jak preventing alloantibody production. Am J Transplant 2009;9:201–9.
inhibitor CP-690,550 preserves the function of CD4CD25FoxP3 174. Lang VR, Mielenz D, Neubert K, et al. The early marginal zone B
regulatory T cells and inhibits effector T cells. Am J Transplant cell-initiated T-independent type 2 response resists the proteasome
2010;10:1785–95. inhibitor bortezomib. J Immunol 2010;185:5637–47.
151. Busque S, Leventhal J, Brennan DC, et al. Calcineurin-inhibitor- 175. Neubert K, Meister S, Moser K, et al. The proteasome inhibitor bort-
free immunosuppression based on the JAK inhibitor CP-690,550: a ezomib depletes plasma cells and protects mice with lupus-like dis-
pilot study in de novo kidney allograft recipients. Am J Transplant ease from nephritis. Nat Med 2008;14:748–55.
2009;9:1936–45. 176. Vogelbacher R, Meister S, Guckel E, et al. Bortezomib and sirolimus
152. Vincenti F, Tedesco SH, Busque S, et al. Randomized phase 2b trial inhibit the chronic active antibody-mediated rejection in experi-
of tofacitinib (CP-690,550) in de novo kidney transplant patients: mental renal transplantation in the rat. Nephrol Dial Transplant
efficacy, renal function and safety at 1 year. Am J Transplant 2010;25:3764–73.
2012;12:2446–56. 177. Shah N, Meouchy J, Qazi Y. Bortezomib in kidney transplantation.
153. Vincenti F, Silva HT, Busque S, et al. Evaluation of the effect of tofaci- Curr Opin Organ Transplant 2015;20:652–6.
tinib exposure on outcomes in kidney transplant patients. Am J 178. Diwan TS, Raghavaiah S, Burns JM, Kremers WK, Gloor JM, Stegall
Transplant 2015;15:1644–53. MD. The impact of proteasome inhibition on alloantibody-producing
154. van GE, Weimar W, Gaston R, et al. Phase 1 dose-escalation study of plasma cells in vivo. Transplantation 2011;91:536–41.
CP-690 550 in stable renal allograft recipients: preliminary findings 179. Everly MJ, Everly JJ, Susskind B, et al. Bortezomib provides effective
of safety, tolerability, effects on lymphocyte subsets and pharmaco- therapy for antibody- and cell-mediated acute rejection. Transplan-
kinetics. Am J Transplant 2008;8:1711–8. tation 2008;86:1754–61.
155. Baan CC, Kannegieter NM, Felipe CR, Tedesco Jr SH. Targeting JAK/ 180. Sberro-Soussan R, Zuber J, Suberbielle-Boissel C, et al. Bortezomib
STAT signaling to prevent rejection after kidney transplantation: a as the sole post-renal transplantation desensitization agent does
reappraisal. Transplantation 2016;100:1833–9. not decrease donor-specific anti-HLA antibodies. Am J Transplant
156. Moore CA, Iasella CJ, Venkataramanan R, et al. Janus kinase inhi- 2010;10:681–6.
bition for immunosuppression in solid organ transplantation: Is 181. Walsh RC, Everly JJ, Brailey P, et al. Proteasome inhibitor-based pri-
there a role in complex immunologic challenges? Hum Immunol mary therapy for antibody-mediated renal allograft rejection. Trans-
2017;78:64–71. plantation 2010;89:277–84.
157. Kovarik JM, Slade A. Overview of sotrastaurin clinical pharmacoki- 182. Woodle ES, Shields AR, Ejaz NS, et al. Prospective iterative trial
netics. Ther Drug Monit 2010;32:540–3. of proteasome inhibitor-based desensitization. Am J Transplant
158. Kovarik JM, JU Steiger, Grinyo JM, et al. Pharmacokinetics of 2015;15:101–18.
sotrastaurin combined with tacrolimus or mycophenolic acid in de 183. Moreno Gonzales MA, Gandhi MJ, Schinstock CA, et al. 32 Doses of
novo kidney transplant recipients. Transplantation 2011;91:317– bortezomib for desensitization is not well tolerated and is associated
22. with only modest reductions in anti-HLA antibody. Transplantation
159. Kovarik JM, Stitah S, Slade A, et al. Sotrastaurin and cyclosporine 2017;101:1222–7.
drug interaction study in healthy subjects. Biopharm Drug Dispos 184. Kwun J, Burghuber C, Manook M, et al. Humoral compensation after
2010;31:331–9. bortezomib treatment of allosensitized recipients. J Am Soc Nephrol
160. Spitaler M, Cantrell DA. Protein kinase C and beyond. Nat Immunol 2017;28:1991–6.
2004;5:785–90. 185. Kortuem KM, Stewart AK. Carfilzomib. Blood 2013;121:893–7.
161. Mecklenbrauker I, Saijo K, Zheng NY, Leitges M, Tarakhovsky A. 186. De Sousa-Amorim E, Revuelta I, Diekmann F, et al. High incidence
Protein kinase C-delta controls self-antigen-induced B-cell tolerance. of paralytic ileus after bortezomib treatment of antibody-mediated
Nature 2002;416:860–5. rejection in kidney transplant recipients: report of 2 cases. Trans-
162. Tan SL, Parker PJ. Emerging and diverse roles of protein kinase C in plantation 2015;99:e170–1.
immune cell signalling. Biochem J 2003;376:545–52. 187. Everly MJ, Terasaki PI, Hopfield J, Trivedi HL, Kaneku H. Protective
163. Merani S, McCall M, Pawlick RL, Edgar RL, Davis J, Toso C, et al. immunity remains intact after antibody removal by means of protea-
AEB071 (sotrastaurin) does not exhibit toxic effects on human islets some inhibition. Transplantation 2010;90:1493–8.
in vitro, nor after transplantation into immunodeficient mice. Islets 188. Bikle D. Nonclassic actions of vitamin D. J Clin Endocrinol Metab
2011;3:338–43. 2009;94:26–34.
164. Bigaud M, Wieczorek G, Beerli C, et al. Sotrastaurin (AEB071) alone 189. Verstuyf A, Carmeliet G, Bouillon R, Mathieu C. Vitamin D: a pleio-
and in combination with cyclosporine A prolongs survival times of tropic hormone. Kidney Int 2010;78:140–5.
non-human primate recipients of life-supporting kidney allografts. 190. Provvedini DM, Tsoukas CD, Deftos LJ, Manolagas SC. 1,25-
Transplantation 2012;93:156–64. dihydroxyvitamin D3 receptors in human leukocytes. Science
165. Budde K, Sommerer C, Becker T, et al. Sotrastaurin, a novel small 1983;221:1181–3.
molecule inhibiting protein kinase C: first clinical results in renal- 191. Takahashi K, Nakayama Y, Horiuchi H, et al. Human neutro-
transplant recipients. Am J Transplant 2010;10:571–81. phils express messenger RNA of vitamin D receptor and respond to
166. Friman S, Arns W, Nashan B, et al. Sotrastaurin, a novel small mol- 1alpha,25-dihydroxyvitamin D3. Immunopharmacol Immunotoxi-
ecule inhibiting protein-kinase C: randomized phase II study in renal col 2002;24:335–47.
transplant recipients. Am J Transplant 2011;11:1444–55. 192. Baeke F, Korf H, Overbergh L, et al. Human T lymphocytes are direct
167. Tedesco-Silva H, Kho MM, Hartmann A, et al. Sotrastaurin in cal- targets of 1,25-dihydroxyvitamin D3 in the immune system. J Ste-
cineurin inhibitor-free regimen using everolimus in de novo kidney roid Biochem Mol Biol 2010;121:221–7.
transplant recipients. Am J Transplant 2013;13:1757–68. 193. Overbergh L, Decallonne B, Valckx D, et al. Identification and
168. Russ GR, Tedesco-Silva H, Kuypers DR, et al. Efficacy of sotrastaurin immune regulation of 25-hydroxyvitamin D-1-alpha-hydroxylase
plus tacrolimus after de novo kidney transplantation: randomized, in murine macrophages. Clin Exp Immunol 2000;120:139–46.
phase II trial results. Am J Transplant 2013;13:1746–56. 194. Stoffels K, Overbergh L, Bouillon R, Mathieu C. Immune regula-
169. Pascher A, De SP, Pratschke J, et al. Protein kinase C inhibitor tion of 1alpha-hydroxylase in murine peritoneal macrophages:
sotrastaurin in de novo liver transplant recipients: a randomized unravelling the IFN-gamma pathway. J Steroid Biochem Mol Biol
phase II trial. Am J Transplant 2015;15:1283–92. 2007;103:567–71.
20 • Other Forms of Immunosuppression 329

195. Dusso AS, Kamimura S, Gallieni M, et al. gamma-Interferon-induced 216. Veyron P, Pamphile R, Binderup L, Touraine JL. New 20-epi-vitamin
resistance to 1,25-(OH)2 D3 in human monocytes and macro- D3 analogs: immunosuppressive effects on skin allograft survival.
phages: a mechanism for the hypercalcemia of various granuloma- Transplant Proc 1995;27:450.
toses. J Clin Endocrinol Metab 1997;82:2222–32. 217. Vos R, Ruttens D, Verleden SE, et al. High-dose vitamin D after
196. Gregori S, Casorati M, Amuchastegui S, Smiroldo S, Davalli AM, lung transplantation: a randomized trial. J Heart Lung Transplant
Adorini L. Regulatory T cells induced by 1 alpha,25-dihydroxyvita- 2017;10.
min D3 and mycophenolate mofetil treatment mediate transplanta- 218. Cippa PE, Kraus AK, Edenhofer I, et al. The BH3-mimetic ABT-737
tion tolerance. J Immunol 2001;167:1945–53. inhibits allogeneic immune responses. Transpl Int 2011;24:722–
197. Penna G, Adorini L. 1 Alpha,25-dihydroxyvitamin D3 inhibits 32.
differentiation, maturation, activation, and survival of dendritic 219. Gabriel SS, Bon N, Chen J, et al. Distinctive expression of Bcl-2 factors
cells leading to impaired alloreactive T cell activation. J Immunol in regulatory T cells determines a pharmacological target to induce
2000;164:2405–11. immunological tolerance. Front Immunol 2016;7:73.
198. van Halteren AG, van EE, de Jong EC, Bouillon R, Roep BO, Mathieu 220. Cippa PE, Kamarashev J, Chen J, et al. Synergistic Bcl-2 inhibition by
C. Redirection of human autoreactive T-cells Upon interaction with ABT-737 and cyclosporine A. Apoptosis 2013;18:315–23.
dendritic cells modulated by TX527, an analog of 1,25 dihydroxyvi- 221. Cippa PE, Gabriel SS, Chen J, et al. Targeting apoptosis to induce
tamin D(3). Diabetes 2002;51:2119–25. stable mixed hematopoietic chimerism and long-term allograft
199. Baeke F, Takiishi T, Korf H, Gysemans C, Mathieu C. Vitamin survival without myelosuppressive conditioning in mice. Blood
D: modulator of the immune system. Curr Opin Pharmacol 2013;122:1669–77.
2010;10:482–96. 222. Cippa PE, Gabriel SS, Kraus AK, et al. Bcl-2 inhibition to over-
200. Jeffery LE, Burke F, Mura M, et al. 1,25-Dihydroxyvitamin D3 and come memory cell barriers in transplantation. Am J Transplant
IL-2 combine to inhibit T cell production of inflammatory cytokines 2014;14:333–42.
and promote development of regulatory T cells expressing CTLA-4 223. Zhang L, You J, Sidhu J, et al. Abrogation of chronic rejection in rat
and FoxP3. J Immunol 2009;183:5458–67. model system involves modulation of the mTORC1 and mTORC2
201. Tang J, Zhou R, Luger D, et al. Calcitriol suppresses antiretinal auto- pathways. Transplantation 2013;96:782–90.
immunity through inhibitory effects on the Th17 effector response. 224. Pike KG, Malagu K, Hummersone MG, et al. Optimization of potent
J Immunol 2009;182:4624–32. and selective dual mTORC1 and mTORC2 inhibitors: the discovery
202. Chen S, Sims GP, Chen XX, Gu YY, Chen S, Lipsky PE. Modulatory of AZD8055 and AZD2014. Bioorg Med Chem Lett 2013;23:1212–
effects of 1,25-dihydroxyvitamin D3 on human B cell differentiation. 6.
J Immunol 2007;179:1634–47. 225. Rosborough BR, Raich-Regue D, Liu Q, Venkataramanan R, Turn-
203. Casteels KM, Mathieu C, Waer M, et al. Prevention of type I diabetes in quist HR, Thomson AW. Adenosine triphosphate-competitive mTOR
nonobese diabetic mice by late intervention with nonhypercalcemic inhibitors: a new class of immunosuppressive agents that inhibit
analogs of 1,25-dihydroxyvitamin D3 in combination with a short allograft rejection. Am J Transplant 2014;14:2173–80.
induction course of cyclosporin A. Endocrinology 1998;139:95–102. 226. Powles T, Wheater M, Din O, et al. A randomised phase 2 study of
204. Gysemans C, Waer M, Laureys J, Bouillon R, Mathieu C. A combina- AZD2014 versus everolimus in patients with VEGF-Refractory Met-
tion of KH1060, a vitamin D(3) analogue, and cyclosporin prevents astatic Clear Cell Renal Cancer. Eur Urol 2016;69:450–6.
early graft failure and prolongs graft survival of xenogeneic islets in 227. Basu B, Dean E, Puglisi M, et al. First-in-human pharmacokinetic
nonobese diabetic mice. Transplant Proc 2001;33:2365. and pharmacodynamic study of the dual m-TORC 1/2 inhibitor
205. Branisteanu DD, Mathieu C, Bouillon R. Synergism between siroli- AZD2014. Clin Cancer Res 2015;21:3412–9.
mus and 1,25-dihydroxyvitamin D3 in vitro and in vivo. J Neuroim- 228. Gorski A, Grieb P, Korczak-Kowalska G, Wierzbicki P, Stepien-
munol 1997;79:138–47. Sopniewska B, Mrowiec T. Cladribine (2-chloro-deoxyadenosine,
206. Branisteanu DD, Waer M, Sobis H, Marcelis S, Vandeputte M, Bouil- CDA): an inhibitor of human B and T cell activation in vitro. Immu-
lon R. Prevention of murine experimental allergic encephalomyeli- nopharmacology 1993;26:197–202.
tis: cooperative effects of cyclosporine and 1 alpha, 25-(OH)2D3. 229. Gorski A, Grieb P, Makula J, Stepien-Sopniewska B, Mrowiec T,
J Neuroimmunol 1995;61:151–60. Nowaczyk M. 2-Chloro-2-deoxyadenosine—a novel immunosup-
207. van EE, Branisteanu DD, Verstuyf A, Waer M, Bouillon R, Mathieu pressive agent. Transplantation 1993;56:1253–7.
C. Analogs of 1,25-dihydroxyvitamin D3 as dose-reducing agents for 230. Schmid T, Hechenleitner P, Mark W, et al. 2-Chlorodeoxyadenosine
classical immunosuppressants. Transplantation 2000;69:1932–42. (cladribine) in combination with low-dose cyclosporin prevents
208. Bertolini DL, Araujo PR, Silva RN, Duarte AJ, Tzanno-Martins rejection after allogeneic heart and liver transplantation in the rat.
CB. Immunomodulatory effects of vitamin D analog KH1060 on Eur Surg Res 1998;30:61–8.
an experimental skin transplantation model. Transplant Proc 231. Oberhuber G, Schmid T, Thaler W, et al. Evidence that 2-chloro-
1999;31:2998–9. deoxyadenosine in combination with cyclosporine prevents rejec-
209. Hullett DA, Cantorna MT, Redaelli C, et al. Prolongation of tion after allogeneic small bowel transplantation. Transplantation
allograft survival by 1,25-dihydroxyvitamin D3. Transplantation 1994;58:743–5.
1998;66:824–8. 232. Si MS, Ji P, Tromberg BJ, et al. Farnesyltransferase inhibition: a
210. Johnsson C, Tufveson G. MC 1288—a vitamin D analogue with novel method of immunomodulation. Int Immunopharmacol
immunosuppressive effects on heart and small bowel grafts. Transpl 2003;3:475–83.
Int 1994;7:392–7. 233. Si MS, Ji P, Lee M, et al. Potent farnesyltransferase inhibitor ABT-
211. Lemire JM, Archer DC, Khulkarni A, Ince A, Uskokovic MR, Step- 100 abrogates acute allograft rejection. J Heart Lung Transplant
kowski S. Prolongation of the survival of murine cardiac allografts by 2005;24:1403–9.
the vitamin D3 analogue 1,25-dihydroxy-delta 16-cholecalciferol. 234. Liu YX, Shen NY, Liu C, Lv Y. Immunosuppressive effects of emodin:
Transplantation 1992;54:762–3. an in vivo and in vitro study. Transplant Proc 2009;41:1837–9.
212. Pakkala I, Taskinen E, Pakkala S, Raisanen-Sokolowski A. MC1288, 235. Fujine K, Abe F, Seki N, Ueda H, Hino M, Fujii T. FR252921, a novel
a vitamin D analog, prevents acute graft-versus-host disease immunosuppressive agent isolated from Pseudomonas fluorescens
in rat bone marrow transplantation. Bone Marrow Transplant no. 408813 II. In vitro property and mode of action. J Antibiot
2001;27:863–7. (Tokyo) 2003;56:62–7.
213. Raisanen-Sokolowski AK, Pakkala IS, Samila SP, Binderup L, Hayry 236. Fujine K, Tanaka M, Ohsumi K, et al. FR252921, a novel immu-
PJ, Pakkala ST. A vitamin D analog, MC1288, inhibits adventitial nosuppressive agent isolated from Pseudomonas fluorescens no.
inflammation and suppresses intimal lesions in rat aortic allografts. 408813. I. Taxonomy, fermentation, isolation, physico-chemical
Transplantation 1997;63:936–41. properties and biological activities of FR252921, FR252922 and
214. Redaelli CA, Wagner M, Gunter-Duwe D, et al. 1alpha,25-dihy- FR256523. J Antibiot (Tokyo) 2003;56:55–61.
droxyvitamin D3 shows strong and additive immunomodulatory 237. Fujine K, Ueda H, Hino M, Fujii T. FR252921, a novel immunosup-
effects with cyclosporine A in rat renal allotransplants. Kidney Int pressive agent isolated from Pseudomonas fluorescens no. 408813
2002;61:288–96. III. In vivo activities. J Antibiot (Tokyo) 2003;56:68–71.
215. Redaelli CA, Wagner M, Tien YH, et al. 1 alpha,25-Dihydroxy- 238. Thomas FT, Tepper MA, Thomas JM, Haisch CE. 15-Deoxyspergua-
cholecalciferol reduces rejection and improves survival in rat liver lin: a novel immunosuppressive drug with clinical potential. Ann NY
allografts. Hepatology 2001;34:926–34. Acad Sci 1993;685:175–92.
330 Kidney Transplantation: Principles and Practice

239. Amemiya H, Koyama I, Kyo M, et al. Outline and long-term progno- 261. Strober S, Slavin S, Gottlieb M, et al. Allograft tolerance after total
sis in 15-deoxyspergualin-treated cases. Japan Collaborative Trans- lymphoid irradiation (TLI). Immunol Rev 1979;46:87–112.
plant Study Group of NKT-01. Transplant Proc 1996;28:1156–8. 262. Waer M, Ang KK, Van der Schueren E, Vandeputte M. Allogeneic
240. Groth CG. Deoxyspergualin in allogeneic kidney and xenoge- bone marrow transplantation in mice after total lymphoid irradia-
neic islet transplantation: early clinical trials. Ann NY Acad Sci tion: influence of breeding conditions and strain of recipient mice.
1993;685:193–5. J Immunol 1984;132:991–6.
241. Takahashi K, Tanabe K, Ooba S, et al. Prophylactic use of a new 263. Waer M, Ang KK, Van der Schueren E, Vandeputte M. Influence
immunosuppressive agent, deoxyspergualin, in patients with kid- of radiation field and fractionation schedule of total lymphoid irra-
ney transplantation from ABO-incompatible or preformed antibody- diation (TLI) on the induction of suppressor cells and stable chi-
positive donors. Transplant Proc 1991;23:1078–82. merism after bone marrow transplantation in mice. J Immunol
242. Lebreton L, Annat J, Derrepas P, Dutartre P, Renaut P. Structure- 1984;132:985–90.
immunosuppressive activity relationships of new analogues of 264. Gottlieb M, Strober S, Hoppe RT, Grumet FC, Kaplan HS. Engraft-
15-deoxyspergualin. 1. Structural modifications of the hydroxygly- ment of allogeneic bone marrow without graft-versus-host disease
cine moiety. J Med Chem 1999;42:277–90. in mongrel dogs using total lymphoid irradiation. Transplantation
243. Boddy AV, Yule SM. Metabolism and pharmacokinetics of oxaza- 1980;29:487–91.
phosphorines. Clin Pharmacokinet 2000;38:291–304. 265. Howard RJ, Sutherland DE, Lum CT, et al. Kidney allograft sur-
244. de Jonge ME, Huitema AD, Rodenhuis S, Beijnen JH. Clinical vival in dogs treated with total lymphoid irradiation. Ann Surg
pharmacokinetics of cyclophosphamide. Clin Pharmacokinet 1981;193:196–200.
2005;44:1135–64. 266. Rynasiewicz JJ, Sutherland DE, Kawahara K, Najarian JS. Total lym-
245. de Jonge ME, Huitema AD, van Dam SM, Rodenhuis S, Beijnen JH. phoid irradiation: critical timing and combination with cyclosporin
Population pharmacokinetics of cyclophosphamide and its metabolites A for immunosuppression in a rat heart allograft model. J Surg Res
4-hydroxycyclophosphamide, 2-dechloroethylcyclophosphamide, 1981;30:365–71.
and phosphoramide mustard in a high-dose combination with Thio- 267. Strober S, Modry DL, Hoppe RT, et al. Induction of specific unre-
tepa and Carboplatin. Ther Drug Monit 2005;27:756–65. sponsiveness to heart allografts in mongrel dogs treated with total
246. Alarabi A, Backman U, Wikstrom B, Sjoberg O, Tufveson G. Plasma- lymphoid irradiation and antithymocyte globulin. J Immunol
pheresis in HLA-immunosensitized patients prior to kidney trans- 1984;132:1013–8.
plantation. Int J Artif Organs 1997;20:51–6. 268. Myburgh JA, Smit JA, Stark JH, Browde S. Total lymphoid irradiation
247. Fuks Z, Strober S, Bobrove AM, Sasazuki T, McMichael A, Kaplan in kidney and liver transplantation in the baboon: prolonged graft
HS. Long term effects of radiation of T and B lymphocytes in survival and alterations in T cell subsets with low cumulative dose
peripheral blood of patients with Hodgkin’s disease. J Clin Invest regimens. J Immunol 1984;132:1019–25.
1976;58:803–14. 269. Sadeghi AM, Laks H, Drinkwater DC, et al. Heart-lung xenotrans-
248. Lan F, Zeng D, Higuchi M, Higgins JP, Strober S. Host condition- plantation in primates. J Heart Lung Transplant 1991;10:442–7.
ing with total lymphoid irradiation and antithymocyte globulin 270. Bollinger RR, Fabian MA, Harland RC, et al. Total lymphoid irradia-
prevents graft-versus-host disease: the role of CD1-reactive natural tion for cardiac xenotransplantation in nonhuman primates. Trans-
killer T cells. Biol Blood Marrow Transplant 2003;9:355–63. plant Proc 1991;23:587–8.
249. Lan F, Zeng D, Higuchi M, Huie P, Higgins JP, Strober S. Predomi- 271. Panza A, Roslin MS, Coons M, et al. One-year survival of heterotopic
nance of NK1.1+TCR alpha beta+ or DX5+TCR alpha beta+ T cells heart primate xenografts treated with total lymphoid irradiation and
in mice conditioned with fractionated lymphoid irradiation pro- cyclosporine. Transplant Proc 1991;23:483–4.
tects against graft-versus-host disease: “natural suppressor” cells. 272. Thomas F, Pittman K, Ljung T, Cekada E. Deoxyspergualin is a unique
J Immunol 2001;167:2087–96. immunosuppressive agent with selective utility in inducing toler-
250. Hertel-Wulff B, Palathumpat V, Schwadron R, Strober S. Prevention ance to pancreas islet xenografts. Transplant Proc 1995;27:417–9.
of graft-versus-host disease by natural suppressor cells. Transplant 273. Tixier D, Levy C, Le Bourgeois JP, Leandri J, Loisance D. [Discor-
Proc 1987;19:536–9. dant heart xenografts. Experimental study in pigs conditioned
251. Bass H, Strober S. Deficits in T helper cells after total lymphoid by total lymphoid irradiation and cyclosporine A]. Presse Med
irradiation (TLI): reduced IL-2 secretion and normal IL-2 receptor 1992;21:1941–4.
expression in the mixed leukocyte reaction (MLR). Cell Immunol 274. Trager DK, Banks BA, Rosenbaum GE, et al. Cardiac allograft
1990;126:129–42. prolongation in mice treated with combined posttransplantation total-
252. Field EH, Becker GC. The immunosuppressive mechanism of total lymphoid irradiation and anti-L3T4 antibody therapy. Transplantation
lymphoid irradiation. I. The effect on IL-2 production and IL-2 receptor 1989;47:587–91.
expression. Transplantation 1989;48:499–505. 275. Hayamizu K, Huie P, Sibley RK, Strober S. Monocyte-derived den-
253. Field EH, Becker GC. Blocking of mixed lymphocyte reaction by dritic cell precursors facilitate tolerance to heart allografts after total
spleen cells from total lymphoid-irradiated mice involves interrup- lymphoid irradiation. Transplantation 1998;66:1285–91.
tion of the IL-2 pathway. J Immunol 1992;148:354–9. 276. Najarian JS, Ferguson RM, Sutherland DE, et al. Fractionated total
254. Palathumpat VC, Vandeputte MM, Waer M. Effects of thymus irra- lymphoid irradiation as preparative immunosuppression in high risk
diation on the immune competence of T cells after total-lymphoid renal transplantation: clinical and immunological studies. Ann Surg
irradiation. Transplantation 1990;50:95–100. 1982;196:442–52.
255. Field EH, Steinmuller D. Nondeletional mechanisms of tolerance in 277. Waer M, Vanrenterghem Y, Roels L, et al. Total lymphoid irradiation
total-lymphoid irradiation-induced bone marrow chimeras. Trans- in renal cadaveric transplantation in diabetics. Lancet 1985;2:1354.
plantation 1993;56:250–3. 278. Waer M, Leenaerts P, Vanrenterghem Y, et al. Factors determining
256. Salam A, Vandeputte M, Waer M. Clonal deletion and clonal anergy the success rate of total lymphoid irradiation in clinical kidney trans-
in allogeneic bone marrow chimeras prepared with TBI or TLI. plantation. Transplant Proc 1989;21:1796–7.
Transpl Int 1994;7(Suppl 1):S457–61. 279. Levin B, Hoppe RT, Collins G, et al. Treatment of cadaveric renal
257. Florence LS, Jiang GL, Ang KK, Stepkowski SM, Kahan BD. In vitro transplant recipients with total lymphoid irradiation, antithymocyte
analysis of T cell-mediated cytotoxicity displayed by rat heart allograft globulin, and low-dose prednisone. Lancet 1985;2:1321–5.
recipients rendered unresponsive by total-lymphoid irradiation and 280. Chow D, Saper V, Strober S. Renal transplant patients treated with total
extracted donor antigen. Transplantation 1990;49:436–44. lymphoid irradiation show specific unresponsiveness to donor antigens
258. Field EH, Rouse TM. Alloantigen priming after total lymphoid irra- the mixed leukocyte reaction (MLR). J Immunol 1987;138:3746–50.
diation alters alloimmune cytokine responses. Transplantation 281. Strober S, Dhillon M, Schubert M, et al. Acquired immune tolerance
1995;60:695–702. to cadaveric renal allografts. A study of three patients treated with
259. Stark JH, Smit JA, Myburgh JA. Nonspecific mixed lymphocyte cul- total lymphoid irradiation. N Engl J Med 1989;321:28–33.
ture inhibitory antibodies in sera of tolerant transplanted baboons 282. Hunt SA, Strober S, Hoppe RT, Stinson EB. Total lymphoid irradia-
conditioned with total lymphoid irradiation. Transplantation tion for treatment of intractable cardiac allograft rejection. J Heart
1994;57:1103–10. Lung Transplant 1991;10:211–6.
260. Nador RG, Hongo D, Baker J, Yao Z, Strober S. The changed balance 283. Levin B, Bohannon L, Warvariv V, Bry W, Collins G. Total lymphoid
of regulatory and naive T cells promotes tolerance after TLI and anti- irradiation (TLI) in the cyclosporine era—use of TLI in resistant car-
T-cell antibody conditioning. Am J Transplant 2010;10:262–72. diac allograft rejection. Transplant Proc 1989;21:1793–5.
20 • Other Forms of Immunosuppression 331

284. Salter SP, Salter MM, Kirklin JK, Bourge RC, Naftel DC. Total lym- 308. Sunder-Plassman G, Druml W, Steininger R, Honigsmann H, Kno-
phoid irradiation in the treatment of early or recurrent heart trans- bler R. Renal allograft rejection controlled by photopheresis. Lancet
plant rejection. Int J Radiat Oncol Biol Phys 1995;33:83–8. 1995;346:506.
285. Asano M, Gundry SR, Razzouk AJ, et al. Total lymphoid irradiation 309. Wolfe JT, Tomaszewski JE, Grossman RA, et al. Reversal of
for refractory rejection in pediatric heart transplantation. Ann Tho- acute renal allograft rejection by extracorporeal photopheresis:
rac Surg 2002;74:1979–85. a case presentation and review of the literature. J Clin Apher
286. Chin C, Hunt S, Robbins R, Hoppe R, Reitz B, Bernstein D. Long-term 1996;11:36–41.
follow-up after total lymphoid irradiation in pediatric heart trans- 310. Barr ML, Meiser BM, Eisen HJ, et al. Photopheresis for the prevention
plant recipients. J Heart Lung Transplant 2002;21:667–73. of rejection in cardiac transplantation. Photopheresis Transplanta-
287. Madden BP, Barros J, Backhouse L, Stamenkovic S, Tait D, Murday tion Study Group. N Engl J Med 1998;339:1744–51.
A. Intermediate term results of total lymphoid irradiation for the 311. O’Hagan AR, Stillwell PC, Arroliga A, Koo A. Photopheresis in the
treatment of non-specific graft dysfunction after heart transplanta- treatment of refractory bronchiolitis obliterans complicating lung
tion. Eur J Cardiothorac Surg 1999;15:663–6. transplantation. Chest 1999;115:1459–62.
288. Pelletier MP, Coady M, Macha M, Oyer PE, Robbins RC. Coronary 312. Salerno CT, Park SJ, Kreykes NS, et al. Adjuvant treatment of refrac-
atherosclerosis in cardiac transplant patients treated with total lym- tory lung transplant rejection with extracorporeal photopheresis.
phoid irradiation. J Heart Lung Transplant 2003;22:124–9. J Thorac Cardiovasc Surg 1999;117:1063–9.
289. Valentine VG, Robbins RC, Wehner JH, Patel HR, Berry GJ, Theodore J. 313. Starzl TE, Marchioro TL, Waddell WR. Human renal homotrans-
Total lymphoid irradiation for refractory acute rejection in heart- plantation in the presence of blood group incompatibilities. Proc Soc
lung and lung allografts. Chest 1996;109:1184–9. Exp Biol Med 1963;113:471–2.
290. Scandling JD, Busque S, Dejbakhsh-Jones S, et al. Tolerance and chi- 314. Opelz G, Terasaki PI. Effect of splenectomy on human renal trans-
merism after renal and hematopoietic-cell transplantation. N Engl J plants. Transplantation 1973;15:605–8.
Med 2008;358:362–8. 315. Pierce JC, Hume DM. The effect of splenectomy on the survival of
291. Scandling JD, Busque S, Shizuru JA, Engleman EG, Strober S. first and second renal homotransplants in man. Surg Gynecol Obstet
Induced immune tolerance for kidney transplantation. N Engl J Med 1968;127:1300–6.
2011;365:1359–60. 316. Stuart FP, Reckard CR, Ketel BL, Schulak JA. Effect of splenectomy
292. Scandling JD, Busque S, Shizuru JA, et al. Chimerism, graft survival, on first cadaver kidney transplants. Ann Surg 1980;192:553–61.
and withdrawal of immunosuppressive drugs in HLA matched and 317. Fryd DS, Sutherland DE, Simmons RL, Ferguson RM, Kjellstrand CM,
mismatched patients after living donor kidney and hematopoietic Najarian JS. Results of a prospective randomized study on the effect
cell transplantation. Am J Transplant 2015;15:695–704. of splenectomy versus no splenectomy in renal transplant patients.
293. Lim TS, O’Driscoll G, Freund J, Peterson V, Hayes H, Heywood J. Transplant Proc 1981;13:48–56.
Short-course total lymphoid irradiation for refractory cardiac trans- 318. Sutherland DE, Fryd DS, Strand MH, et al. Results of the Minnesota
plantation rejection. J Heart Lung Transplant 2007;26:1249–54. randomized prospective trial of cyclosporine versus azathioprine-
294. Edelson R, Berger C, Gasparro F, et al. Treatment of cutaneous T-cell antilymphocyte globulin for immunosuppression in renal allograft
lymphoma by extracorporeal photochemotherapy. Preliminary recipients. Am J Kidney Dis 1985;5:318–27.
results. N Engl J Med 1987;316:297–303. 319. Alexander JW, First MR, Majeski JA, et al. The late adverse effect of
295. Perotti C, Torretta L, Viarengo G, et al. Feasibility and safety of a new splenectomy on patient survival following cadaveric renal trans-
technique of extracorporeal photochemotherapy: experience of 240 plantation. Transplantation 1984;37:467–70.
procedures. Haematologica 1999;84:237–41. 320. Peters TG, Williams JW, Harmon HC, Britt LG. Splenectomy and
296. Perez MI, Edelson RL, John L, Laroche L, Berger CL. Inhibition of death in renal transplant patients. Arch Surg 1983;118:795–9.
antiskin allograft immunity induced by infusions with photo- 321. Lucas BA, Vaughn WK, Sanfilippo F, Peters TG, Alexander JW.
inactivated effector T lymphocytes (PET cells). Yale J Biol Med Effects of pretransplant splenectomy: univariate and multivariate
1989;62:595–609. analyses. Transplant Proc 1987;19:1993–5.
297. Pepino P, Berger CL, Fuzesi L, et al. Primate cardiac allo- and xeno- 322. Alexandre GP, Squifflet JP, De BM, et al. Present experiences in a
transplantation: modulation of the immune response with photo- series of 26 ABO-incompatible living donor renal allografts. Trans-
chemotherapy. Eur Surg Res 1989;21:105–13. plant Proc 1987;19:4538–42.
298. Perez M, Edelson R, Laroche L, Berger C. Inhibition of antiskin 323. Reding R, Squifflet JP, Pirson Y, et al. Living-related and unrelated
allograft immunity by infusions with syngeneic photoinactivated donor kidney transplantation: comparison between ABO-compatible
effector lymphocytes. J Invest Dermatol 1989;92:669–76. and incompatible grafts. Transplant Proc 1987;19:1511–3.
299. Yoo EK, Rook AH, Elenitsas R, Gasparro FP, Vowels BR. Apoptosis 324. Squifflet JP, De MM, Malaise J, Latinne D, Pirson Y, Alexandre GP.
induction of ultraviolet light A and photochemotherapy in cuta- Lessons learned from ABO-incompatible living donor kidney trans-
neous T-cell Lymphoma: relevance to mechanism of therapeutic plantation: 20 years later. Exp Clin Transplant 2004;2:208–13.
action. J Invest Dermatol 1996;107:235–42. 325. Ishikawa A, Itoh M, Ushlyama T, Suzuki K, Fujita K. Experience of
300. Rook AH, Suchin KR, Kao DM, et al. Photopheresis: clinical appli- ABO-incompatible living kidney transplantation after double filtra-
cations and mechanism of action. J Investig Dermatol Symp Proc tion plasmapheresis. Clin Transplant 1998;12:80–3.
1999;4:85–90. 326. Tyden G, Kumlien G, Genberg H, Sandberg J, Lundgren T, Fehrman I.
301. Baron ED, Heeger PS, Hricik DE, Schulak JA, Tary-Lehmann M, Ste- ABO incompatible kidney transplantations without splenectomy,
vens SR. Immunomodulatory effect of extracorporeal photopheresis using antigen-specific immunoadsorption and rituximab. Am J
after successful treatment of resistant renal allograft rejection. Pho- Transplant 2005;5:145–8.
todermatol Photoimmunol Photomed 2001;17:79–82. 327. Tyden G, Kumlien G, Genberg H, Sandberg J, Lundgren T, Fehrman I.
302. Gatza E, Rogers CE, Clouthier SG, et al. Extracorporeal photopheresis ABO-incompatible kidney transplantation and rituximab. Trans-
reverses experimental graft-versus-host disease through regulatory plant Proc 2005;37:3286–7.
T cells. Blood 2008;112:1515–21. 328. Orandi BJ, Zachary AA, Dagher NN, et al. Eculizumab and sple-
303. Griffith TS, Kazama H, VanOosten RL, et al. Apoptotic cells induce nectomy as salvage therapy for severe antibody-mediated rejection
tolerance by generating helpless CD8+ T cells that produce TRAIL. after HLA-incompatible kidney transplantation. Transplantation
J Immunol 2007;178:2679–87. 2014;98:857–63.
304. Dall’Amico R, Murer L, Montini G, et al. Successful treatment of 329. Orandi BJ, Lonze BE, Jackson A, et al. Splenic irradiation for the
recurrent rejection in renal transplant patients with photopheresis. treatment of severe antibody-mediated rejection. Am J Transplant
J Am Soc Nephrol 1998;9:121–7. 2016;16:3041–5.
305. Genberg H, Kumlien G, Shanwell A, Tyden G. Refractory acute renal 330. Montgomery RA, Lonze BE, King KE, et al. Desensitization in
allograft rejection successfully treated with photopheresis. Trans- HLA-incompatible kidney recipients and survival. N Engl J Med
plant Proc 2005;37:3288–9. 2011;365:318–26.
306. Horina JH, Mullegger RR, Horn S, et al. Photopheresis for renal 331. Orandi BJ, Luo X, Massie AB, et al. Survival benefit with kidney
allograft rejection. Lancet 1995;346:61. transplants from HLA-incompatible live donors. N Engl J Med
307. Kumlien G, Genberg H, Shanwell A, Tyden G. Photopheresis for 2016;374:940–50.
the treatment of refractory renal graft rejection. Transplantation 332. Takahashi K, Saito K. ABO-incompatible kidney transplantation.
2005;79:123–5. Transplant Rev (Orlando) 2013;27:1–8.
332 Kidney Transplantation: Principles and Practice

333. Clark WF, Huang SS, Walsh MW, Farah M, Hildebrand AM, Sontrop 340. Takahashi K, Saito K, Takahara S, et al. Results of a multicenter
JM. Plasmapheresis for the treatment of kidney diseases. Kidney Int prospective clinical study in Japan for evaluating efficacy and safety
2016;90:974–84. of desensitization protocol based on rituximab in ABO-incompatible
334. Inui M, Miyazato T, Furusawa M, et al. Successful kidney transplan- kidney transplantation. Clin Exp Nephrol 2017;21(4):705–13.
tation after stepwise desensitization using rituximab and bortezomib Available online at: https://doi.org/10.1007/s10157-016-1321-5.
in a highly HLA-Sensitized and ABO incompatible high titer patient. 341. Tanabe K, Ishida H, Inui M, et al. ABO-incompatible kidney trans-
Transplant Direct 2016;2:e92. plantation: long-term outcomes. Clin Transpl 2013:307–12.
335. Allen NH, Dyer P, Geoghegan T, Harris K, Lee HA, Slapak M. Plasma 342. Cardella CJ, Sutton DM, Falk JA, Katz A, Uldall PR, deVeber GA.
exchange in acute renal allograft rejection: a controlled trial. Trans- Effect of intensive plasma exchange on renal transplant rejection
plantation 1983;35:425–8. and serum cytotoxic antibody. Transplant Proc 1978;10:617–9.
336. Kirubakaran MG, Disney AP, Norman J, Pugsley DJ, Mathew TH. 343. Nojima M, Yoshimoto T, Nakao A, et al. Combined therapy of deoxys-
A controlled trial of plasmapheresis in the treatment of renal allograft pergualin and plasmapheresis: a useful treatment for antibody-
rejection. Transplantation 1981;32:164–5. mediated acute rejection after kidney transplantation. Transplant
337. Taube DH, Williams DG, Cameron JS, et al. Renal transplantation Proc 2005;37:930–3.
after removal and prevention of resynthesis of HLA antibodies. Lan- 344. KDIGO clinical practice guideline for the care of kidney transplant
cet 1984;1:824–8. recipients. Am J Transplant 2009;9:S1–155.
338. Brynger H, Rydberg L, Samuelsson B, Blohme I, Lindholm A, 345. Roberts DM, Jiang SH, Chadban SJ. The treatment of acute antibody-
Sandberg L. Renal transplantation across a blood group barrier— mediated rejection in kidney transplant recipients-a systematic
‘A2’ kidneys to ‘O’ recipients. Proc Eur Dial Transplant Assoc review. Transplantation 2012;94:775–83.
1983;19:427–31. 346. Palmer A, Taube D, Welsh K, Bewick M, Gjorstrup P, Thick M.
339. Schwartz J, Padmanabhan A, Aqui N, et al. Guidelines on the use of Removal of anti-HLA antibodies by extracorporeal immunoadsorp-
therapeutic apheresis in clinical practice-evidence-based approach tion to enable renal transplantation. Lancet 1989;1:10–2.
from the Writing Committee of the American Society of Apheresis:
the seventh special issue. J Clin Apher 2016;31:149–338.
21 Approaches to
the Induction of Tolerance
KATHRYN J. WOOD and EDWARD K. GEISSLER

CHAPTER OUTLINE Introduction Mesenchymal Stromal Cells


Historical Perspective Information From Analyzing Tolerant
Definition of “Tolerance” Recipients
Need for Tolerance in Clinical Strategies With the Potential to Induce
Transplantation Immunologic Tolerance to an Allograft
Understanding the Immunologic Mixed Chimerism
Mechanisms Behind Tolerance Induction Costimulation Blockade
Overview of T Cell Activation The B7:CD28/CTLA-4 Pathway
Mechanisms of Tolerance to Donor Antigens CD40-154 Pathway
Mechanisms of Tolerance Induction and Targeting CD3 and Accessory Molecules
Maintenance Leukocyte Depletion at the Time of
Persistence of Donor Antigen Transplantation
Deletion of Donor-Reactive Leukocytes Cell Therapy
Regulation of Immune Responses Treg Cell Therapy
Regulatory T Cells Mreg Cell Therapy
Regulatory B Cells tolDC Therapy
Regulatory Macrophages Mesenchymal Stromal Cell Therapy
Tolerogenic Dendritic Cells
Myeloid-Derived Suppressor Cells

Introduction because of the lack of immunosuppression needed when


an organ was transplanted between monozygotic twins.
HISTORICAL PERSPECTIVE Allografts that were attempted subsequently failed because
of uncontrolled acute rejection responses mounted by the
In 1951 Billingham and Medawar published a landmark immune system. The quest to identify methods of both
article entitled “The Technique of Free Skin Grafting in immunosuppression and tolerance induction in trans-
Mammals.”1 These classic experiments provide the foun- plantation began.
dation for what would become the field of transplantation
immunology and the groundwork for many concepts in
modern immunology, including immunologic memory.1
DEFINITION OF “TOLERANCE”
Further work by Medawar and his team, based upon ear- Generally, the concept of tolerance (operational) refers to the
lier writings of Ray D. Owen,2 involved skin grafting dizy- persistent survival of a transplanted allograft in the absence
gotic mammalian twin calves. The observation that these of continuing immunosuppressive therapy and an ongoing
grafts were accepted by both hosts led to the hypothesis destructive immune response targeting the graft. This func-
that a phenomenon of immunologic tolerance to the skin tional definition is appropriate, because multiple immuno-
grafts was achieved secondary to “foreign” blood cells logic mechanisms together with donor and recipient factors
that persisted in each twin as a consequence of placental are involved in both inducing and maintaining tolerance to
fusion.3 a defined set of donor antigens in vivo. Achieving functional
These breakthroughs in research translated to the clinic tolerance in transplant recipients will mandate that specific
in 1954, when Joseph Murray and colleagues performed allograft-destructive responses are “switched off” while the
the first successful kidney transplant between monozy- global immune response to pathogens and carcinogens
gotic twins at the Peter Bent Brigham Hospital in Boston, remains intact. The most robust form of transplantation
Massachusetts. The success of this procedure was in part tolerance thus has to be donor-specific, as opposed to mere

333
334 Kidney Transplantation: Principles and Practice

immuno-incompetence, a requirement that can be tested TABLE 21.1 Immunosuppressive Agents Used in Solid-
experimentally by grafting third-party transplants and by Organ Transplantation
challenging tolerant recipients to respond to virus infec-
tions and tumors. The concept of graft-specific tolerance is Class of Agent Agent
essential both to maintain long-term survival of graft and Corticosteroid Prednisone
host, and to eliminate the adverse events associated with Methyl prednisolone
lifelong nonspecific immunosuppression. Antiproliferative Azathioprine
Mycophenolate mofetil
Mycophenolate sodium
NEED FOR TOLERANCE IN CLINICAL Calcineurin inhibitor Cyclosporin
Tacrolimus
TRANSPLANTATION mTOR inhibitor Sirolimus
Everolimus
The immune response to an allograft is an ongoing dialogue Polyclonal antilymphocyte ALG
between the innate and adaptive immune system that if left antibodies ATG
unchecked will lead to the rejection of transplanted cells, Monoclonal antibodies (with Muromonab (CD3)
tissues, or organs4 (see Chapter 32). Elements of the innate target) Basiliximab (IL2α receptor-CD25)
immune system, including macrophages, neutrophils, and Alemtuzumab (CD52)
Rituximab (CD20)
complement, are activated as a consequence of tissue injury Costimulation blockade Belatacept (LEA 29Y - CTLA4-Ig)
sustained during cell isolation or organ retrieval and isch-
emia reperfusion. Activation of the innate immune system ALG, anti-lymphocyte globulin; ATG, anti-thymocyte globulin.
inevitably leads to the initiation and amplification the adap-
tive response that involves T cells, B cells, and antibodies. T
cells require a minimum of two signals for activation, anti- Unfortunately, all of these agents are globally nonspecific
gen recognition (often referred to signal 1) and costimula- in their suppressive activity, and each has some deleteri-
tion (referred to as signal 2). The majority of B cells require ous side effects.
help from T cells to initiate antibody production. Antibod- These immunosuppressive drugs can be used with good
ies reactive to donor antigens, including major and minor success to prevent or control acute allograft rejection; how-
histocompatibility antigens and blood group antigens, ever, they are less effective at controlling the long-term
can trigger or contribute to rejection early, and late, after response to injury and activation of the immune system.
transplantation. They also appear to be unable to induce the development
Multiple factors determine the decision as to how the of unresponsiveness or tolerance to the donor alloantigens
immune response to a transplant will be triggered and consistently, at least in the way they are used clinically at
evolve, including where the antigen is “seen” and the present. For nearly all transplant recipients, continued sur-
conditions that are present at the time—in particular, vival of the allograft depends on life-long administration of
the presence or absence of inflammation associated several immunosuppressive drugs. The exception to this
with activation of the innate response. In general, the statement is liver transplantation where in a proportion of
innate response is neither specific nor is it altered signifi- pediatric and adult recipients it is possible to wean a small
cantly with multiple antigenic challenges. In contrast, proportion of selected patients (<10%–20%) treated with
the adaptive response is specific for a particular anti- immunosuppressive drugs off their immunosuppression,
gen or combination of antigens and “remembers” when especially in patients with stable graft function over the first
it encounters the same antigen again, augmenting its 4 to 5 years.5–8
activity and the rapidity of the response at each encoun- The inability of current immunosuppressive drug regi-
ter. When the immune system encounters an antigen, it mens to induce tolerance to donor antigens in the majority
has to decide which type of response to make. In most of patients may be due in part to the nonspecific nature of
cases, even though one component of the immune sys- the immunosuppression resulting from their inability to dis-
tem may dominate and lead to rejection, the process is tinguish between the potentially harmful immune response
usually multifactorial, resulting from the integration of mounted against the organ graft and immune responses
multiple mechanisms. that could be beneficial, protecting the recipient from infec-
Understanding the molecular and cellular mechanisms tious pathogens and providing mechanisms to control the
that lead to allograft rejection has provided insights lead- development of malignant cells. In general, current immu-
ing to the development of therapeutics that suppress this nosuppressive drugs act by interfering with lymphocyte
unwanted immune response after transplantation. A activation and/or proliferation irrespective of the antigen
diverse collection of small-molecule and biologic immu- specificity of the responding cells (Fig. 21.1). These mecha-
nosuppressive agents are approved and available for use nisms do not discriminate between effector cells that could
in the clinic that have the potential to control or inhibit be damaging to the transplant and immune regulatory cells
allograft rejection. In the context of solid-organ transplan- that have the potential to control allograft rejection.9 This
tation, the drugs that currently are available for clinical lack of immunologic specificity means that the immune
use include azathioprine, cyclosporine, tacrolimus, myco- system of a patient treated with one or more of these ther-
phenolate mofetil, rapamycin, antithymocyte globulin, apeutic agents is compromised not only in its ability to
anti-CD25 monoclonal antibodies, belatacept, and ste- respond to the transplant, but also in its ability to respond
roids (Table 21.1). Each immunosuppressive agent acts on to any other antigenic stimuli that may be encountered
a different aspect of the immune response to an allograft after transplantation. Therefore patients are more suscep-
and can therefore be used effectively in combination. tible to infections10 (see Chapters 15, 16, 17, 18 and 19)
21 • Approaches to the Induction of Tolerance 335

participate in innate and adaptive immune responses. The


Polyclonal antibodies
Alemtusumab thymus is the key organ that shapes the T cell repertoire.
Resting T cell precursors leave the bone marrow and migrate to
lymphocyte
the thymus where they rearrange the genes that encode
Belatacept the antigen recognition structure, the T cell receptor
(TCR). Thymocytes that express TCRs with low affinity
Activation
for self-antigen presented by self major histocompatibil-
ity complexes (MHC) molecules are “neglected” and die
Cyclosporine
Tacrolimus
as they will be of no use to the host. In contrast, thymo-
cytes expressing TCRs with a high affinity for self-antigen
Azathioprine undergo programmed cell death and are “deleted” from
Mycophenolate mofetil
IL-2 the repertoire as they could respond to self-antigens in
the periphery and therefore be harmful to the host. This
Basiliximab Sirolimus leaves the T cells with receptors that have an intermediate
Everolimus affinity to enter the bloodstream where they recirculate
between blood and peripheral lymphoid tissue. A sub-
population of T cells that will be discussed later, so-called
thymus-derived or naturally occurring regulatory T cells
Proliferation (Treg), are also selected in the thymus and migrate to the
Fig. 21.1 Schematic indicating the sites of action of common immu-
periphery. A mature T cell repertoire is developed through
nosuppressive agents during an immune response. Each immunosup- this thymic selection process that is not only diverse, but
pressive agent targets a specific step in the activation and proliferation can also react to foreign antigen while still remaining tol-
of T lymphocytes. erant to self-antigens.
Naïve T cells encounter antigen in the form of a peptide
MHC complex on the surface of antigen-presenting cells
and are at a higher risk for developing cancer11–13 (see (APCs). Antigen presentation to T cells can be performed
Chapters 34 and 35). by a variety of APCs, including dendritic cells (DCs), mac-
The development of immunologic tolerance or specific rophages, and B cells, although dendritic cells are the most
unresponsiveness to donor alloantigens in the short term immunostimulatory of all the APCs and the most potent at
or the long term after transplantation appears to offer the stimulating naïve T cells to respond.14
best possibility of achieving effectiveness and specificity in As a direct consequence of organ retrieval and implan-
the control of the immune system after transplantation in tation, the tissue within the transplant is injured and
either the absence or at least reduced loads of nonspecific stressed.15,16 Cells of the innate immune system express
immunosuppressive agents. If tolerance to donor alloanti- invariant pathogen-associated pattern recognition recep-
gens could be achieved reliably, it would ensure that only tors (PRRs) that enable them to detect not only repeating
lymphocytes in the patient’s immune repertoire responding structural units expressed by pathogens, referred to as
to donor antigens were suppressed or controlled, leaving pathogen-associated molecular patterns (PAMPs),17 but
the majority of lymphocytes immune competent and able also markers of tissue injury or damage-associated molec-
to perform their normal functions after transplantation, ular patterns (DAMPs).18 Local tissue damage and isch-
including protecting the body from infection and cancer emia reperfusion injury generates many potential DAMPS,
after transplantation. This chapter is therefore dedicated to including reactive oxygen species, heat shock proteins,
discussion of the mechanisms underlying tolerance induc- heparin sulfate, and high mobility group box 1 (HMBG1),
tion and strategies used to induce unresponsiveness in after capture by the receptor for advanced glycation end
transplanted allografts. products (RAGE) complex and fibrinogen, that can bind to
PRRs. There are several families of PRRs, including trans-
membrane proteins present at the cell surface, such as
Understanding the Immunologic toll-like receptors (TLRs), and inside the cell, including the
NOD-like receptors (NODR).
Mechanisms Behind Tolerance The sensing of DAMPS by PRRs results in potent
Induction activation of the inflammasome,19 upregulating the
transcription of genes and production of microRNAs20
OVERVIEW OF T CELL ACTIVATION involved in inflammatory responses setting up ampli-
fication and feedback loops that augment the response
Understanding the cellular and molecular mechanisms of and trigger adaptive immunity. The end result is produc-
activation and immune system regulation is important for tion of inflammatory mediators including the proinflam-
the development of novel tolerance induction approaches matory cytokines, interleukin (IL)-1, IL-6, and tumor
in the context of transplantation and autoimmunity. The necrosis factor (TNF), type I interferons, chemokines
next section of the chapter sets the scene for discussing the (chemoattractant cytokines), and the rapid expression
different approaches to tolerance induction being explored of P-selectin (CD62P) by endothelial cells. These events
most actively at present. identify the transplant as a site of injury and inflamma-
Hematopoietic stem cells (HSC) present in the bone tion, modifying the activation status, permeability, and
marrow give rise to all of the leukocyte populations that viability of endothelial cells lining the vessels, triggering
336 Kidney Transplantation: Principles and Practice

the release of soluble molecules, including antigens from immune response. Immunostimulatory APCs are brought
the graft, inducing the production of acute phase pro- into close proximity to naïve T cells that may have TCRs
teins, including complement factors systemically and capable of recognizing either intact donor alloantigens
in some cases by the organ itself, stimulating the mat- via the direct or semidirect pathways of allorecognition
uration and migration of donor-derived DCs from the or donor peptides presented by recipient MHC molecules
transplant to recipient lymphoid tissue.21,22 This process via the indirect pathway. An immunologic synapse (IS) is
results in upregulation of donor MHC, costimulatory and formed by the close interaction between the APC and the T
adhesion molecules by the donor-derived APC, enabling cell that is dependent on the successful dynamic rearrange-
them to become potent stimulators of naïve T cells. The ment and polarization of the filamentous actin in the DCs
presentation of donor alloantigens to recipient T cells by cytoskeletal membrane to bring the MHC-peptide complex
donor-derived APCs results in T cell activation via the in close relation to the TCR, thereby initiating an activation
direct pathway of allorecognition.23 response.25 Specific T cell membrane compartments termed
Activation of the innate immune system within the “lipid rafts” serve as recruitment centers for costimulatory
allograft also triggers the recruitment of inflammatory leu- molecules to concentrate in the cytoskeleton allowing for
kocytes of recipient origin into the graft. These recipient closer interactions with molecules on the APC (Fig. 21.2).
APCs have the capacity to acquire donor histocompatibility For a T cell to become fully activated, a threshold num-
antigens from the graft tissue and process them into pep- ber of TCRs need to be engaged. T cell receptor recogni-
tides that can be presented to T cells via the indirect path- tion of a donor MHC-peptide complex present on an APC,
way of allorecognition. A third pathway of presentation of often referred to as signal 1, results in signal transduction
alloantigen to T cells has also been described, the so-called through the cluster of differentiation (CD3) proteins that
semidirect pathway of allorecognition whereby as a result associate with the TCR at the T cell surface. This signal
of close contact between recipient APCs and donor cells, transduction initiates a cascade of biochemical signaling
sections of membrane containing histocompatibility mol- pathways that are contributed to by interactions between
ecules are transferred from one cell to the other for presen- accessory, costimulatory, and adhesion molecules and ulti-
tation to T cells.24 mately culminates in cytokine production and proliferation
The interaction between APCs of donor or recipient ori- of the triggered T cell and its differentiation into an effector
gin and T lymphocytes is pivotal to the adaptive arm of the cell (Fig. 21.3).

CD40 CD40L
B7-1/B7-2 CD28/CTLA-4
MCH with Peptide CD3
TCR
CD4
ICAM-1 LFA-1
LFA-3 CD2
Transplanted allograft

SLC

Passenger leukocytes Rearrangement

Maturation
DC T cell

IS

Lymph node
Fig. 21.2 Formation of the immunologic synapse. Passenger leukocytes from a transplanted allograft emigrate from the organ and under the influ-
ence of secondary lymphoid tissue chemokine (SLC) migrate to the lymph nodes and spleen. En route these dendritic cells (DC) undergo maturation
and upregulation/rearrangement of their cell surface markers using mechanisms linked to lipid rafting. Once in the lymph node T cell activation ensues
upon the formation of the immunologic synapse (IS). T cell activation requires at least two signals. Signal 1 is delivered to the T cell when MHC class II
peptide complexes on the APC are recognized specifically by the T cell receptor/CD3 complex expressed by the T cell. CD4 (T cell) interacts with the
MHC class II molecule, fulfilling an adhesion and a signaling function. Second signals or costimulation is provided by additional cell surface interactions.
CD28 (T cell) can bind to B7.2 (CD86) and B7.1 (CD80) expressed by the APC. This interaction delivers a signal to the T cell that lowers the threshold for
T cell activation. CD40 on the APC can bind to its ligand, CD40L (CD154) (T cell). This interaction provides additional signals to the T cell but, in contrast
to the CD28 pathway, also delivers signals to the APC resulting in an increase in expression of B7.1 and B7.2. To ensure that the T cell engages the APC
for sufficient time for the signaling events to occur, adhesion molecules, including ICAM-1 (intracellular adhesion molecule 1) and LFA-1 (lymphocyte
function antigen 1), also engage each other.
21 • Approaches to the Induction of Tolerance 337

Accessory and costimulatory molecules that have been to ensure that once antigen recognition by TCR (signal 1)
shown to be important in triggering T cell activation on the has occurred, the threshold for activation of a naïve T cell is
T cell side include CD4, CD11b/CD18 (LFA-1), CD28, and overcome by delivering signal 2.
CD154 (CD40 ligand).26 These molecules must engage their The two-signal model of T cell activation is well accepted,
ligands on APCs, MHC class II, intracellular adhesion mol- but it is important to note that this is a simplification. The
ecule (ICAM), CD86/80 (B7-1/B7-2), and CD40 respectively cytokine and chemokine milieu present at the time these

Donor or Tolerogenic DC
recipient DC
TGF-β TH1
IDO cell
Inhibition of effector
PDL1 T cell proliferation and
HLA-G TH17 differentiation
cell
IL-10
CD50 or CD86 MHC class II
IDO
CTLA-4 LAG3 TNF?
Therapies to enhance IL-10
Treg cell function
• Ex vivo expansion Treg cell Naive
T cell
• Rapamycin
• Anti-thymocyte globulin CD40L CD40 DC
Adenosine
IL-10 Inhibition of DC
Naive Breg
TGF-β T cell cell and monocyte
Granzymes IL-10 functions
IL-35
IL-2 deprivation IL-10 CD95L
CTLA-4
CD50 or CD86 CD95 Monocyte
Treg cells block effector Apoptosis
Effector
T cell proliferation and T cell of effector
induce apoptosis lymphocytes
A

DC T cell

T cell Trogocytosis
DC DC Apoptosis
T cell
IL-10

IL-10 DN CD95
Tr1
Tolerogenic DC Treg cell CD95L
cell

CD6–CD28– CD5+IL10+
or
Treg cell Treg cell
IFN-γ
IL-10

DN
Treg cell
Treg cell
Naive
CD6+
T cell
Naive
T cell
Breg cell CD5+CD28– T cells Tolerogenic DC
IL-10
B C D
Fig. 21.3 Mechanisms used by adaptive regulatory immune cells in transplantation. (A) tTreg cells that can respond to donor alloantigens
through cross reactivity will be present in the recipient at the time of transplantation. These will be recruited to the allograft where they can suppress
ischemia reperfusion injury. In the draining lymphoid tissue nTreg cells will inhibit T cell proliferation. Breg and tolerogeneic DC will engage naïve T
cells inducing iTreg cells that will contribute to Treg cell-mediated suppression of allograft rejection within the allograft through a variety of mecha-
nisms, including production of IL-10 and TGFb, inhibition of APC function, and through alteration of amino acid and energy metabolism. (B) Tr1 cells are
FOXP3-regulatory T cells induced in the presence of IL-10 produced by either Treg cells, tolerogenic DC, or Breg, either in the draining lymphoid tissue
or within the allograft. They can suppress both APC and T cell function. (C) CD8+ Treg cells contribute to immune regulation. CD8+CD28− cells can inhibit
APC function, and while in the presence of IL-10 naïve CD8+ T cells, can be converted to CD8+Tr cells that function in a similar manner to Tr1 cells. (D)
CD4−CD8− (DN) T cells function by downregulating expression of costimulatory molecules thereby inhibiting the ability of APC to stimulate an immune
response and inducing apoptosis of DC. In addition, DN T cells can acquire alloantigen through trogocytosis, enabling them to present antigen to T cells
that results in T cell apoptosis. (Reproduced with permission from Nature Reviews Immunology28 copyright [2012] Macmillan Magazines Ltd.)
338 Kidney Transplantation: Principles and Practice

molecular engagements occur affects the differentiation MECHANISMS OF TOLERANCE INDUCTION AND
pathway a T cell takes and the course of the response. Cyto-
MAINTENANCE
kines and chemokines can modulate expression of the cell
surface molecules mentioned previously in addition to the Persistence of Donor Antigen
expression of cytokine and chemokine receptors themselves. An overriding feature in all of the mechanisms of toler-
This modulation can result in differential signaling in the T ance mentioned previously is the persistent presence of
cell and APC, tipping the balance of the response from full to donor antigen throughout the period of tolerance in vivo.
partial activation or, in some circumstances, inactivation of Many experimental models have established that donor
the cells involved, modifying dramatically the downstream antigen must be present continuously to maintain a tol-
events (i.e., cell migration patterns and the generation of erant state, before or after transplantation, irrespective
effector cells). Activation signals in the form of cytokines of the precise mechanisms involved.29–31 The source of
propagate the responses initiated by signals 1 and 2 and are the antigen can be donor-derived cells introduced before
often referred to as the third signal in T cell activation. transplantation, as is the case in models of mixed chime-
rism,32 or the graft itself after transplantation.30 In the
MECHANISMS OF TOLERANCE TO DONOR absence of antigen, tolerance is lost gradually, because
the mechanisms responsible for maintaining tolerance are
ANTIGENS no longer stimulated. During the induction and mainte-
The human immune system has evolved naturally to nance phases of tolerance, the presence of alloantigen is
respond to challenges in a precise and controlled way. A the key factor driving the outcome. As is often the case
constant balance exists to ensure an effective but not exces- with the immune system, the same element can influence
sive response to unwanted stimuli. Many mechanisms of the response both positively and negatively. In the case of
tolerance are, in fact, continuously used by the body to pre- donor antigen, presentation in the wrong context, such as
vent reactions against self-antigens that would ultimately in a proinflammatory environment as outlined previously,
lead to autoimmune pathologies.27 It is when this balance could lead to activation with the potential of destroying
in the immune system is disrupted that immune pathology the tolerant state and triggering graft rejection, but once
leading to disease can occur. Many of these same mecha- tolerance is established, persistence of antigen is critical
nisms and regulatory cell populations can be harnessed to for maintaining the tolerant state.
induce and maintain tolerance to alloantigens, at least in
animal models.28 Deletion of Donor-Reactive Leukocytes
The mechanisms identified as responsible for inducing T Cells. The death or deletion of lymphocytes capable
or maintaining tolerance to donor antigens include the of recognizing and responding to self-antigens or, after
following: transplantation, donor alloantigens is a very effective
  
mechanism for eliminating lymphocytes from the immune
Deletion of donor-reactive cells centrally in the thymus
□ 
repertoire that have the potential to damage the host or
and in the periphery
the graft, thereby creating unresponsiveness or tolerance
T cell ignorance, or a state of T cell unresponsiveness
□ 
to self or donor alloantigens. Both T cells and B cells can
that is relevant to grafts placed at “immunologically
be deleted from the repertoire in this manner. Importantly,
privileged” sites such as the cornea or brain
if this is the only mechanism in operation to either induce
Exhaustion, in which the ability of donor-reactive cells
□ 
or maintain tolerance, deletion needs to be sustained
to harm the allograft is eliminated as a result of over-
indefinitely.
stimulation and cell death
Central tolerance by clonal deletion of T cells in the thymus
Anergy, defined as a state of unresponsiveness that is
□ 
is the major mechanism by which tolerance to self-antigens
refractory to further stimulation despite the continuing
is induced. This mechanism can be exploited for inducing
presence of antigen after transplantation
tolerance to donor antigens.
Immunoregulation—an active process whereby the
□ 
Central deletion of donor alloantigen-reactive T cells has
immune response to an allograft is controlled by popula-
been particularly successful in the context of therapeutic
tions of regulatory immune cells
   strategies using donor bone marrow in combination with
To exploit regulation of the immune response to an organ nonmyeloablative therapy, such as T cell depletion or
graft for therapeutic purposes, a clearer understanding of costimulation blockade, for the induction of tolerance.33
the mechanisms by which this phenomenon operates is The clinical applicability of this strategy can be demon-
required. Although theoretically regulation could function strated by kidney transplant recipients who have previously
exclusively through a single mechanism, such as deletion undergone bone marrow transplantation from the same
of donor-reactive T cells and B cells from the repertoire (as donor because of hematologic indications.34 The hemato-
will be discussed next), at present there is little evidence to poietic macrochimerism developing in these patients is in
support this as the only or even the dominant mechanism itself evidence for tolerance, and leads to long-term graft
for inducing and maintaining unresponsiveness to cell and acceptance without the need for life-long immunosuppres-
organ transplants. The more likely scenario is that differ- sion. In mixed allogeneic chimeras in the mouse, donor-
ent mechanisms work in concert and that distinct combi- derived dendritic cells were shown to reside and persist in
nations of mechanisms are brought into play depending on the recipient thymus,35 resulting in continuous deletion of
donor and recipient characteristics, immunosuppression, donor-reactive thymocytes leading to the absence of donor-
infection, and so on, as the immune response to the trans- reactive T cells in the periphery and hence tolerance to
plant evolves. donor alloantigens.
21 • Approaches to the Induction of Tolerance 339

The challenge of these approaches is to achieve a suffi- random to enable as many foreign protein and carbohy-
cient level of chimerism reliably without using a treatment drate antigens to be recognized as possible, it inevitably
regimen that is excessively toxic or leads to potentially dev- leads to the generation of B cells that express B cell recep-
astating graft-versus-host disease (GVHD). Moreover, data tors (BCRs) that can recognize self-antigens. Estimates
regarding the necessity for durable chimerism are conflict- vary, but suggest that up to 70% of the immature B cells
ing. Data from some clinical studies suggest that transient produced are self-reactive. Of the order of one-third of
rather than persistent chimerism may be sufficient in the these immature B cells are eliminated by receptor edit-
presence of other immunosuppressive agents to achieve ing whereby new gene rearrangements result in the pro-
tolerance in some individuals,36 whereas others suggest duction of an alternate light chain that can pair with the
that only the achievement of full donor chimerism is consis- existing heavy chain, altering the antigen recognition
tent with the ability to withdraw immunosuppression and properties of the expressed BCR. When an immature B cell
maintain graft survival.37 recognizes self-antigen with high avidity, it rapidly inter-
Antigen-reactive T cells may also be deleted from the T nalizes the antigen and undergoes a period of developmen-
cell repertoire in the periphery38 either in the presence of tal arrest. As a result, lymph node homing receptors such
high doses of antigen or because of the persistent presence as CD62 ligand (CD62L) are not expressed, and the recep-
of antigen such as would occur after transplantation. The tors for B-cell activating factor (BAFF), a cytokine required
introduction of high doses of defined antigens intravenously for B cell survival, are not induced. In addition, recombi-
or orally has been shown to result in deletion of mature T nase-activating genes remain switched on, which allows
cells in the peripheral lymphoid organs.39,40 Both CD4+ and the BCR to be replaced by editing the light chain. Any B
CD8+ T cells can be eliminated by this mechanism, but in cell undergoing this process will die after 1 to 2 days if it
many cases deletion is incomplete even when high doses of fails to express a nonautoreactive receptor. Death through
antigen are used. this pathway does not require Fas and is in part because
The mechanisms by which T cells are deleted in the thy- of antigen-induced expression of the Bcl-2 interacting
mus and the periphery is an area of active investigation. mediator of cell death (Bim), which inhibits B cell survival
Two distinct modes of apoptosis have been implicated as the proteins from the Bcl-2 family.
mechanism essential for T cell death in these settings. Acti- Receptor editing is a mechanism that could be used
vation induced cell death (AICD) is a process through which to delete immature B cells capable of recognizing donor
T cells undergo cell death in the periphery.41 After restimu- alloantigens from the B cell repertoire and particularly in
lation through TCR, a number of different molecules includ- situations where the repertoire is reshaped after leukocyte
ing CD95 (Fas), tumor necrosis factor receptor 1 (TNFR1), depletion48 or the induction of mixed chimerism36 and
and tumor necrosis factor-related apoptosis inducing ligand when organs are transplanted into young infants.49
receptor (TRAILR) can play a role in AICD depending on If receptor editing fails to eliminate all of the self-reactive
the circumstances, triggering a complex series of signaling B cells generated, residual immature B cells expressing
events, which ultimately lead to caspase activation, DNA highly self-reactive receptors are triggered to die by inter-
fragmentation, cytoskeletal degradation, and cell death. action with self-antigen. This mechanism of control or
Either high doses of antigen or repetitive stimulation is nec- regulation has been studied using immunoglobulin trans-
essary for AICD in the periphery.42 genic mice and the data obtained suggest that B cells are
The Fas pathway may also play a role, in combination deleted efficiently when the antigen they recognize is mem-
with other mechanisms, in deletion of T cells at particular brane-bound. The efficiency of this process was found to
sites in the periphery, so-called immune privileged sites that be dependent on the probability that the immature B cells
include the testis and the eye.43 At these sites, transplanta- encountered the relevant self-antigen and therefore its effi-
tion of allogeneic tissues results in prolonged survival rela- ciency is clearly related to antigen density/frequency.
tive to that obtained after transplantation of the same tissue Deletion of B cells capable of recognizing donor alloanti-
at other sites. Fas-ligand expression has been shown to be gens from the immune repertoire of a transplant recipient
an important contributor enabling these sites to maintain is a mechanism that can be harnessed in transplantation.
their immune privilege status. The Fas pathway also has This mechanism of regulation is most efficient when the
been implicated in deletional tolerance after administration antigens recognized are present at high doses. Infant recipi-
of allogeneic bone marrow.44 However, attempts to harness ents of ABO-incompatible heart allografts have been shown
the immunologic potential of these immune privileged sites to delete B cells capable of making antibodies to blood group
have been met with varying degrees of success,45 suggest- antigens presented on the heart transplant.50 Mixed chime-
ing that multiple mechanisms are involved. These include rism strategies that result in the coexistence of donor and
upregulation of expression of CD152 (CTLA4), a negative recipient bone marrow should also enable donor-reactive B
regulator of T cell activation on T cells,46 and other costim- cells to be deleted if the level of chimerism achieved is suf-
ulatory molecules, such as programmed cell death 1 recep- ficient to ensure that B cells encounter donor cells.51
tors (PD-1) that can inhibit lymphocyte activation when it
binds to its ligands PD-L1 and PD-L2.47 Regulation of Immune Responses
Although the concept of antigen-specific suppression is not
B Cells. The elimination of B cells from the repertoire is new,52 there has been a resurgence of interest in the char-
also a mechanism that has evolved to ensure that poly- acterization and functional dissection of T cell mediated
reactive B cells capable of binding self-antigens are elimi- suppression, now more often called immunoregulation,
nated in the bone marrow before they enter the periphery. since it became possible to identify the cells responsible both
As rearrangement of immunoglobulin genes occurs at phenotypically and functionally.
340 Kidney Transplantation: Principles and Practice

Importantly, transplantation provided some of the earli- that CD4+ T cells expressing high levels of CD25 could regu-
est evidence for suppression or immune regulation by T cells late rejection, Hara and colleagues showed that cotransfer of
in vivo. Data from neonatal tolerance studies suggested that CD4+CD25+ T cells from tolerant animals led to indefinite skin
additional mechanisms beyond deletion of donor-reactive graft survival in 80% of immunodeficient mice reconstituted
cells were involved.53,54 In the 1970s it was demonstrated with naïve CBA effector T cells.78
that antigen-specific unresponsiveness could be transferred In the absence of any previous exposure to alloantigen
from one recipient to another.55 there are usually insufficient numbers of tTreg to prevent
It is now clear that different populations of immune rejection of a fully allogeneic (MHC + minor histocompat-
regulatory cells can play a role in controlling the immune ibility antigen mismatched) graft, because the frequency of
response after transplantation, including Treg, regulatory T cells present in the repertoire capable of making a destruc-
B cells (Breg), myeloid-derived suppressor cells (MDSC), tive response to the graft far outnumbers the relatively
dendritic cells, and regulatory macrophages (Mreg).28,56,57 small number of tTreg present and rejection occurs.78 The
fact that tTreg cannot prevent destruction of an allograft
Regulatory T Cells in the absence of immunosuppression does not mean that
Many different types of T cells with regulatory activity the cells do not function. However, under these circum-
have been described including: CD4+ T cells58,59; CD8+ stances, the balance between rejection and regulation is
T cells60–62; CD4−CD8− double-negative T cells63; natural against regulation as the suppressive activity of any Treg
killer (NK) T cells64; and γδ T cells.65 Because CD4+ T cells present is overwhelmed by the destructive response medi-
with regulatory functions have been studied in greater ated by effector T cells. The presence of preexisting donor
depth to date, this section of the chapter will focus on this alloantigen-reactive memory T cells in the recipient can
population of regulatory cells. also overwhelm immune regulation as the kinetics of
CD25+ forkhead box P3 (Foxp3)+ Treg can arise via two activation of the memory cells is very rapid and unless
distinct developmental pathways. First, as mentioned pre- very high numbers of Treg are present at the outset of the
viously, thymus-derived or naturally occurring (tTreg) response, the balance between rejection and regulation is
differentiate in the thymus and are thought to function pushed markedly in favor of rejection.79 Importantly, this
primarily to suppress responses to self-antigen and hence critical balance between graft destruction and acceptance
prevent autoimmune disease. Evidence for this comes from can be shifted in a number of ways, notably by employing
studies in patients with rare genetic defects and in mice with strategies that increase the relative frequency and/or the
either naturally occurring or genetically engineered defects. activation status and consequently the functional activity
For example, profound immune dysregulation leading to of tTreg that can then respond to donor alloantigens before
autoimmunity is observed in patients with the immune dys- or in the early period after transplantation, including by the
regulation, polyendocrinopathy, enteropathy, and X-linked infusion of ex vivo expanded Treg,80–82 and/or by inhibiting
(IPEX) syndrome. IPEX patients have been found to have a or reducing the activity of the effector cells, for example by
point mutation in the gene encoding factor Foxp3,66,67 the using immunosuppressive agents.83
master gene for T cell regulation, resulting in functional Although the observation that mice with long-term
impairment of Treg activity in vitro.68 Scurfy mice also have surviving allografts contain populations of alloantigen-­
mutations in foxp3 and a similar immune profile.69 Sec- reactive CD25+ Treg was important, these experiments
ond, when CD4+ T cells encounter antigen in a tolerogenic were unable to distinguish between Treg that were gener-
microenvironment in the periphery, such as when antigen ated by the induction strategy itself and those that arose
is presented by immature DCs or in the presence of immu- simply by the presence of the accepted allograft. In terms
nosuppressive cytokines, the CD4+ T cells differentiate into of developing potential clinical approaches, it is important
“adaptive” or antigen-induced Treg, also known as periph- to clarify whether induction strategies that ultimately lead
eral Treg (pTreg).70 In the context of transplantation it can to long-term operational tolerance can drive Treg develop-
be argued that this pathway may be an important route ment independently of the graft itself. Data demonstrating
to generating donor alloantigen-reactive Treg after trans- that exposure to alloantigen in the absence of a transplant
plantation71 and be used to sustain the unresponsive state can lead to the induction of CD4+CD25+ Treg were obtained
through a process often referred to as infectious tolerance.72 in a number of studies. For example, when CD4+CD25+
Interestingly, despite their distinct developmental origins, were isolated from mice 28 days after pretreatment with
both tTreg and pTreg rely on sustained expression of high donor alloantigen in combination with nondepleting anti-
levels of foxp3 transcription for their suppressive function. CD4 therapy, these cells prevented rejection of a test skin
A clear indication that long-term allograft survival in the graft in a sensitive adoptive transfer model.84 Critically,
absence of long-term immunosuppression, a status referred to protection of the test graft was not observed with similar
as operational transplantation tolerance, involved the pres- populations isolated from naïve, or mice treated with anti-
ence of T cells with the ability to regulate the function of naïve CD4-only or DST-only, demonstrating that tolerance medi-
alloreactive T cells, thereby preventing the rejection of a fresh ated by CD25+ Treg can indeed be induced in vivo before
graft was reported.73,74 Subsequently, this form of cellular transplantation if recipients are exposed to donor alloanti-
regulation was found to be associated with CD4+ T cells, and gen under permissive conditions. Moreover, data demon-
Hall and colleagues were the first to suggest that CD25 might strating that the presence of the allograft alone can lead to
be a useful marker for identifying CD4+ T cells with regulatory the development of T cells with regulatory properties that
activity.75 Similar data were obtained in a rat renal allograft can protect a challenge graft from rejection,30 even when
model where operational tolerance was induced by donor-spe- the allograft itself has been rejected,85 have been reported.
cific blood transfusion (DST).76,77 As a direct demonstration These examples demonstrate that T cells with regulatory
21 • Approaches to the Induction of Tolerance 341

activity can be induced in the presence of alloantigen in the production and has been reported to be necessary for acti-
form of the allograft or an infusion of alloantigen or indeed vation of Breg, enabling them to manifest their functional
both, and that these cells can contribute to controlling sub- activity and suppress Th1 differentiation. It has been sug-
sequent responses to alloantigen in vivo. gested that there is a link between regulatory B cells and
Another important issue is understanding where Treg T cells, with Breg acting as potent generators of Treg. Breg
that can control allograft rejection function most effectively have been described in both mouse and human. Mouse
and where they can be found or detected in vivo. There is evi- Breg express T cell Ig domain and mucin domain protein 1
dence that the location in which Treg function may change (Tim-1).96 Although human regulatory B cells comprise a
with time after transplantation. Early in the response after small subset of the total B cell pool, they share some proper-
transplantation, Treg have been shown to be present and ties with their mouse counterparts, including an immature
functionally active in the draining lymph nodes, whereas phenotype.56
later in the posttransplant course Treg have been shown In transplantation, there is evidence that Breg may play
to function within the allograft itself.86,87 Indeed there is a role in controlling immune responses to alloantigens.
increasing evidence that an important site of immune regu- In experimental models, a shift in both peripheral and
lation is within the allograft itself, where Treg function to intragraft gene expression from IgG to IgM was observed,
create an environment that is permissive of immune con- and IgM+, but not IgG+ B cell clusters within rat kidney
trol.87–89 Moreover, reexposing Treg to antigen in a tissue allografts.97 In mice, Tim-1 ligation on B cells induced Tim-
may enable them to become more potent suppressors and 1+ B cells with regulatory activity,96 suggesting a poten-
therefore more effective at controlling rejection.90 tial therapeutic strategy for increasing the number of Breg
Although a significant body of work has demonstrated in vivo.
that Treg can control responses to alloantigen, most stud- Interestingly, in renal transplant recipients with a func-
ies have used either in vitro assays or experimental models tioning graft in the absence of immunosuppression, a B
whereby cells are adoptively transferred into immunodefi- cell signature was found to be associated with this state of
cient recipients where allograft rejection is driven by rela- operational tolerance to donor alloantigens.98,99 A distinct
tively small numbers of effector T cells compared with the but also B cell dominated signature of tolerance was identi-
number that would be found in a full immune repertoire fied in a separate cohort of long-term immunosuppression-
in vivo. Arguably, a much more relevant question for clini- free renal transplant recipients.100 The potential role that
cal application of this approach is what role do Treg play in immunosuppressive drug therapy plays in enabling a B cell
an intact immune system? In transplantation, Treg-specific signature to emerge in these immunosuppression-free renal
inactivation was used to show that in the anti-CD4/DST tol- transplant recipients has also been investigated. Not sur-
erance induction model described previously, the survival prisingly, data are emerging demonstrating that different
of primary heart allografts in normal, lymphoreplete recipi- immunosuppressive drugs have a significant effect.101–103
ents is also unequivocally dependent on iTreg driven by the
tolerance induction protocol.91 These data suggest that it Regulatory Macrophages
should indeed be possible to boost the function of Tregs in Macrophages have both protective and pathogenic func-
nonlymphopenic transplant patients. tions and can be divided into subgroups on the basis of their
The mechanisms used by Treg to control the activity of tissue location and their functional properties.104,105 Mature
other cell populations include cell-cell contact; CD152 has macrophages are present in tissues where they contribute to
been shown to play a critical role in this respect84 and the immune surveillance by sensing tissue damage or infection.
creation of a microenvironment through molecules such as Macrophages activated via TLRs differentiate into “classi-
IL-1078 and transforming growth factor (TGF)-β.92 cally activated” macrophages (also referred to as M1 mac-
The microenvironment created by the presence of Treg rophages) and produce inflammatory cytokines, including
in the lymphoid tissues and the allograft contribute to the TNF and IL-1. In transplantation, macrophage activation
phenomenon of linked unresponsiveness, a powerful mech- occurs initially as a result of the tissue injury that is associ-
anism that allows tolerance to “spread” from the initiating ated with ischemia and reperfusion and can contribute to
antigen to those present on cells in the immediate vicinity. early graft damage.62 Alternatively activated macrophages
For example, if a recipient’s immune system is exposed to (also referred to as M2 macrophages) are induced by expo-
a defined alloantigen before transplantation either alone sure to the cytokine IL-4, are less proinflammatory than
or in combination with immunomodulating agents, the classically activated macrophages, and in some settings can
immune response to that antigen will be modulated and create a microenvironment that downregulates inflamma-
subsequently the unresponsiveness achieved will be linked tory immune responses.106 Indeed, alternatively activated
to other molecules present on the transplanted tissue.93,94 macrophages can inhibit the production of proinflamma-
tory cytokines by classically activated macrophages. Alter-
Regulatory B Cells natively activated macrophages also play an important role
B cells with the capacity to suppress the development of in wound healing and tissue repair by producing growth
autoimmune diseases in mice were first described in the factors that can stimulate epithelial cells and fibroblasts.107
1980s. Breg express high levels of CD1d, CD21, CD24, and This function is important in organ transplantation in the
IgM and moderate levels of CD19, although some hetero- early posttransplant period where wound healing will allow
geneity has been described, suggesting that different sub- tissue homeostasis to be reestablished. However, later in
sets of Breg may exist.95 One of the characteristics of B cells the response, tissue repair responses may be less desirable
with regulatory activity is their ability to secrete IL-10. because they have been shown to contribute to delayed
CD40 stimulation appears to be required to stimulate IL-10 allograft failure by causing occlusion of blood vessels within
342 Kidney Transplantation: Principles and Practice

the allograft, a process referred to as transplant arterioscle- solid-organ allografts and bone marrow grafts,114 and their
rosis or transplant-associated vasculopathy. tolerogenic effects can be enhanced by other immunomod-
Macrophages are positioned within the lymphoid archi- ulatory agents, such as drugs that block the CD40–CD40L
tecture to interact with lymphocytes and therefore have costimulatory axis.118 However, as the characterization of
the potential to influence each other’s functional activity. DC has progressed it is clear that the state of maturity of
The interaction of Treg cells with macrophages can result the DC is not the only factor that enables them to induce
in the macrophages acquiring the properties of alterna- unresponsiveness.
tively activated or regulatory macrophages108 (see Fig. Plasmacytoid DC (pDCs) produce type-I IFNs in response
21.2). Although the interaction of macrophages with B1 B to single-stranded nucleic acids, which activate pDCs via
cells results in the formation of a regulatory macrophage both TLR-dependent and TLR-independent pathways, thus
population that produces reduced levels of inflammatory promoting antiviral innate and adaptive immune responses.
cytokines and increased levels of IL-10.109 Thus macro- Compared with conventional DCs, pDCs express lower levels
phages can influence the character of an adaptive immune of the costimulatory molecules CD80 and CD86 and higher
response, and they also can change their physiology as a levels of the inhibitory molecule PD-L1, and consequently,
result of interactions with other populations of regulatory exhibit poor immunostimulatory activity. Indeed, pDC inter-
immune cells during an immune response. action with naïve T cells has been shown to promote the gen-
It has been proposed that “regulatory macrophages” eration of Treg cells in the thymus and in the periphery. The
may represent an additional, distinct macrophage popula- molecular mechanisms used by pDCs to promote tolerance
tion whose main physiologic role is to dampen inflamma- are complex.119 In experimental models, pDCs can acquire
tory immune responses and prevent the immunopathology alloantigen in the allograft and then migrate to the draining
associated with prolonged classical macrophage activa- lymphoid tissue where they interact with T cells and induce
tion.110 In vitro, regulatory macrophages are efficient the generation of CCR4+CD4+CD25+Foxp3+ Treg cells.120 In
APCs that induce highly polarized antigen-specific T mice, prepDC appear to be the principal cell type that facili-
cell responses dominated by the production of Th2-type tates hematopoietic stem cell engraftment and induction of
cytokines. A specific subpopulation of human regulatory donor-specific skin graft tolerance in allogeneic recipients.121
macrophages, referred to as Mreg, can be isolated from In humans, pDCs produce significant amounts of IL-10,
the peripheral blood and are characterized by their mor- low levels of IFN-γ, and no detectable IL-4, IL-5, or TGF-β,
phology, expression of HLA-DR, and high levels of CD86 creating a microenvironment that promotes immune regu-
with low or absent cell-surface expression of CD14, lation.121 Tolerogenic human DCs that secrete high levels of
CD16, CD80, CD163, and CD282 (TLR2); Mreg strongly IL-10 in the absence of IL-12 induce adaptive IL-10-produc-
suppress mitogen-driven T cell proliferation in vitro.111 ing regulatory type-1 (Tr1) T cells.122 Data demonstrating
Human Mreg express high levels of indoleamine-pyrrole that DC regulate the suppressive ability, expansion, and/
2,3-dioxygenase (IDO), have the capacity to regulate or differentiation of Treg cells, with the loss of DC result-
immune responses to alloantigens, can delete activated T ing in reduced numbers of Treg cells, has been reported in
cells, and in a pilot clinical study donor-derived Mreg were mice.123 Thus tolerogenic DCs and T cells with regulatory
shown to reduce the need for immunosuppressive drugs activity may be linked and act in concert with one another
when administered intravenously to kidney transplant in some situations.
recipients.111 Recently a marker for specific for Mreg has The tolerogenic phenotype of human dendritic cells has
been identified.112 been characterized by high cell surface expression of the
Groups have also shown that host macrophages can also inhibitory receptor ILT3.124 In DCs, immunoglobulin-like
have a protective effect after transplantation. Reducing the transcript 3 (ILT3) signaling on binding cognate ligands
pool of the host macrophages in recipient mice increased such as MHC class I, HLA-G, and CD1d inhibits tyrosine
donor T cell expansion and aggravated GVHD mortality phosphorylation, NF-κB, and mitogen-activated protein
after allogeneic stem cell transplantation, suggesting that kinase (MAPK) p38 activity, transcription of certain costim-
host macrophages may have an important protective effect ulatory molecules, and the secretion of proinflammatory
by both engulfing and inhibiting the proliferation of donor cytokines and chemokines. In addition, ILT3 can interact
allogeneic T cells.113 As with the advancement of Treg with its ligands on T cells to promote inhibitory signaling in
research, further studies to specifically identify regulatory T cells. Stimulation through ILT3 appears to be a prerequi-
macrophage populations will help further dissect their role site for DC tolerization. Both ILT3high tolerogenic dendritic
in vivo. cells and soluble ILT3 have been shown to induce CD4+ T
cell anergy and differentiation of antigen-specific CD8+ sup-
Tolerogenic Dendritic Cells pressor T cells.125
DCs are crucial for priming antigen-specific T cell responses, Higher numbers of pDCs than myeloid DCs have been
including T cell responses to alloantigens,14 but they can found in the peripheral blood of pediatric liver transplant
also promote the development of immunologic unrespon- recipients who were operationally tolerant to their allograft
siveness, either in their own right or by interacting with and in those receiving low-dose immunosuppressive ther-
other leukocyte populations.22,28,114–116 apy during prospective immunosuppressive drug weaning,
Initially, immature conventional myeloid DCs that compared with patients on maintenance immunosuppres-
express low levels of MHC class II and costimulatory mol- sion.126 In addition, higher expression levels of PD-L1 and
ecules at the cell surface were identified as the dominant CD86 by pDC were found to correlate with elevated num-
form of DC that had the capacity to induce T cell toler- bers of CD4+CD25highFoxp3+ Treg cells in liver transplant
ance.117 Indeed, immature DCs can promote tolerance to recipients who were free from immunosuppressive drug
21 • Approaches to the Induction of Tolerance 343

regimens.127 These data suggest that pDCs and Treg cells transplantation, indirect evidence for a role of MDSCs has
may contribute to immune regulation in liver transplant come from the observation that transplantation tolerance
recipients. could not be induced in mice that did not express MHC class
The ability of different populations of DC to induce toler- II on circulating leukocytes, although given that many other
ance combined with clinical observations suggest that both leukocytes’ populations may also be altered in this setting.137
myeloid DCs and pDCs can promote tolerance to alloanti- The mechanisms used by MDSCs to promote tolerance to allo-
gens, and that DC maturation in itself is not the distinguish- antigens require further clarification. Some evidence suggests
ing feature that separates immunogenic DC functions from that they may act partly through the induction or sparing of
tolerogenic ones.128 Indeed, maturation is more of a contin- Treg.136 Alternatively, MDSCs have been found to upregu-
uum than an “on-off” switch, and a “semimature” state, in late heme oxygenase 1, an enzyme that has immunoregu-
which DCs are phenotypically mature but remain poor pro- latory activity through the inhibition of DC maturation and
ducers of proinflammatory cytokines, and appears to be bet- preservation of IL-10 function in addition to cytoprotective
ter linked with tolerogenic function. Maturation-resistant properties.138 It has been shown recently that granulocyte
regulatory DC do show some promise in nonhuman pri- macrophage-colony stimulating factor (GM-CSF) signal-
mates at prolonging kidney allograft survival.129 However, ing pathways through IFN-γ receptor/interferon regulatory
the use of DCs to facilitate the induction of operational toler- factor-1 and AKT/mechanistic target of rapamycin (mTOR)
ance is not without risk. DCs are arguably better known for provide monocyte licensing for suppressor function.139
the ability to prime the immune system. Indeed, DCs pulsed
with antigen are being used clinically as vaccines to stimu- Mesenchymal Stromal Cells
late immune responses to tumor antigens. Using DCs as a Mesenchymal stromal cells (MSCs) are a subpopulation
cellular therapy in transplantation may carry the risk of of multipotent cells within the bone marrow that support
sensitizing the recipient. One possible approach to reducing hematopoiesis and possess immunomodulatory and repar-
this risk is to combine DC administration and costimulatory ative properties.140 Bone marrow derived MSCs have the
blockade, with the objective of presenting donor alloan- ability to migrate to sites of inflammation, including to an
tigens to induce T cell unresponsiveness. Thus far, thera- allograft,141 but when injected intravenously are trapped in
peutic application of donor antigen pulsed DC has recently the lung and difficult to find alive thereafter.142 One mecha-
shown some modest positive effect on kidney allograft sur- nism by which MSCs may exert their immunosuppressive
vival in nonhuman primates.130 effects is by undergoing apoptosis in vivo; moreover, already
apoptotic MSCs can be effective immunomodulators by
Myeloid-Derived Suppressor Cells delivering cellular material that is phagocytized.143 Regard-
MDSC have been associated with many antigen specific and ing live cells, MSCs exposed to an inflammatory microen-
nonspecific suppressive functions, including regulation of vironment have been found to be capable of regulating
innate immunity, T cell activation, and tumor immunity. many immune effector functions through specific interac-
MDSCs are a heterogeneous population of progenitor cells tions with leukocytes that participate in both innate and
that can accumulate in tissues during an inflammatory adaptive responses. The proinflammatory cytokines IFNγ,
immune response, where they differentiate into macro- TNF-α, and/or IL-1β have been shown to activate bone
phages, DCs, and granulocytes. The expansion and activa- marrow derived MSCs144; studies indicate that their sup-
tion of MDSCs are regulated by factors produced by other pressive functions depend on activation signals associated
cells that are present in the same microenvironment, with inflammation,145,146 which makes these cells poten-
including stromal cells, activated T cells, and in tumors, the tially important in the situation of organ transplantation,
tumor cells themselves. where retrieval and transplantation of the allograft inevi-
Several MDSC subsets have been described in both mice and tably results in ischemia, reperfusion injury, and an inflam-
humans.131 Despite their heterogeneity, common phenotypi- matory microenvironment within the graft. Indeed, in vivo,
cal markers are expressed by most MDSCs, including Gr1 and MSC activation by IFN-γ has been shown to be essential for
CD11b in mice, and CD33, CD11b, CD34, and low levels of preventing GVHD.147 Once activated MSCs can control the
MHC class II in humans. Activated MDSCs suppress prolifera- activity of T cells,148 B cells,149 DCs,150 NK cells,151 and mac-
tion and cytokine production by effector T cells, B cells, and nat- rophages.149,152 Research indicates that mediation of their
ural killer (NK) cells in vitro through mechanisms that include immunomodulatory properties may depend on the presence
their expression of inducible nitric oxide synthase (iNOS) 1, of macrophages,153 which convert to an antiinflammatory
which induces nitric oxide production, and expression of argi- phenotype that expresses suppressive cytokines such as
nase 1, which depletes arginine. MDSCs also can inhibit T cell IL-10152,154 and take on the function of suppressive mac-
proliferation and modify T cell differentiation pathways. For rophages. MSC themselves have also been shown to possess
example, they can promote Treg cell differentiation in a pro- immunosuppressive activity via production of IDO, prosta-
cess requiring interferonγ (IFNγ) and IL-10.132 Interestingly, glandin E2 (PGE2), and PD-L1.155–158 Moreover, among the
interactions between MDSCs and macrophages result in a shift cocktail of other factors secreted by MSCs include matrix
of macrophages toward an alternatively activated phenotype metalloproteinases (MMPs),141,159,160 with the production
and increased production of IL-10 by MDSCs.133 of MMP2 being linked directly to a decrease in CD25 expres-
In experimental transplant models, MDSCs have been sion on CD4+ T cells and the inhibition of alloantigen-driven
shown to promote tolerance to alloantigens. Direct evidence proliferation resulting in long-term survival of allogeneic
of a tolerogenic role for MDSCs has been obtained for heart islets of Langerhans.141 Furthermore, regarding the poten-
and islet allografts in mice134,135 and by iNOS-expressing tial enhancement of Treg, MSC are known to produce fac-
MDSCs in a rat kidney allograft model.136 In bone marrow tors such as TGF-β,161–164 and through their activation by
344 Kidney Transplantation: Principles and Practice

the inflammatory cytokine IL-17A, promote Treg numbers response—the so-called liver effect.179 Based upon these
via the cyclo-oxygenase-2/PGE2 pathway.145 The immu- observations it has been suggested that liver allografts have
nomodulatory properties of MSCs on B cell function could the capacity to promote the development of immunologic
also contribute to suppressing graft rejection by inhibiting unresponsiveness. A number of mechanisms have been
alloantibody production.165 Clearly, the mechanisms by proposed to account for the liver effect, including many of
which MSC suppress the immune response are growing those discussed previously, for example, the large antigen
in complexity and are actively being studied, especially in load delivered by the liver itself, the presence of a large num-
the context of the inflammatory conditions associated with bers of passenger leukocytes that could result in deletion of
organ transplantation. donor-reactive cells, and the establishment of long-lasting
microchimerism in some recipients in addition to the pro-
INFORMATION FROM ANALYZING TOLERANT duction of soluble MHC class I by the liver.180 As a result,
RECIPIENTS liver transplant recipients have proved to be an interesting
population of immunosuppression-free patients for investi-
Operational tolerance, whereby an allograft remains func- gations designed to determine whether a molecular signa-
tional and rejection-free for more than 1 year without ture of tolerance exists.
immunosuppression, is often described as the holy grail Estimates vary, but on the order of 10% to 20% select
of transplantation. Clearly, to be able to achieve opera- adult and pediatric liver transplant recipients appear to
tional tolerance would have a major benefit for the patient, have the potential to be weaned from immunosuppression
reducing the morbidity and mortality associated with life- without the risk of rejection.6,7 Early investigations focused
long immunosuppression. However, operational tolerance on phenotyping the populations of leukocytes that were
remains a relatively rare event in the clinical setting and present in immunosuppression-free liver transplant recipi-
thus far not reliably predictable through biomarkers. ents; the majority were weaned from immunosuppression
A small number of bone marrow transplant recipients as a result of participation in well controlled clinical wean-
who subsequently required a renal transplant were trans- ing protocols.5,6,173 Interestingly, Foxp3+ cells were found
planted with a kidney from their bone marrow donor.166–168 to be a component of lymphocytes present in the periph-
In these cases long-term immunosuppression was unneces- eral blood and infiltrating the liver allografts.181 Further
sary because the recipient already was unresponsive to the analysis using transcriptional profiling demonstrated that
donor alloantigens as a result of the allogeneic chimerism there was a molecular signature of tolerance associated
that developed after the successful bone marrow trans- with tolerance in immunosuppression-free liver trans-
plant. Clinical reports of patients exhibiting spontaneous plant recipients174,182 and that this signature can be used
tolerance to an allograft after withdrawal of immunosup- to guide weaning of immunosuppression. In other words,
pression in the absence of a bone marrow transplant are still the molecular signature and the transplant biopsy can be
infrequent but cases are accruing.6,169–172 Based on these used to provide a risk assessment of which patients are more
case reports, a number of groups proposed that by study- likely to wean from immunosuppression without the risk of
ing these patients in depth it would be possible to define a rejection, which is an exciting development for the future
molecular signature of tolerance in immunosuppression- of clinical liver transplantation.174 This biomarker signa-
free kidney and liver transplant recipients5,98,99,173,174 (see ture for liver transplant tolerance, however, is best derived
later). through sampling of the liver tissue itself, rather than blood
The majority of patients who are able to stop immuno- markers.
suppression without rejection of their allograft arrive at In kidney transplantation, there are far fewer patients
that point as a result of either (1) discontinuation of immu- able to continue to have a functioning allograft in the
nosuppression by the clinical team as a consequence of the absence of immunosuppression. Nevertheless, a small
side effects of immunosuppression, nonadherence/compli- number of patients who are immunosuppression-free can
ance; or (2) as a result of weaning of immunosuppression be identified and have been studied in depth to determine
based on a clinical protocol (particularly in liver transplant whether there is a molecular signature of tolerance after
recipients) designed to minimize immunosuppressive drugs kidney transplantation. Two independent studies, one
without compromising the function of the allograft. There in Europe and the other in the US, found that there was a
is also a smaller group of patients who have been treated signature of tolerance in immunosuppression-free kidney
with protocols designed with the deliberate aim of inducing transplant recipients that could be cross-validated between
tolerance to a kidney transplant that will be discussed in the two cohorts of patients studied.98,99 Interestingly, the
detail later.34,36,37,175–177 signature of tolerance in kidney transplant recipients was
It is generally accepted that there is a hierarchy with distinct from that identified in immunosuppression-free
respect to the ease of inhibition or suppression of immune liver transplant recipients. The predominance of B cells in
responses directed against different organs. Liver allografts the peripheral blood in the absence of donor-specific anti-
and skin grafts (when they are not part of a vascular- body was found in the “kidney” signature as mentioned pre-
ized composite allograft) are at the two opposite ends of viously and was supported by gene expression data. Further
this spectrum. In experimental transplantation, liver work examining the prevalence of this signature in kidney
allografts are sometimes accepted spontaneously in the transplant recipients still being treated with immunosup-
absence of any immunosuppression or immunomodu- pression has demonstrated that different drugs in clinical
lation.178 Furthermore, in clinical transplantation, it is use have a distinct influence on the molecular signature in
often noted that the liver “protects” other organs that are a particular patient.101–103 More work is therefore required
transplanted alongside it from the full force of the rejection to determine whether a molecular signature can be used
21 • Approaches to the Induction of Tolerance 345

to identify patients currently on immunosuppression who conditioning regimen of total lymphoid irradiation and
could potentially benefit from withdrawal or minimization antithymocyte globulin after transplantation resulted in
of immunosuppression without risk to the graft. the engraftment of donor HSC. The persistent mixed chime-
rism achieved enabled immunosuppression to be discontin-
ued without rejection episodes or clinical manifestations of
Strategies With the Potential to GVHD in HLA-matched pairs.176
The limitations of myeloablative therapy prompted the
Induce Immunologic Tolerance to development and refinement of alternate approaches in
an Allograft animal models to enable high-dose bone marrow infusions
in combination with nonmyeloablative conditioning regi-
The strategies for tolerance induction being explored most mens that promote deletion of donor-reactive cells in the
actively at present invoke one or more of the mechanisms thymus. Interestingly, both costimulation blockade and T
of tolerance outlined previously, including the continu- cell depletion (see later) have been shown to be potentially
ous deletion of donor-reactive leukocytes by establishing useful in this context.188 The demonstration that transient
the presence of high levels of donor cells in the recipient macrochimerism, via nonmyeloablative conditioning, could
(mixed chimerism), short-term depletion and/or deletion achieve tolerance to renal allografts transplanted along
of donor-reactive leukocytes combined with the establish- with donor bone marrow in nonhuman primate models was
ment of immunoregulation and suppression of responses to the impetus for the translation of this approach to clinical
donor alloantigens in the longer term after transplantation, transplantation.188–190
and costimulation blockade leading to the induction of T Mixed chimerism was first used successfully to treat a
cell unresponsiveness in the presence of an organ graft.87 patient with multiple myeloma who had end-stage renal
Data emerging from the analysis of samples from patients failure and received a kidney from an HLA identical living
enrolled in tolerance induction protocols again suggest donor.34 The pretransplant workup included thymic irra-
that no single mechanism can account for or dominate the diation, whole-body irradiation, splenectomy, and donor
tolerance induction or maintenance process in transplant marrow infusion, and then relied on the administration of
recipients.36,98,99,183,184 a short course of cyclosporine posttransplantation. As in
Most of the approaches being explored in kidney trans- the primate studies, macrochimerism was only detectable
plant recipients, with the exception of some protocols for in the early period after transplantation, and after the first
inducing mixed or full donor chimerism, do not aim to 2 months the percentage of donor cells declined. Neverthe-
induce tolerance to donor antigens before transplanta- less, withdrawal of immunosuppressive therapy (cyclo-
tion. Instead they attempt to use novel strategies that are sporin) at 10 weeks posttransplantation did not trigger
relatively nonspecific in their mode of action at the time of rejection and immunosuppression-free survival (i.e., opera-
transplantation to create an environment that promotes tional tolerance persisted).
the development of operational tolerance to the graft in As a result of these encouraging data and further refine-
the long term in the presence of the allograft. This often ment of the protocols in experimental models, the mixed
means that patients are treated with immunosuppressive chimerism approach for tolerance induction was extended
drugs in the short term after transplantation with weaning to HLA-mismatched kidney transplants.36 The first two
of immunosuppression later in the transplant course. The patients enrolled in this trial were treated successfully and
safety of the patient is always a prime concern and therefore immunosuppression was withdrawn. However, the third
the development of tolerance to the graft in the long term patient developed irreversible humoral rejection 10 days
allows time for the careful analysis of the immune status of after transplantation. Careful review of this case showed
each transplant recipient before a decision to minimize or that this patient had high levels of preexisting alloantibody
withdraw immunosuppression is made. activity, but no detectable donor-specific antibody (DSA)
before transplantation. The protocol was therefore modified
to include Rituximab (anti-CD20). Stable renal allograft
MIXED CHIMERISM
function has been maintained in seven of the eight subjects
The coexistence of stable mixtures of donor and recipient in whom immunosuppression was discontinued, although
cells resulting in a state of allospecific tolerance is an idea long-term monitoring of all of these patients continues. Fur-
that was initially restricted to the field of bone marrow and ther modifications of the protocol were also introduced as
HSC transplantation, as mentioned. experience was obtained. To date, 10 patients have been
Many different approaches have been used to achieve enrolled with 6 of them maintaining long-term graft sur-
macrochimerism. Total lymphoid irradiation alone or in vival in the complete absence of immunosuppression. The
combination with bone marrow infusion has been shown other patients were returned to immunosuppressive drug
to be effective at inducing tolerance in some recipients in therapy at various times after transplantation when graft
rodents, primates, and human patients.176,185–187 Despite function was deteriorating.177
the requirement for irradiation that has arguably inhibited The analysis of samples from patients enrolled in this trial
the development and clinical application of these proto- showed that the chimeric state was achieved but persisted
cols to their fullest extent, strategies using total lymphoid only transiently. Evidence for unresponsiveness to donor
irradiation have continued to be refined, leading to more alloantigens was obtained through in vitro assays, and
recent reports that this approach can be used more safely data supporting the idea that regulatory mechanisms were
and effectively to induce tolerance to a kidney transplant. operating were obtained by analyzing foxp3 expression by
For example, Scandling and colleagues reported that a polymerase chain reaction (PCR).36 Further analysis of
346 Kidney Transplantation: Principles and Practice

serial samples has revealed that multiple mechanisms are their ligands CD28 and CD152 (CTLA-4). CD28 is expressed
responsible for the tolerant state with evidence for deletion constitutively by T cells, whereas CD152 is only expressed
of donor-reactive T cells occurring in the long term.184 later after activation. CD86 appears to interact preferen-
Macrochimerism as a strategy for inducing tolerance to tially with CD28 and may be the most important ligand for
kidney allografts has also been investigated in a clinical T cell activation, whereas CD80 may bind preferentially
trial performed at Northwestern University, Chicago.37 Fol- to CD152.195,196 In contrast to CD28, CD152 negatively
lowing myeloablative conditioning using fludarabine and regulates T cell activation when it engages its ligand on
total body irradiation, donor-derived hematopoietic stem the APC. Thus targeting CD28 will inhibit T cell activation,
cells together with facilitating cells were infused the day whereas targeting CD152 will potentiate T cell activation.
after kidney transplantation and the patients treated with Both approaches are of interest in different contexts as dem-
cyclophosphamide on day 3. Facilitating cells are a mixed onstrated by the development of CTLA-4Ig for preventing
population of CD8-αβTCR- cells present in the bone marrow allograft rejection and the development of anti-CD152 as
that can promote engraftment of hematopoietic stem cells a cancer therapy.197 Interestingly, CD152 is constitutively
and prevent the development of GVHD.191,192 Patients are expressed and plays a key role in the functional activity of
monitored for the development of macrochimerism in the regulatory T cells.84,198
peripheral blood. If macrochimerism is sustained, the recip- When CTLA4Ig, an immunoglobulin fusion protein of
ients have normal graft function in the absence of DSA, CTLA4, was produced, it was shown to inhibit graft rejec-
and the protocol biopsy is “normal,” patients are weaned tion in xenogeneic and allogeneic systems.199,200 In rodent
from immunosuppression. To date, of 31 patients enrolled models, CTLA4Ig therapy alone was found capable of induc-
in this trial, 20 have been weaned off immunosuppression ing tolerance to the graft,199 an effect that was enhanced
successfully; other patients are in the weaning stage of the when donor antigen was included in the treatment pro-
protocol. This is undoubtedly an impressive outcome. How- tocol.201 However, this effect did not translate to primate
ever, it is important to note that this protocol is not without models where CTLA4Ig monotherapy was not found to
risk. To date there have been two graft losses, two cases of induce long-term graft survival.202 Indeed a mutated form
GVHD, and two deaths. of CTLA4-Fc, LEA29Y or Belatacept was found to be a more
potent inhibitor of allograft rejection than the native mol-
ecule in primate renal transplant models.203 Belatacept
COSTIMULATION BLOCKADE
differs from CTLA4Ig by two amino acid sequences, which
As discussed previously, the activation of a T cell is depen- confers an approximately two-fold greater binding capacity
dent on multiple signals.26 When antigen recognition to CD80 and CD86. Belatacept has been approved for clini-
occurs in the absence of costimulation, T cells become cal use for the indication of prophylaxis of organ rejection in
anergic or undergo apoptosis.193 Monoclonal antibodies adult kidney transplant recipients when used in conjunction
and recombinant fusion proteins targeting costimulatory with basiliximab induction, mycophenolate mofetil (MMF),
molecules are capable of inducing unresponsiveness to and corticosteroids as maintenance immunosuppression.
donor antigens in vivo by allowing antigen recognition to Other reagents that target CD28 have also been devel-
take place in the absence of costimulation. The molecules oped. A CD28-superagonist, TGN1412, showed enhanced
that participate in costimulation pathways have attracted expansion of regulatory T cells in preclinical studies and
interest as targets for development of novel therapeutics was taken into a Phase I clinical trial. However, six healthy
not only because they have immunosuppressive proper- volunteers treated with TGN1412 experienced a mas-
ties and the ability to prevent rejection, but also to facilitate sive expansion of inflammatory T cells that resulted in
the induction of immunologic unresponsiveness to donor a cytokine storm. The trial was terminated as all volun-
alloantigens. teers experienced multiorgan failure.204 Despite this, the
Members of the immunoglobulin (Ig) and tumor necro- development of antibody-mediated selective costimula-
sis factor (TNF):TNFR superfamilies make up many of the tory blockade remains an attractive therapeutic strategy
costimulatory molecules that are integral to positive and as specifically blocking CD28, for example, would still
negative costimulation of T cell responses. Research and allow CTLA-4 signaling, potentially enhancing T cell sup-
development has been focused on two pairs of ligand- pression.205 Monovalent Fab fragments (Fv) that selec-
receptor interactions that seem to play key roles in posi- tively block CD28 have been developed to prevent signals
tive costimulation, CD80/CD86 interacting with CD28 through CD28 while allowing negative signals via CTLA-4
expressed by T cells and CD40 interacting with CD154, to remain intact. CD28 single chain Fv (scFv) fragments
which are members of the Ig and TNF:TNFR superfamilies, have been shown to significantly prolong allograft survival
respectively. CD28 blockade by a mutated version of CTLA- in primates, increasing the number of Treg in the periphery
4Ig (Belatacept) for transplantation has been licensed for as well as in the graft,206 and have been shown in a more
clinical use in renal transplantation194 (US Food and Drug recent study to induce alloantigen specific tolerance while
Administration in June 2011; European Medicines Agency preserving T cell activity against pathogens.207 This costim-
in April 2011). ulation blocking approach remains in clinical development.

THE B7:CD28/CTLA-4 PATHWAY CD40-154 PATHWAY


CD80(B7-1) and CD86(B7-2) are expressed as cell surface The CD40-CD154 pathway has been targeted using mono-
molecules by APC and are responsible for delivering addi- clonal antibody therapy to inhibit graft rejection.208 CD154,
tional or second signals to T cells when they interact with or CD40-ligand, is a type 2 membrane protein of the TNF
21 • Approaches to the Induction of Tolerance 347

family and is expressed predominantly by activated CD4+ with a depleting anti-CD4 monoclonal antibody.230,231
T cells and by a small proportion of CD8+ T cells, NK cells, Refinements of these types of protocols have resulted in
and eosinophils,209 and more recently CD154 has also been the ability to achieve long-term T cell unresponsiveness
found on platelets.210,211 Structural models predict that to protein and alloantigens in the absence of T cell deple-
CD154 forms a homotrimer that binds to its ligand, CD40, tion in experimental models. In fact, many other acces-
on the surface of APC. CD40 is expressed by B cells, macro- sory molecules, other than anti-CD4 and anti-CD8, have
phages, DCs, and thymic epithelium, and is inducible on the been targeted and shown capable of inducing tolerance to
surface of endothelial cells and fibroblasts.212 alloantigens.
The CD40-154 pathway interaction is pivotal for the OKT3, a mouse antihuman CD3 monoclonal antibody,
induction of humoral and cellular responses, with the impor- received approval for human use in 1986 in kidney trans-
tance of CD154 for B cell activation first being demonstrated plant patients undergoing rejection and later in liver and
in in vitro studies. A CD40-Ig fusion protein and a blocking cardiac transplant recipients.232 Although widely used,
monoclonal antibody to CD154 were shown to inhibit B cell OKT3 brought with it the undesired complications of the
cycling, proliferation, and differentiation into plasma cells human antimouse antibody response and a first dose reac-
in response to T cell dependent antigens.213 In vivo stud- tion characterized by fevers, chills, gastrointestinal, respira-
ies using the anti-CD154 monoclonal antibody, CD40, or tory, and cardiac complications233,234 as a result of T cell
CD154 knockout mice214,215 all demonstrated a crucial role activation and subsequent cytokine release. Consequently,
for this interaction in the generation of primary and second- many investigators have invested time into the construc-
ary humoral responses to T cell dependent antigens, class tion of pharmacotherapeutics that mimic the efficacy of
switching to antigen switching rice heterotrimeric G-protein OKT3 with less immunogenicity. A few of these OKT3-
gamma 1 (RGG1) responses, and development of germi- derived molecules in preliminary studies, such as hu12F6,
nal centers. The lack of humoral response in the absence of hOKT3gamma1(Ala-Ala), and ChAglyCD3 have proven
CD40-CD40L interaction is due not only to a lack of signaling to be more effective in T cell suppression and less immuno-
through CD40 on the B cell surface, but also the inhibition genic compared with OKT3.235–237 In transplantation mod-
of priming of CD4+ T cells through CD40L216 as a result of els, anti-CD3 therapy can be very effective at preventing
the bidirectional nature of the CD40-CD154 pathway. Sig- allograft rejection. However, timing of antibody administra-
nals through CD40 have been shown to upregulate expres- tion is critical. If anti-CD3 is given at the time of transplan-
sion of CD80 and CD86 and induce IL-12.217 Activation of tation, graft survival is not prolonged. However, if initiation
DCs through CD40 promotes their ability to present antigen of antibody therapy is delayed to the point where T cell acti-
to T cells; this may explain why targeting CD154 and block- vation has been triggered, anti-CD3 therapy results in pro-
ing its ability to interact with CD40 has a profound effect on longed allograft survival.238
T cell dependent immune responses in vivo.218 Thus target- Along with anti-CD3, antibodies to CD11a (leukocyte
ing the CD40-CD154 pathway could act to prevent both cell-­ function associated antigen-1, LFA-1) and its ligands,
mediated and antibody-mediated rejection. ICAM 1, 2, and 3, have been investigated, and data sug-
Anti-CD154 was found capable of inducing long- gests potential to prolong graft survival.239,240 However,
term acceptance of cardiac, renal, and islet allografts in an adverse side effect profile in clinical trial has resulted
several murine and nonhuman primate models either in withdrawal of efalizumab (anti-LFA1) from clinical
when used as monotherapy or in conjunction with anti- development.
CD28.200,202,219–221 Disappointingly, anti-CD154 therapy Operational tolerance induced by these strategies has
was found to have the unexpected complication of throm- been shown to develop over several weeks after the initial
bogenesis when it was translated to the clinic.222 Indeed antigen encounter31,241 and is dependent on the amount
it was found subsequently that CD154 plays key roles in of antigen infused.242 With high doses of donor bone mar-
coagulation implicated in platelet activation and the stabi- row, deletion also may be used as one of the mechanisms of
lization of thrombi.210 tolerance initially,39,243 whereas with lower doses of anti-
Monoclonal antibodies targeting CD40 have also been gen, immunoregulation is the mechanism in operation. In
found to be capable of preventing rejection in rodent and mice and rats this type of tolerance has been shown to be
primate studies,223 circumventing the complication of infectious72,244; in other words it can be transferred from
thrombogenesis associated with CD154 interference. one generation of cells to another. The maintenance of tol-
CD40-specific antibodies have already been developed for erance in these systems requires the persistent presence of
testing in human kidney transplantation (e.g., ASKP1240), antigen in the form of the organ when the thymus is still
and results from these trials are currently pending.224 Fur- functional.30
thermore, CD40 blockade potential is continuing to be
explored in experimental nonhuman primate transplant
models.225,226
Leukocyte Depletion at the Time of
Transplantation
TARGETING CD3 AND ACCESSORY MOLECULES Many tolerance induction strategies that have been inves-
Initially, administration of depleting anti-CD4 and anti-CD8 tigated in small and large animal studies result in the deple-
monoclonal antibodies were shown to result in prolonged tion of leukocytes (antithymocyte globulin, anti-CD52) or
graft survival.227–229 That this treatment strategy resulted T cells (anti-CD3 with or without immunotoxin, CD2, CD4,
in antigen-specific tolerance was shown first most clearly and CD8). In small animals, the short-term depletion of T
when a protein antigen was administered in conjunction cells appears to be sufficient in some situations for tolerance
348 Kidney Transplantation: Principles and Practice

to develop and be maintained in the long term. The success nonhuman primates have already shown some encourag-
rate can be enhanced by removing the thymus before trans- ing enhancement of kidney allograft survival,129 and evi-
plantation to prevent repopulation of the periphery with dence of their capacity to dampen immune reactions to
T cells after transplantation.245 In humans, however, leu- antigenic challenge.256
kocyte depletion with the anti-CD52 monoclonal antibody
alemtuzumab in combination with immunosuppressive MESENCHYMAL STROMAL CELL THERAPY
drugs is not sufficient to induce operational tolerance, but
can enable some degree of immunosuppression minimiza- Although more than 100 clinical trials investigating the
tion to control residual donor-reactive cells.246–251 immunomodulatory and proreparative effects of MSC are in
progress (www.clinicaltrials.gov) this form of cell therapy
is still at an early stage of development, especially in kidney
Cell Therapy transplantation. Nonetheless, there are already more than
five clinical MSC trials in kidney transplantation that have
Cellular therapies using regulatory T cells, regulatory macro- reported results, as has been summarized by van Kooten
phages (Mreg), tolerogenic dendritic cells (tolDCs), and MSCs et al.257 The largest trial thus far reported that infusion of
to suppress rejection or GVHD are being developed for use in autologous MSC resulted in a lower incidence of acute rejec-
clinical transplantation as a strategy to promote the devel- tion, a decreased risk of opportunistic infection, and better
opment of specific unresponsiveness252 that will result in at estimated renal function at 1 year posttransplantation.258
least a reduction in general immunosuppressive burden. Other investigators targeting subclinical rejection and fibro-
sis have observed some hints of positive effects from MSC
treatment.259 However, smaller cohorts of treated patients
Treg CELL THERAPY
have reported the possibility that MSC might aggravate
Treg have been infused into bone marrow transplant recipients kidney injury under certain conditions. In general, the het-
with the objective of limiting GVHD. Expanded Treg were used erogeneity in indications, clinical protocols, cell infusion
to treat two patients who developed GVHD after bone marrow timing, and tissue source of MSC in these trials has made it
transplantation with clinical improvement demonstrated in very difficult to draw any conclusions at this early stage of
both patients.253 No safety concerns were reported from two- clinical testing.
dose escalation studies when ex vivo expanded cord blood or
donor-derived Treg were infused into adult patients receiv- Acknowledgments
ing hematopoietic stem cell transplantation (HSCT).254,255 The work from the authors’ own laboratories described in
Moreover, in the second study GVHD was prevented in the this review was supported by grants from The Wellcome
absence of any immunosuppressive treatment regimen, and Trust, Medical Research Council, British Heart Founda-
lymphoid reconstitution occurred with improved immunity tion, The Deutsche Forschungsgemeinschaft, and The
to opportunistic pathogens, and no loss of the graft-versus- European Union ONE Study, Optistem, TRIAD, AFACTT
leukemia effect.255 In organ transplantation, The ONE Study, (European Union COST action – BM1305), and BioDRIM
a multicenter Phase I/II study funded by the European Union networks.
FP7 program, is investigating the safety of infusion of ex vivo
expanded polyclonal Treg and donor-reactive Treg into kidney References
transplant recipients (www.onestudy.org). Although regula- 1. Billingham RE, Medawar PB. Technique of free skin grafting in
tory immune cell therapy in organ transplantation is still in its mammals. J Exp Biol 1951;28:385–91.
2. Owen RD. Immunogenetic consequences of vascular anastomoses
very initial stages, The ONE Study consortium is expected to between bovine twins. Science 1945;102:400–1.
provide early results at least with regard to the general safety of 3. Billingham RE, Lampkin GH, Medawar PB, Williams HLI. Tolerance
various cellular therapies in kidney transplantation, as men- to homografts, twin diagnosis, and the freemarting condition in cat-
tioned later. tle. Heredity 1952;6:201–12.
4. Wood KJ, Goto R. Mechanisms of rejection: current perspectives.
Transplantation 2012;93(1):1–10. Available online at: https://doi.
Mreg CELL THERAPY org/10.1097/TP.1090b1013e31823cab31844.
5. Martínez-Llordella M, Puig-Pey I, Orlando G, et al. Multiparameter
Regulatory macrophages (Mreg) produced from blood leu- immune profiling of operational tolerance in liver transplantation.
kocytes of organ donors have been administered intrave- Am J Transplant 2007;7(2):309–19.
6. Takatsuki M, Uemoto S, Inomata Y, et al. Weaning of immunosup-
nously to two living donor kidney transplant recipients with pression in living donor liver transplant recipients. Transplantation
no deleterious effect on graft function, enabling tacrolimus 2001;72:449–54.
immunosuppressive therapy to be reduced within the first 7. Feng S, Ekong UD, Lobritto SJ, et al. Complete immunosuppres-
24 weeks after transplantation.111 A follow-up study using sion withdrawal and subsequent allograft function among pedi-
Mreg in kidney transplant recipients is currently being per- atric recipients of parental living donor liver transplants. JAMA
2012;307(3):283–93.
formed as part of The ONE Study. 8. Sanchez-Fueyo A. Strategies for minimizing immunosuppression:
state of the art. Liver Transpl 2016;22(S1):68–70.
9. Whitehouse G, Gray E, Mastoridis S, et al. IL-2 therapy restores regu-
TolDC THERAPY latory T-cell dysfunction induced by calcineurin inhibitors. Proc
Natl Acad Sci USA 2017;114(27):7083–8.
Also as part of The ONE Study, tolDC are being tested 10. Grossi PA, Costa AN, Fehily D, et al. Infections and organ transplan-
for their safety in living donor kidney transplant recipi- tation: new challenges for prevention and treatment—a colloquium.
ents. In anticipation of these results, preclinical studies in Transplantation 2012;93:S4–39.
21 • Approaches to the Induction of Tolerance 349

11. Webster AC, Craig JC, Simpson JM, Jones MP, Chapman JR. Identi- pared with a nonmyeloablative conditioning regime. Transplanta-
fying high risk groups and quantifying absolute risk of cancer after tion 1998;66:96–102.
kidney transplantation: a cohort study of 15183 recipients. Am J 36. Kawai T, Cosimi AB, Spitzer TR, et al. HLA-mismatched renal trans-
Transplant 2007;7(9):2140–51. plantation without maintenance immunosuppression. N Engl J Med
12. Hall EC, Pfeiffer RM, Segev DL, Engels EA. Cumulative incidence of can- 2008;358(4):353–61.
cer after solid organ transplantation. Cancer 2013;119(12):2300–8. 37. Leventhal J, Abecassis M, Miller J, et al. Chimerism and tolerance
13. Geissler EK. Post-transplantation malignancies: here today, gone without GVHD or engraftment syndrome in HLA-mismatched com-
tomorrow? Nat Rev Clin Oncol 2015;12(12):705–17. bined kidney and hematopoietic stem cell transplantation. Sci Transl
14. Steinman RM, Witmer MD. Lymphoid dendritic cells are potent stim- Med 2012;4(124):124–8.
ulators of the primary mixed leukocyte reaction in mice. Proc Natl 38. Webb SR, Sprent J. Induction of neonatal tolerance to Mlsa antigens
Acad Sci USA 1978;75(10):5132–6. by CD8+ T cells. Science 1990;248(4963):1643–6.
15. Mühlberger I, Perco P, Fechete R, Mayer B, Oberbauer R. Biomark- 39. Bemelman F, Honey K, Adams E, Cobbold S, Waldmann H. Bone
ers in renal transplantation ischemia reperfusion injury. Trans- marrow transplantation induces either clonal deletion or infectious
plantation 2009;88(3S):S14–9. Available online at: https://doi. tolerance depending on the dose. J Immunol 1998;160:2645–8.
org/10.1097/TP.1090b1013e3181af1065b1095. 40. Kearney E, Pape K, Loh D, Jenkins M. Visualisation of peptide-specific
16. Lu L, Zhou H, Ni M, et al. Innate immune regulations and liver isch- T cell immunity and peripheral tolerance induction in vivo. Immu-
emia-reperfusion injury. Transplantation 2016;100(12):2601–10. nity 1994;1:327–39.
17. Takeuchi O, Akira S. Pattern recognition receptors and inflamma- 41. Krammer PH, Arnold R, Lavrik IN. Life and death in peripheral T
tion. Cell 2010;140(6):805–20. cells. Nat Rev Immunol 2007;7(7):532–42.
18. Land WG, Agostinis P, Gasser S, Garg AD, Linkermann A. Trans- 42. Wells A, Li X, Li Y, et al. Requirement for T cell apoptosis in the induc-
plantation and damage-associated molecular patterns (DAMPs). Am tion of peripheral transplantation tolerance. Nat Med 1999;5:1303–7.
J Transplant 2016;16(12):3338–61. 43. Bellgrau D, Duke R. Apoptosis and CD95 ligand in immune privi-
19. Ogura Y, Sutterwala FS, Flavell RA. The inflammasome: first line of leged sites. Int Rev Immunol 1999;18:547–62.
the immune response to cell stress. Cell 2006;126(4):659–62. 44. George J, Sweeney S, Kirklin J, Simpson E, Goldstein D, Thomas J. An
20. Anglicheau D, Muthukumar T, Suthanthiran M. MicroRNAs: essential role for Fas ligand in transplantation tolerance induced by
small RNAs with big effects. Transplantation 2010;90(2):105–12. donor bone marrow. Nat Med 1998;4:333–5.
Available online at: https://doi.org/110.1097/TP.1090b1013e318 45. Turvey S, Gonzalez-Nicolini V, Kingsley C, et al. Fas ligand trans-
1e1913c1092. fected myoblasts and islet cell transplantation. Transplantation
21. Larsen C, Morris P, Austyn J. Migration of dendritic leukocytes form 2000;69:1972–6.
cardiac allografts into host spleens: a novel pathway for initiation of 46. Walunas T, Lenschow D, Bakker C, et al. CTLA-4 can function as a
rejection. J Exper Med 1990;171:307–14. negative regulator of T cell activation. Immunity 1994;1:405–13.
22. van Kooten C, Lombardi G, Gelderman KA, et al. Dendritic cells as 47. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance
a tool to induce transplantation tolerance: obstacles and opportuni- and autoimmunity. Immunol Rev 2010;236:219–42.
ties. Transplantation 2011;91(1):2–7. Available online at: https:// 48. Haynes R, Harden P, Judge P, et al. Alemtuzumab-based induc-
doi.org/10.1097/TP.1090b1013e31820263b31820263. tion treatment versus basiliximab-based induction treatment in
23. Marino J, Paster J, Benichou G. Allorecognition by T lymphocytes kidney transplantation (the 3C Study): a randomised trial. Lancet
and allograft rejection. Front Immunol 2016;7(582). 2014;384(9955):1684–90.
24. Afzali B, Lombardi G, Lechler RI. Pathways of major histocom- 49. West L, Pollock-Barziv S, Dipchand A, et al. ABO-incompatible heart
patibility complex allorecognition. Curr Opin Organ Trans- transplantation in infants. N Engl J Med 2001;344:793–800.
plant 2008;13(4):438–44. Available online at: https://doi. 50. Fan X, Ang A, Pollock-Barziv S, et al. Donor-specific B-cell toler-
org/410.1097/MOT.1090b1013e328309ee328331. ance after ABO-incompatible infant heart transplantation. Nat Med
25. Fooksman DR, Vardhana S, Vasiliver-Shamis G, et al. Functional 2004;10:1227–33.
anatomy of T cell activation and synapse formation. Annu Rev 51. Sykes M, Sachs D. Mixed chimerism. Philos Trans R Soc Lond B Biol
Immunol 2010;28:79–105. Sci 2001;356:707–26.
26. Kinnear G, Jones ND, Wood KJ. Costimulation blockade: cur- 52. Billingham RE, Brent L, Medawar PB. Quantitative studies on tissue
rent perspectives and implications for therapy. Transplantation transplantation immunity. III. Actively acquired tolerance. Philos
2013;95(4):527–35. Trans R Soc Lond B Biol Sci 1956;239:357–412.
27. O’Garra A, Vieira P. Regulatory T cells and mechanisms of immune 53. Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of
system control. Nat Med 2004;10(8):801–5. foreign cells. Nature 1953;172:603–6.
28. Wood KJ, Bushell A, Hester J. Regulatory immune cells in transplan- 54. Wood KJ, Bushell AR, Jones ND. The discovery of immunological
tation. Nat Rev Immunol 2012;12(6):417–30. tolerance: now more than just a laboratory solution. J Immunol
29. Bushell A, Morris P, Wood K. Anti-CD4 antibody combined with 2010;184(1):3–4.
random blood transfusion leads to authentic transplantation tol- 55. Gershon R, Kondo K. Infectious immunological tolerance. Immunol-
erance in the mouse: a protocol with significant clinical potential. ogy 1971;21:903–14.
Transplantation 1994;58:133–40. 56. Stolp J, Turka LA, Wood KJ. B cells with immune-regulating function
30. Hamano K, Rawsthorne M, Bushell A, Morris P, Wood K. Evidence in transplantation. Nat Rev Nephrol 2014;10:389.
that the continued presence of the organ graft and not peripheral 57. Broichhausen C, Riquelme P, Geissler EK, Hutchinson JA. Regula-
donor microchimerism is essential for the maintenance of toler- tory macrophages as therapeutic targets and therapeutic agents
ance to alloantigen in anti-CD4 treated recipients. Transplantation in solid organ transplantation. Curr Opin Organ Transplant
1996;62:856–60. 2012;17(4):332–42.
31. Scully R, Qin S, Cobbold S, Waldmann H. Mechanisms in CD4 58. Wood KJ, Sakaguchi S. Regulatory T cells in transplantation toler-
antibody-mediated transplantation tolerance: kinetics of induction, ance. Nat Immunol Rev 2003;3:199–210.
antigen dependency and role of regulatory T cells. Eur J Immunol 59. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regu-
1994;24:2383–92. latory T cells in the human immune system. Nat Rev Immunol
32. Khan A, Tomita Y, Sykes M. Thymic dependence of loss of tolerance 2010;10(7):490–500.
in mixed allogeneic bone marrow chimeras after depletion of donor 60. Reibke R, Garbi N, Ganss R, Hammerling GJ, Arnold B, Oelert T.
antigen. Transplantation 1996;62:380–7. CD8+ regulatory T cells generated by neonatal recognition of periph-
33. Zuber J, Sykes M. Mechanisms of mixed chimerism-based transplant eral self-antigen. Proc Natl Acad Sci USA 2006;103(41):15142–7.
tolerance. Trends Immunol 2017;38(11):829–43. 61. Vlad G, Suciu-Foca N. Resurgence or emergence of CD8+ Ts. Hum
34. Spitzer T, Delmonico F, Tolkoff-Rubin N, et al. Combined histocom- Immunol 2008;69(11):679–80.
patibility leukocyte antigen-matched donor bone marrow and renal 62. Li XC. The significance of non-T-cell pathways in graft rejection: implica-
transplantation for multiple myeloma with end stage renal disease: tions for transplant tolerance. Transplantation 2010;90(10):1043–7.
the induction of allograft tolerance through mixed lymphohemato- Available online at: https://doi.org/1010.1097/TP.1040b1013e318
poietic chimerism. Transplantation 1999;68:480–4. 1efcfe1049.
35. Manilay J, Pearson D, Sergio J, Swenson K, Sykes M. Intrathymic 63. Thomson CW, Lee BP, Zhang L. Double-negative regulatory T cells:
deletion of alloreactive T cells in mixed bone marrow chimeras pre- non-conventional regulators. Immunol Res 2006;35(1-2):163–78.
350 Kidney Transplantation: Principles and Practice

64. Monteiro M, Almeida CF, Caridade M, et al. Identification of regu- 88. Graca L, Cobbold SP, Waldmann H. Identification of regulatory T
latory Foxp3+ invariant NKT cells induced by TGF-beta. J Immunol cells in tolerated allografts. J Exper Med 2002;195(12):1641–6.
2010;185(4):2157–63. 89. Kendal AR, Waldmann H. Infectious tolerance: therapeutic poten-
65. Hayday A, Tigelaar R. Immunoregulation in the tissues by gamma­ tial. Curr Opin Immunol 2010;22(5):560–5.
delta T cells. Nat Rev Immunol 2003;3(3):233–42. 90. Rosenblum MD, Gratz IK, Paw JS, Lee K, Marshak-Rothstein A,
66. Bennett CL, Christie F, Ramsdell F, et al. The immune dysregula- Abbas AK. Response to self antigen imprints regulatory memory in
tion, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is tissues. Nature 2011;480(7378):538–42.
caused by mutations of FOXP3. Nat Genet 2001;27:20–1. 91. Bushell A, Wood K. GITR ligation blocks allograft protection by
67. Kobayashi I, Shiari R, Yamada M, et al. Novel mutations of FOXP3 induced CD25+CD4+ regulatory T cells without enhancing effector
in two Japanese patients with immune dysregulation, polyendo- T-cell function. Am J Transplant 2007;7(4):759–68.
crinopathy, enteropathy, X linked syndrome (IPEX). J Med Genet 92. Josien R, Douillard P, Guillot C, et al. A critical role for transforming
2001;38(12):874–6. growth factor beta in donor transfusion induced allograft tolerance.
68. Bacchetta R, Passerini L, Gambineri E, et al. Defective regulatory and J Clin Invest 1998;102:1920–6.
effector T cell functions in patients with FOXP3 mutations. J Clin 93. Madsen JC, Superina RA, Wood KJ, Morris PJ. Immunological unre-
Invest 2006;116(6):1713–22. sponsiveness induced by recipient cells transfected with donor MHC
69. Khattri R, Cox T, Yasayko S, Ramsdell F. An essential role for Scurfin genes. Nature 1988;332:161–4.
in CD4+CD25+ T regulatory cells. Nat Immunol 2003;4:304–6. 94. Davies JD, Leong LY, Mellor A, Cobbold SP, Waldmann H. T cell sup-
70. Josefowicz SZ, Rudensky A. Control of regulatory T cell lineage com- pression in transplantation tolerance through linked recognition. J
mitment and maintenance. Immunity 2009;30(5):616–25. Immunol 1996;156:3602–7.
71. Wood KJ, Bushell A, Jones ND. Immunologic unresponsiveness 95. Mauri C, Blair PA. Regulatory B cells in autoimmunity: develop-
to alloantigen in vivo: a role for regulatory T cells. Immunol Rev ments and controversies. Nature Rev Rheumatol 2010;6(11):
2011;241(1):119–32. 636–43.
72. Qin S, Cobbold SP, Pope H, et al. “Infectious” transplantation toler- 96. Ding Q, Yeung M, Camirand G, et al. Regulatory B cells are identified
ance. Science 1993;259:974–7. by expression of TIM-1 and can be induced through TIM-1 ligation
73. Hall BM, Jelbart ME, Dorsch SE. Suppressor T cells in rats with pro- to promote tolerance in mice. J Clin Invest 2011;121(9):3645–56.
longed cardiac allograft survival after treatment with cyclosporine. 97. Le Texier L, Thebault P, Lavault A, et al. Long-term allograft toler-
Transplantation 1984;37(6):595–600. ance is characterized by the accumulation of B cells exhibiting an
74. Hall B, Jelbart M, Gurley K, Dorsch S. Specific unresponsiveness in inhibited profile. Am J Transplant 2011;11(3):429–38.
rats with prolonged cardiac allograft survival after treatment with 98. Sagoo P, Perucha E, Sawitzki B, et al. Development of a cross-plat-
cyclosporine: mediation of specific suppression by T helper/inducer form biomarker signature to detect renal transplant tolerance in
cells. J Exper Med 1985;162:1683–94. humans. J Clin Invest 2010;120(6):1848–61.
75. Hall B, Pearce N, Gurley K, Dorsch S. Specific unresponsiveness in 99. Newell KA, Asare A, Kirk AD, et al. Identification of a B cell signature
rats with prolonged cardiac allograft survival after treatment with associated with renal transplant tolerance in humans. J Clin Invest
cyclosporine. III. Further characterisation of the CD4+ suppressor 2010;120(6):1836–47.
cell and its mechanisms of action. J Exper Med 1990;171:141–57. 100. Pallier A, Hillion S, Danger R, et al. Patients with drug-free long-term
76. Quigley RL, Wood KJ, Morris J. Mediation of antigen-induced sup- graft function display increased numbers of peripheral B cells with a
pression of renal allograft rejection by a CD4 (W3/25+) T cell. Trans- memory and inhibitory phenotype. Kidney Int 2010;78(5):503–13.
plantation 1989;47:684–8. 101. Rebollo-Mesa I, Nova-Lamperti E, et al. Biomarkers of toler-
77. Quigley RL, Wood KJ, Morris PJ. Transfusion induces blood donor- ance in kidney transplantation: are we predicting tolerance or
specific suppressor cells. J Immunol 1989;142(2):463. response to immunosuppressive treatment? Am J Transplant
78. Hara M, Kingsley C, Niimi M, et al. IL-10 is required for regula- 2016;16(12):3443–57.
tory T cells to mediate tolerance to alloantigens in vivo. J Immunol 102. Asare A, Kanaparthi S, Lim N, et al. B cell receptor genes associ-
2001;166:3789–96. ated with tolerance identify a cohort of immunosuppressed patients
79. Yang J, Brook M, Carvalho-Gaspar M, et al. Allograft rejection medi- with improved renal allograft graft function. Am J Transplant
ated by memory T cells is resistant to regulation. Proc Natl Acad Sci 2017;17(10):2627–39.
USA 2007;104:19954–9. 103. Bottomley MJ, Chen M, Fuggle S, Harden PN, Wood KJ. Application
80. Graca L, Thompson S, Lin C-Y, Adams E, Cobbold SP, Wald- of operational tolerance signatures are limited by variability and
mann H. Both CD4(+)CD25(+) and CD4(+)CD25(-) regulatory type of immunosuppression in renal transplant recipients: a cross-
cells mediate dominant transplantation tolerance. J Immunol sectional study. Transplant Direct 2017;3(1):e125.
2002;168(11):5558–65. 104. Murray PJ, Wynn TA. Protective and pathogenic functions of mac-
81. Francis RS, Feng G, Tha-In T, Lyons I, Wood KJ, Bushell A. Induction rophage subsets. Nat Rev Immunol 2011;11:723.
of transplantation tolerance converts potential effector T cells into graft 105. Riquelme P, Geissler EK, Hutchinson JA. Alternative approaches to
protective regulatory T cells. Eur J Immunol 2011;41(3):726–38. myeloid suppressor cell therapy in transplantation: comparing regu-
82. Nadig SN, Wieckiewicz J, Wu DC, et al. In vivo prevention of trans- latory macrophages to tolerogenic DCs and MDSCs. Transplant Res
plant arteriosclerosis by ex vivo-expanded human regulatory T cells. 2012;1(1):17.
Nat Med 2010;16(7):809–13. 106. Martinez FO, Helming L, Gordon S. Alternative activation of macro-
83. Hester J, Schiopu A, Nadig SN, Wood KJ. Low-dose rapamy- phages: an immunologic functional perspective. Annu Rev Immu-
cin treatment increases the ability of human regulatory T cells nol 2009;27:451–83.
to inhibit transplant arteriosclerosis in vivo. Am J Transplant 107. Hu MS, Walmsley GG, Barnes LA, et al. Delivery of monocyte lin-
2012;12(8):2008–16. eage cells in a biomimetic scaffold enhances tissue repair. JCI Insight
84. Kingsley CI, Karim M, Bushell AR, Wood KJ. CD25+CD4+ regulatory 2017;2(19).
T cells prevent graft rejection: CTLA-4- and IL-10-dependent immu- 108. Tiemessen MM, Jagger AL, Evans HG, van Herwijnen MJ, John S,
noregulation of alloresponses. J Immunol 2002;168(3):1080–6. Taams LS. CD4+CD25+Foxp3+ regulatory T cells induce alternative
85. Tullius S, Nieminen M, Bechstein W, et al. Chronically rejected rat activation of human monocytes/macrophages. Proc Natl Acad Sci
kidney allografts induce donor-specific tolerance. Transplantation USA 2007;104(49):19446–51.
1997;64:158–61. 109. Wong SC, Puaux AL, Chittezhath M, et al. Macrophage polar-
86. Carvalho-Gaspar M, Jones ND, Luo S, Martin L, Brook MO, Wood ization to a unique phenotype driven by B cells. Eur J Immunol
KJ. Location and time-dependent control of rejection by regulatory 2010;40(8):2296–307.
T cells culminates in a failure to generate memory T cells. J Immunol 110. Fleming BD, Mosser DM. Regulatory macrophages: setting the
2008;180(10):6640–8. threshold for therapy. Eur J Immunol 2011;41(9):2498–502.
87. Issa F, Hester J, Milward K, Wood KJ. Homing of regulatory T cells to 111. Hutchinson JA, Riquelme P, Sawitzki B, et al. Cutting edge: immu-
human skin is important for the prevention of alloimmune-mediated nological consequences and trafficking of human regulatory mac-
pathology in an in vivo cellular therapy model. PLoS One rophages administered to renal transplant recipients. J Immunol
2012;7(12):e53331. 2011;187(5):2072–8.
21 • Approaches to the Induction of Tolerance 351

112. Riquelme P, Amodio G, Macedo C, et al. DHRS9 is a stable marker 137. Yamada A, Chandraker A, Laufer TM, Gerth AJ, Sayegh MH,
of human regulatory macrophages. Transplantation 2017; Auchincloss Jr H. Cutting edge: recipient MHC Class II expression is
101(11):2731–8. required to achieve long-term survival of murine cardiac allografts
113. Hashimoto D, Chow A, Greter M, et al. Pretransplant CSF-1 after costimulatory blockade. J Immunol 2001;167(10):5522–6.
therapy expands recipient macrophages and ameliorates GVHD 138. De Wilde V, Van Rompaey N, Hill M, et al. Endotoxin-induced
after allogeneic hematopoietic cell transplantation. J Exper Med myeloid-derived suppressor cells inhibit alloimmune responses via
2011;208(5):1069–82. heme oxygenase-1. Am J Transplant 2009;9(9):2034–47.
114. Morelli AE, Thomson AW. Tolerogenic dendritic cells and the quest 139. Ribechini E, Hutchinson JA, Hergovits S, et al. Novel GM-CSF signals
for transplant tolerance. Nat Rev Immunol 2007;7:610. via IFN-gR/IRF-1 and AKT/mTOR license monocytes for suppressor
115. Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic function. Blood Advances 2017;1(14):947–60.
cells. Annu Rev Immunol 2003;21:685–711. 140. English K, French A, Wood KJ. Mesenchymal stromal cells: facilita-
116. Thomson AW. Tolerogenic dendritic cells: all present and correct? tors of successful transplantation? Cell Stem Cell 2010;7(4):431–42.
Am J Transplant 2010;10(2):214–9. 141. Ding Y, Xu D, Feng G, Bushell A, MR J, Wood KJ. Mesenchymal stem
117. Lu L, McCaslin D, Starzl TE, Thomson AW. Bone marrow-derived cells prevent the rejection of fully allogeneic islet grafts by the immu-
dendritic cell progenitors (NLDC 145+, MHC class II+, B7-1dim, nosuppressive activity of matrix metalloproteinase-2 and -9. Diabe-
B7-2−) induce alloantigen-specific hyporesponsiveness in murine T tes 2009; 58(8):1797–806.
lymphocytes. Transplantation 1995;60(12):1539–45. 142. Lee RH, Pulin AA, Seo MJ, et al. Intravenous hMSCs improve myo-
118. Lu L, Li W, Fu F, et al. Blockade of the CD40-CD40 ligand pathway cardial infarction in mice because cells embolized in lung are acti-
potentiates the capacity of donor-derived dendritic cell progenitors vated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell
to induce long-term cardiac allograft survival. Transplantation 2009;5(1):54–63.
1997;64:1808–15. 143. Galleu A, Riffo-Vasquez Y, Trento C, et al. Apoptosis in mesenchymal
119. Swiecki M, Colonna M. Unraveling the functions of plasmacytoid stromal cells induces in vivo recipient-mediated immunomodula-
dendritic cells during viral infections, autoimmunity, and tolerance. tion. Sci Transl Med 2017;9(416).
Immunol Rev 2010;234(1):142–62. 144. Ren G, Zhang L, Zhao X, et al. Mesenchymal stem cell-mediated
120. Ochando JC, Homma C, Yang Y, et al. Alloantigen-presenting plas- immunosuppression occurs via concerted action of chemokines and
macytoid dendritic cells mediate tolerance to vascularized grafts. Nat nitric oxide. Cell Stem Cell 2008;2(2):141–50.
Immunol 2006;7:652. 145. Bai M, Zhang L, Fu B, et al. IL-17A improves the efficacy of mesen-
121. Gilliet M, Liu Y-J. Generation of human CD8 T regulatory cells by chymal stem cells in ischemic-reperfusion renal injury by increas-
CD40 ligand-activated plasmacytoid dendritic cells. J Exper Med ing Treg percentages by the COX-2/PGE2 pathway. Kidney Int
2002;195(6):695–704. 2017:814–25.
122. Levings MK, Gregori S, Tresoldi E, Cazzaniga S, Bonini C, Roncarolo 146. Krampera M, Cosmi L, Angeli R, et al. Role for interferon-gamma in
MG. Differentiation of Tr1 cells by immature dendritic cells requires the immunomodulatory activity of human bone marrow mesenchy-
IL-10 but not CD25+CD4+ Tr cells. Blood 2005;105(3):1162–9. mal stem cells. Stem Cells (Dayton, Ohio) 2006;24(2):386–98.
123. Darrasse-Jeze G, Deroubaix S, Mouquet H, et al. Feedback control of 147. Polchert D, Sobinsky J, Douglas G, et al. IFN-gamma activation of
regulatory T cell homeostasis by dendritic cells in vivo. J Exper Med mesenchymal stem cells for treatment and prevention of graft versus
2009;206(9):1853–62. host disease. Eur J Immunol 2008;38(6):1745–55.
124. Chang CC, Ciubotariu R, Manavalan JS, et al. Tolerization of den- 148. Sheng H, Wang Y, Jin Y, et al. A critical role of IFN gamma in prim-
dritic cells by TS cells: the crucial role of inhibitory receptors ILT3 and ing MSC-mediated suppression of T cell proliferation through up-
ILT4. Nat Immunol 2002;3(3):237–43. regulation of B7-H1. Cell Res 2008;18(8):846–57.
125. Vlad G, Cortesini R, Suciu-Foca N. CD8+ T suppressor cells and the 149. Asari S, Itakura S, Ferreri K, et al. Mesenchymal stem cells suppress
ILT3 master switch. Hum Immunol 2008;69(11):681–6. B-cell terminal differentiation. Exper Hematol 2009;37(5):604–15.
126. Mazariegos GV, Zahorchak AF, Reyes J, Chapman H, Zeevi A, 150. Spaggiari GM, Abdelrazik H, Becchetti F, Moretta L. MSCs inhibit
Thomson AW. Dendritic cell subset ratio in tolerant, weaning and monocyte-derived DC maturation and function by selectively inter-
non-­tolerant liver recipients is not affected by extent of immunosup- fering with the generation of immature DCs: central role of MSC-
pression. Am J Transplant 2005;5(2):314–22. derived prostaglandin E2. Blood 2009;113(26):6576–83.
127. Tokita D, Sumpter TL, Raimondi G, et al. Poor allostimulatory func- 151. Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari
tion of liver plasmacytoid DC is associated with pro-apoptotic activ- MC, Moretta L. Mesenchymal stem cells inhibit natural killer-
ity, dependent on regulatory T cells. J Hepatol 2008;49(6):1008–18. cell proliferation, cytotoxicity, and cytokine production: role
128. Matta BM, Castellaneta A, Thomson AW. Tolerogenic plasmacytoid of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood
DC. Eur J Immunol 2010;40(10):2667–76. 2008;111(3):1327–33.
129. Ezzelarab MB, Zahorchak AF, Lu L, et al. Regulatory dendritic cell 152. Nemeth K, Leelahavanichkul A, Yuen PS, et al. Bone marrow stro-
infusion prolongs kidney allograft survival in nonhuman primates. mal cells attenuate sepsis via prostaglandin E2-dependent repro-
Am J Transplant 2013;13(8):1989–2005. gramming of host macrophages to increase their interleukin-10
130. Ezzelarab MB, Raich-Regue D, Lu L, et al. Renal allograft survival in production. Nat Med 2009;15(1):42–9.
nonhuman primates infused with donor antigen-pulsed autologous 153. Carty F, Mahon BP, English K. The influence of macrophages on
regulatory dendritic cells. Am J Transplant 2017;17(6):1476–89. mesenchymal stromal cell therapy: passive or aggressive agents?
131. Boros P, Ochando JC, Chen SH, Bromberg JS. Myeloid-derived sup- Clin Exper Immunol 2017;188(1):1–11.
pressor cells: natural regulators for transplant tolerance. Hum 154. Francois M, Romieu-Mourez R, Li M, Galipeau J. Human MSC
Immunol 2010;71(11):1061–6. suppression correlates with cytokine induction of indoleamine
132. Haile LA, von Wasielewski R, Gamrekelashvili J, et al. Myeloid- 2,3-­dioxygenase and bystander M2 macrophage differentiation. Mol
derived suppressor cells in inflammatory bowel disease: a new immu- Ther 2012;20(1):187–95.
noregulatory pathway. Gastroenterology 2008;135(3):871–81. 155. Reading JL, Vaes B, Hull C, et al. Suppression of IL-7-dependent effec-
133. Gallina G, Dolcetti L, Serafini P, et al. Tumors induce a subset of tor T-cell expansion by multipotent adult progenitor cells and PGE2.
inflammatory monocytes with immunosuppressive activity on CD8+ Mol Ther 2015;23(11):1783–93.
T cells. J Clin Invest 2006;116(10):2777–90. 156. Chinnadurai R, Copland IB, Ng S, et al. Mesenchymal stromal cells
134. Garcia MR, Ledgerwood L, Yang Y, et al. Monocytic suppressive cells derived from Crohn’s patients deploy indoleamine 2,3-dioxygenase-
mediate cardiovascular transplantation tolerance in mice. J Clin mediated immune suppression, independent of autophagy. Mol Ther
Invest 2010;120(7):2486–96. 2015;23(7):1248–61.
135. Chou HS, Hsieh CC, Charles R, et al. Myeloid-derived suppressor cells 157. Duffy MM, Pindjakova J, Hanley SA, et al. Mesenchymal stem cell
protect islet transplants by B7-H1 mediated enhancement of T regu- inhibition of T-helper 17 cell- differentiation is triggered by cell-cell
latory cells. Transplantation 2012;93(3):272–82. contact and mediated by prostaglandin E2 via the EP4 receptor. Eur
136. Dugast AS, Haudebourg T, Coulon F, et al. Myeloid-derived J Immunol 2011;41(10):2840–51.
suppressor cells accumulate in kidney allograft tolerance 158. Luz-Crawford P, Noel D, Fernandez X, et al. Mesenchymal stem cells
and specifically suppress effector T cell expansion. J Immunol repress Th17 molecular program through the PD-1 pathway. PLoS
2008;180(12):7898–906. One 2012;7(9):e45272.
352 Kidney Transplantation: Principles and Practice

159. Ding Y, Bushell A, Wood KJ. Mesenchymal stem-cell immunosup- 183. Morris H, DeWolf S, Robins H, et al. Tracking donor-reactive T cells:
pressive capabilities: therapeutic implications in islet transplanta- evidence for clonal deletion in tolerant kidney transplant patients.
tion. Transplantation 2010;89(3):270–3. Sci Transl Med 2015;7(272):272–10.
160. Lu C, Li XY, Hu Y, Rowe RG, Weiss SJ. MT1-MMP controls human 184. Sprangers B, DeWolf S, Savage TM, et al. Origin of enriched regu-
mesenchymal stem cell trafficking and differentiation. Blood latory T cells in patients receiving combined kidney-bone marrow
2010;115(2):221–9. transplantation to induce transplantation tolerance. Am J Trans-
161. English K, Ryan JM, Tobin L, Murphy MJ, Barry FP, Mahon BP. Cell plant 2017;17(8):2020–32.
contact, prostaglandin E2 and transforming growth factor beta 1 185. Myburgh JA, Smit JA, Stark JH, Browde S. Total lymphoid irradiation
play non-redundant roles in human mesenchymal stem cell induc- in kidney and liver transplantation in the baboon: prolonged graft
tion of CD4+CD25High forkhead box P3+ regulatory T cells. Clin Exper survival and alteration in cell subsets with low cumulative dose regi-
Immunol 2009;156(1):149–60. mens. J Immunol 1984;132:1019–25.
162. Casiraghi F, Azzollini N, Cassis P, et al. Pretransplant infusion of mes- 186. Slavin S, Strober S, Fuks Z, Kaplan HS. Induction of specific tissue
enchymal stem cells prolongs the survival of a semiallogeneic heart transplantation tolerance using fractionated total lymphoid irradia-
transplant through the generation of regulatory T cells. J Immunol tion in adult mice: long-term survival of allogeneic bone marrow and
2008;181(6):3933–46. skin grafts. J Exper Med 1977;146:34–48.
163. Chen L, Tredget EE, Wu PY, Wu Y. Paracrine factors of mesenchy- 187. Strober S, Dhillon M, Schubert M, et al. Acquired immune tolerance
mal stem cells recruit macrophages and endothelial lineage cells and to cadaveric renal allografts: a study of three patients treated with
enhance wound healing. PLoS One 2008;3(4):e1886. total lymphoid irradiation. N Engl J Med 1989;321:28–33.
164. Hsiao ST, Asgari A, Lokmic Z, et al. Comparative analysis of para- 188. Sachs DH, Sykes M, Kawai T, Cosimi AB. Immuno-intervention for
crine factor expression in human adult mesenchymal stem cells the induction of transplantation tolerance through mixed chime-
derived from bone marrow, adipose, and dermal tissue. Stem Cells rism. Semin Immunol 2011;23(3):165–73.
Dev 2012;21(12):2189–203. 189. Kawai T, Cosimi A, Colvin R, et al. Mixed allogeneic chimerism and
165. Ge W, Jiang J, Baroja ML, et al. Infusion of mesenchymal stem cells and renal allograft tolerance in cynomolgus monkeys. Transplantation
rapamycin synergize to attenuate alloimmune responses and promote 1995;59:256–62.
cardiac allograft tolerance. Am J Transplant 2009;9(8):1760–72. 190. Monaco AP. Chimerism in organ transplantation: conflicting experi-
166. Jacobsen N, Taaning E, Ladefoged J, Kristensen J, Pedersen F. Toler- ments and clinical observations. Transplantation 2003;75(Suppl.
ance to an HLA-B,DR disparate kidney allograft after bone marrow 9):13s–6s.
transplantation from the same donor. Lancet 1994;343:800. 191. Colson Y, Wren S, Schubert M, et al. A nonlethal conditioning
167. Sayegh M, Fine N, Smith J, Renneke H, Milford E, Tilney N. Immu- approach to achieve durable multilineage mixed chimerism and
nologic tolerance to renal allografts after bone marrow transplants tolerance across major, minor and hematopoietic histocompatibility
from the same donors. Ann Intern Med 1991;114:954. barriers. J Immunol 1995;155. 4197–88.
168. Sorof J, Koerper M, Portale A, Potter D, DeSantes K, Morton C. Renal 192. Ildstad ST, Leventhal J, Wen Y, Yolcu E. Facilitating cells: translation
transplantation without chronic immunosuppression after T cell of hematopoietic chimerism to achieve clinical tolerance. Chimerism
depleted HLA-mismatched bone marrow transplantation. Trans- 2015;6(1–2):33–9.
plantation 1995;59:1633–5. 193. Schwartz RH. A cell culture model for T lymphocyte clonal anergy.
169. Devlin J, Doherty D, Thomson L, et al. Defining the outcome of Science 1990;248:1349–56.
immunosuppression withdrawal after liver transplantation. Hepa- 194. Vincenti F, Larsen C, Durrbach A, et al. Costimulation blockade with
tology (Baltimore, MD) 1998;27(4):926–33. belatacept in renal transplantation. N Engl J Med 2005;353(8):770–81.
170. Owens ML, Maxwell J, Goodnight J, Wolcott MW. Discontinuance 195. Thompson C. Distinct roles for the costimulatory ligands B7-1 and
of immunosuppression in renal transplant patients. Arch Surg B7-2 in T helper cell differentiation? Cell 1995;81:879–982.
1975;110(12):1450–1. 196. Stamper CC, Zhang Y, Tobin JF, et al. Crystal structure of the B7-1/
171. Brouard S, Dupont A, Giral M, et al. Operationally tolerant and mini- CTLA-4 complex that inhibits human immune responses. Nature
mally immunosuppressed kidney recipients display strongly altered 2001;410(6828):608–11.
blood T-cell clonal regulation. Am J Transplant 2005;5(2):330–40. 197. Sharma P, Wagner K, Wolchok JD, Allison JP. Novel cancer immu-
172. Roussey-Kesler G, Giral M, Moreau A, et al. Clinical operational toler- notherapy agents with survival benefit: recent successes and next
ance after kidney transplantation. Am J Transplant 2006;6(4):736–46. steps. Nat Rev Cancer 2011;11(11):805–12.
173. Li Y, Koshiba T, Yoshizawa A, et al. Analyses of peripheral blood 198. Read S, Malmstrom V, Powrie F. Cytotoxic T lymphocyte associated
mononuclear cells in operational tolerance after pediatric living antigen 4 plays an essential role in the function of CD25+CD4+
donor liver transplantation. Am J Transplant 2004;4(12):2118–25. regulatory cells that control intestinal inflammation. J Exper Med
174. Bohne F, Martínez-Llordella M, et al. Intra-graft expression of 2000;192:295–302.
genes involved in iron homeostasis predicts the development of 199. Lenschow DJ, Zeng Y, Thistlethwaite JR, et al. Long-term survival
operational tolerance in human liver transplantation. J Clin Invest of xenogeneic pancreatic islet grafts induced by CTLA4lg [see com-
2012;122(1):368–82. ments]. Science 1992;257(5071):789–92.
175. Strober S, Slavin S, Gottlieb M, et al. Allograft tolerance after total 200. Larsen P, Elwood E, Alexander D, et al. Long-term acceptance of
lymphoid irradiation (TLI). Immunol Rev 1979;46:87–112. skin and cardiac allografts after blocking CD40 and CD28 pathways.
176. Scandling JD, Busque S, Dejbakhsh-Jones S, et al. Tolerance and chi- Nature 1996;381:434–8.
merism after renal and hematopoietic-cell transplantation. N Engl J 201. Pearson T, Alexander D, Hendrix R, et al. CTLA4-Ig plus bone marrow
Med 2008;358(4):362–8. induces long-term allograft survival and donor-specific unrespon-
177. Kawai T, Sachs DH, Sprangers B, et al. Long-term results in recipients siveness in the murine model. Transplantation 1996;61:997–1004.
of combined HLA-mismatched kidney and bone marrow transplan- 202. Kirk A, Harlan D, Armstrong N, et al. CTLA4-Ig and anti-CD40
tation without maintenance immunosuppression. Am J Transplant ligand prevent renal allograft rejection in primates. Proc Natl Acad
2014;14(7):1599–611. Sci USA 1997;94:8789–94.
178. Calne RY, Sells RA, Pena JR, et al. Induction of immunological toler- 203. Larsen CP, Pearson TC, Adams AB, et al. Rational development of
ance by porcine liver allografts. Nature 1969;223:472–6. LEA29Y (belatacept), a high-affinity variant of CTLA4-Ig with potent
179. Calne R, Davies H. Organ graft tolerance: the liver effect. Lancet immunosuppressive properties. Am J Transplant 2005;5(3):443–53.
1994;343(8889):67–8. 204. Suntharalingam G, Perry MR, Ward S, et al. Cytokine storm in a
180. Starzl T, Murase N, Thomson A, Demetris A. Liver transplants con- phase 1 trial of the anti-CD28 monoclonal antibody TGN1412.
tribute to their own success. Nat Med 1996;2:163–5. N Engl J Med 2006;355(10):1018–28.
181. Li Y, Zhao X, Cheng D, et al. The presence of Foxp3 expressing T cells 205. Zaitsu M, Issa F, Hester J, Vanhove B, Wood KJ. Selective block-
within grafts of tolerant human liver transplant recipients. Trans- ade of CD28 on human T cells facilitates regulation of alloimmune
plantation 2008;86(12):1837–43. Available online at: https://doi. responses. JCI Insight 2017;2(19).
org/1810.1097/TP.1830b1013e31818febc31814. 206. Poirier N, Azimzadeh AM, Zhang T, et al. Inducing CTLA-4-­
182. Martínez-Llordella M, Lozano JJ, Puig-Pey I, et al. Using transcrip- dependent immune regulation by selective CD28 blockade pro-
tional profiling to develop a diagnostic test of operational tolerance motes regulatory T cells in organ transplantation. Sci Transl Med
in liver transplant recipients. J Clin Invest 2008;118(8):2845–57. 2010;2(17):17ra10.
21 • Approaches to the Induction of Tolerance 353

207. Dillinger B, Ahmadi-Erber S, Soukup K, et al. CD28 blockade ex vivo possible role for the CD4 (L3T4) and CD11a (LFA-1) molecules in
induces alloantigen-specific immune tolerance but preserves T-cell self-non-self discrimination. Eur J Immunol 1988;18:1079–88.
pathogen reactivity. Front Immunol 2017;8:1152. 232. Group OMTS. A randomized clinical trial of OKT3 monoclonal anti-
208. Larsen C, Alexander D, Hollenbaugh D, et al. CD40-gp39 interac- body for acute rejection of cadaveric renal transplants. N Engl J Med
tions play a critical role during allograft rejection. Transplantation 1985;313(6):337–42.
1996;61:4–9. 233. Gaston RS, Deierhoi MH, Patterson T, et al. OKT3 first-dose reac-
209. Clarkson MR, Sayegh MH. T-Cell costimulatory pathways in allograft tion: association with T cell subsets and cytokine release. Kidney Int
rejection and tolerance. Transplantation 2005;80(5):555–63. 1991;39(1):141–8.
210. Andre P, Prasad KSS, Denis CV, et al. CD40L stabilizes arterial 234. Thistlethwaite Jr JR, Stuart JK, Mayes JT, et al. Complications and
thrombi by a beta3 integrin-dependent mechanism. Nat Med monitoring of OKT3 therapy. Am J Kidney Dis 1988;11(2):112–9.
2002;8(3):247–52. 235. Li B, Wang H, Dai J, et al. Construction and characterization of a
211. Larsen CP, Knechtle SJ, Adams A, Pearson T, Kirk AD. A new humanized anti-human CD3 monoclonal antibody 12F6 with effective
look at blockade of T-cell costimulation: a therapeutic strategy for immunoregulation functions. Immunology 2005;116(4):487–98.
long-term maintenance immunosuppression. Am J Transplant 236. Herold KC, Gitelman SE, Masharani U, et al. A single course of anti-
2006;6(5p1):876–83. CD3 monoclonal antibody hOKT3γ1(Ala-Ala) results in improve-
212. Larsen CP, Pearson TC. The CD40 pathway in allograft rejection, ment in C-peptide responses and clinical parameters for at least 2
acceptance, and tolerance. Curr Opin Immunol 1997;9:641–7. years after onset of Type 1 diabetes. Diabetes 2005;54(6):1763–9.
213. Noelle R, Roy M, Shepherd D, Stamenkovic I, Ledbetter J, Aruffo A. 237. Keymeulen B, Vandemeulebroucke E, Ziegler AG, et al. Insulin needs
A 39-kDa protein on activated helper T cells binds CD40 and trans- after CD3-antibody therapy in new-onset type 1 diabetes. N Engl J
duces the signal for cognate activation of B cells. Proc Natl Acad Sci Med 2005;352:2598–608.
USA 1992;89:6550–4. 238. Goto R, You S, Zaitsu M, Chatenoud L, Wood KJ. Delayed anti-CD3
214. Kawabe T, Naka T, Yoshida K, et al. The immune responses in CD40- therapy results in depletion of alloreactive T cells and the domi-
deficient mice: impaired immunoglobulin class switching and germi- nance of Foxp3+CD4+ graft infiltrating cells. Am J Transplant
nal centre formation. Immunity 1994;1:167–78. 2013;13(7):1655–64.
215. Xu J, Foy T, Laman J, et al. Mice deficient for the CD40 ligand. Immu- 239. Badell IR, Russell MC, Thompson PW, et al. LFA-1-specific ther-
nity 1994;1:423–31. apy prolongs allograft survival in rhesus macaques. J Clin Invest
216. Grewal I, Foellmer H, Grewal K, et al. Requirement for CD40 ligand 2010;120(12):4520–31.
in costimulation induction, T cell activation, and experimental aller- 240. Posselt AM, Bellin MD, Tavakol M, et al. Islet transplantation in Type
gic encephalomyelitis. Science 1996;274:1864–7. 1 diabetics using an immunosuppressive protocol based on the anti-
217. Gurunathan S, Irvine K, Wu C, et al. CD40 ligand/trimer DNA LFA-1 antibody Efalizumab. Am J Transplant 2010;10(8):1870–80.
enhances both humoral and cellular immune responses and induces 241. Pearson TC, Darby C, Bushell AR, West L, Morris PJ, Wood KJ.
protective immunity to infectious and tumor challenge. J Immunol The assessment of transplantation tolerance induced by anti-
1998;161:4563–71. CD4 monoclonal antibody in the murine model. Transplantation
218. Tomita Y, Satomi M, Bracamonte-Baran W, et al. Kinetics of allo- 1993;55:361–7.
antigen-specific regulatory CD4 T cell development and tissue distri- 242. Pearson TC, Madsen JC, Larsen C, Morris PJ, Wood KJ. Induction of
bution after donor-specific transfusion and costimulatory blockade. transplantation tolerance in the adult using donor antigen and anti-
Transplant Direct 2016;2(5):e73. CD4 monoclonal antibody. Transplantation 1992;54:475–83.
219. Parker D, Greiner D, Phillips N, et al. Survival of mouse pancre- 243. Bushell A, Jones E, Gallimore A, Wood KJ. The generation of
atic islet allografts in recipients treated with allogeneic small lym- CD25+CD4+ regulatory cells that prevent allograft rejection
phocytes and antibody to CD40 ligand. Proc Natl Acad Sci USA does not compromise immunity to a viral protein. J Immunol
1995;92:9560–4. 2005;174:3290–7.
220. Markees T, Phillips N, Noelle R, et al. Prolonged survival of mouse 244. Kendal AR, Chen Y, Regateiro FS, et al. Sustained suppression by
skin allografts in recipients treated with donor splenocytes and anti- Foxp3+ regulatory T cells is vital for infectious transplantation toler-
body to CD40 ligand. Transplantation 1997;64:329–35. ance. J Exper Med 2011;208(10):2043–53.
221. Kirk A, Burkly L, Batty D, et al. Treatment with humanised 245. Monaco AP, Wood ML, Russel PS. Studies on heterologous anti-lym-
monoclonal antibody against CD154 prevents acute renal allograft phocyte serum in mice: III. Immunologic tolerance and chimerism
rejection in nonhuman primates. Nat Med 1999;5:686–93. produced across the H-2 locus with adult thymectomy and anti-lym-
222. Kirk AD, Knechtle SJ, Sollinger HW, Vincenti F, Stecher S, Nadeau K. phocyte serum. Ann NY Acad Sci 1966;129:190–206.
Preliminary results of the use of humanized anti-CD154 in human 246. Watson C, Bradley J, Friend P, et al. Alemtuzumab (CAMPATH 1H)
renal allotransplantation. Am J Transplant 2001;1:S191. induction therapy in cadaveric kidney transplantation - efficacy and
223. Zhang T, Pierson III RN, Azimzadeh AM. Update on CD40 and CD154 safety after five years. Am J Transplant 2005;5:1347–53.
blockade in transplant models. Immunotherapy 2015;7(8):899–911. 247. Trzonkowski P, Zilvetti M, Chapman S, et al. Homeostatic repopula-
224. Malvezzi P, Jouve T, Rostaing L. Costimulation blockade in kidney trans- tion by CD28-CD8+ T cells in alemtuzumab-depleted kidney trans-
plantation: an update. Transplantation 2016;100(11):2315–23. plant recipients treated with reduced immunosuppression. Am J
225. Oura T, Hotta K, Lei J, et al. Immunosuppression with CD40 Transplant 2008;8(2):338–47.
costimulatory blockade plus rapamycin for simultaneous islet- 248. Trzonkowski P, Zilvetti M, Friend P, Wood KJ. Recipient memory-
kidney transplantation in nonhuman primates. Am J Transplant like lymphocytes remain unresponsive to graft antigens after
2017;17(3):646–56. CAMPATH-1H induction with reduced maintenance immunosup-
226. Kim J, Kim DH, Choi HJ, et al. Anti-CD40 antibody-mediated costim- pression. Transplantation 2006;82(10):1342–51.
ulation blockade promotes long-term survival of deep-lamellar por- 249. Hanaway MJ, Woodle ES, Mulgaonkar S, et al. Alemtuzumab induc-
cine corneal grafts in non-human primates. Xenotransplantation tion in renal transplantation. N Engl J Med 2011;364(20):1909–19.
2017;24(3). 250. Morgan RD, O’Callaghan JM, Knight SR, Morris PJ. Alemtuzumab
227. Cobbold S, Waldmann H. Skin allograft rejection by L3T4+ and LYT- induction therapy in kidney transplantation: a systematic review
2+ T cell subsets. Transplantation 1986;41:634–9. and meta-analysis. Transplantation 2012;93(12):1179–88.
228. Madsen JC, Peugh WN, Wood KJ, Morris PJ. The effect of anti- 251. Knechtle SJ, Pascual J, Bloom DD, et al. Early and limited use of tacro-
L3T4 monoclonal antibody on first-set rejection of murine cardiac limus to avoid rejection in an alemtuzumab and sirolimus regimen
allografts. Transplantation 1987;44(6):849–52. for kidney transplantation: clinical results and immune monitoring.
229. Shizuru JA, Gregory AK, Chao CT-B, Fathman CG. Islet allograft sur- Am J Transplant 2009;9(5):1087–98.
vival after a single course of treatment of recipient with antibody to 252. McMurchy AN, Bushell A, Levings MK, Wood KJ. Moving to toler-
L3T4. Science 1987;237:278–80. ance: clinical application of T regulatory cells. Semin Immunol
230. Benjamin RJ, Cobbold SP, Clark MR. Tolerance to rat mono- 2011;23(4):304–13.
clonal antibodies: implications for serotherapy. J Exper Med 253. Trzonkowski P, Bieniaszewska M, Juscinska J, et al. First-in-man
1986;163:1539–52. clinical results of the treatment of patients with graft versus host dis-
231. Benjamin RJ, Qin S, Wise MP, Cobbold SP, Waldmann H. Mecha- ease with human ex vivo expanded CD4+CD25+CD127- T regula-
nisms of monoclonal antibody-facilitated tolerance induction: a tory cells. Clin Immunol 2009;133(1):22–6.
354 Kidney Transplantation: Principles and Practice

254. Brunstein CG, Miller JS, Cao Q, et al. Infusion of ex vivo expanded T 257. van Kooten C, Rabelink TJ, de Fijter JW, Reinders ME. Mesenchymal
regulatory cells in adults transplanted with umbilical cord blood: safety stromal cells in clinical kidney transplantation: how tolerant can it
profile and detection kinetics. Blood 2011;117(3):1061–70. be? Curr Opin Organ Transplant 2016;21(6):550–8.
255. Di Ianni M, Falzetti F, Carotti A, et al. Tregs prevent GVHD and pro- 258. Tan J, Wu W, Xu X, et al. Induction therapy with autologous mesen-
mote immune reconstitution in HLA-haploidentical transplanta- chymal stem cells in living-related kidney transplants: a randomized
tion. Blood 2011;117(14):3921–8. controlled trial. JAMA 2012;307(11):1169–77.
256. Moreau A, Vandamme C, Segovia M, et al. Generation and in vivo 259. Reinders ME, de Fijter JW, Roelofs H, et al. Autologous bone mar-
evaluation of IL10-treated dendritic cells in a nonhuman pri- row-derived mesenchymal stromal cells for the treatment of allograft
mate model of AAV-based gene transfer. Mol Ther Meth Clin Dev rejection after renal transplantation: results of a phase I study. Stem
2014;1:14028. Cell Translation Med 2013;2(2):107–11.
22 Transplantation in
the Sensitized Recipient and
Across ABO Blood Groups
CARRIE SCHINSTOCK and MARK STEGALL

CHAPTER OUTLINE Sensitized Patients Therapeutic Approaches to Overcome


Alloantibody Detection Alloantibody
Clinical Approaches to Sensitized Patients Desensitization
Deceased Donor Transplantation Prevention or Treatment of Early Active AMR
Kidney Paired Donation Prevention or Treatment of Late AMR and
Kidney Transplantation in the Presence of Chronic AMR
DSA The Path to New Drug Approval in High-Risk
Immunologic Risk Patients
Hyperacute Rejection Mechanistic View of Antibody Production
and Injury
Early Active Antibody-Mediated Rejection
ABO-Incompatible Transplantation
Late Antibody-Mediated Rejection: Active
and Chronic AMR Conclusion

Naturally occurring antibodies against blood group anti- Sensitized Patients


gens and acquired alloantibodies against donor human
leukocyte antigens (HLAs; secondary to a prior kidney Historically, some of the first evidence for alloantibody was
transplant, blood transfusions, or pregnancy) may pose the retrospective study of Patel and Terasaki in 1969.3 This
major barriers to a successful renal transplantation. study showed that ability of the recipient’s serum to lyse
Because approximately 20% of renal allograft candi- donor cells in vitro was associated with allograft loss within
dates may be blood group incompatible with their living hours of transplantation in a high percentage of cases. Thus
donor and more than one-third will demonstrate some for many years the presence of a “positive crossmatch”
level of anti-HLA antibody pretransplant, understanding because of DSA was generally considered to be an abso-
the unique issues pertaining to donor-specific antibody lute contraindication to kidney transplantation. However,
(DSA) is important for the proper management of these in the late 1990s, several groups began to employ proto-
patients. cols aimed at overcoming the antibody barrier. In the early
Over the past decade innovative strategies have been 2000s, an increased understanding of the histology associ-
utilized to avoid incompatible transplantation, includ- ated with antibody and new techniques that enhanced the
ing the expansion of multicenter kidney paired dona- ability to understand the specificity and strength of DSA
tion programs and the increased priority for deceased helped clarify the risks and outcomes of transplantation for
donor allocation to highly sensitized candidates in the patients with DSA. In the past decade, the development of
US. Yet evidence is mounting that a role for incompatible paired living donation and increased national sharing for
transplantation (ABO and/or HLA) still exists in select highly sensitized patients has provided more options for
circumstances.1,2 these patients, yet many still are not transplanted. Recently
This chapter discusses the current options for transplan- an increasing number of clinical trials have been instituted
tation in sensitized renal transplant candidates or those to test new agents that raise hopes that new therapy might
with ABO-incompatible living donors. Specific emphasis is further improve the transplantation rates and the long-term
placed on what is known regarding the mechanisms and outcomes of highly sensitized renal transplant candidates.
treatment of both early and late antibody-mediated injury,
including the results of some recent therapeutic trials.
Finally, current knowledge regarding the mechanism of Alloantibody Detection
antibody production in the setting of renal transplantation
is outlined, highlighting important gaps in current knowl- To understand the therapeutic options for sensitized
edge in this emerging field. patients, one first must understand the various assays used

355
356 Kidney Transplantation: Principles and Practice

TABLE 22.1 Alloantibody Detection Assays a donor against whom the recipient has no DSA, but does
not give information about the level of alloantibody. Cur-
SCREENING ASSAYS rently, the cPRA can be used as a factor in deceased donor
Panel-reactive antibody (T cell only) allocation, as will be discussed later. The PRA was histori-
Solid-phase bead or ELISA assay cally determined when the transplant candidate’s serum
DONOR-SPECIFIC ALLOANTIBODY DETECTION TESTS was added to a panel of cells that represented the donor
Anti–Class I Sensitivity pool (hence the name “panel-reactive antibody”), but that
method is now infrequently used.
T-cell cytotoxicity (NIH-CDC) assay Lowest When a potential donor is identified, both solid-phase
T-cell AHG-CDC assay Highest
T-cell flow cytometric crossmatch and crossmatch assays are performed and interpreted
Solid-phase bead or ELISA assay together in context (i.e., sensitization history). However,
as the breadth of antibodies detectable by the solid-phase
Anti–Class I, II, or Both Sensitivity assay has expanded, some experts support the use of solid-
B-cell cytotoxicity (NIH-CDC) assay Lowest phase assays alone for a virtual crossmatch.11
B-cell flow cytometric crossmatch Highest Despite the technologic advances in HLA detection, there
Solid-phase bead or ELISA assay remains a considerable lack of standardization of practice
AHG-CDC, antihuman globulin complement-dependent cytotoxicity; ELISA, among transplant centers regarding the method, interpre-
enzyme-linked immunosorbent assay; NIH-CDC, National Institutions of tation, and reporting of the various tests. This lack of stan-
Health complement-dependent cytotoxicity. dardization, combined with the small number of patients
who undergo incompatible transplantation at individual
to determine the presence of alloantibody (Table 22.1). A centers, limits the ability to understand immunologic risk
more detailed description of these assays is presented else- in various settings. Major efforts are underway interna-
where in this book (see Chapter 10), but some key concepts tionally to standardize practices, and our understanding of
will be covered here. The sensitivity of the assay is impor- the immunologic risk of incompatible transplantation has
tant in determining the presence and amount of antibody. advanced.12
For example, complement-dependent cytotoxicity (CDC)
assays in which the recipient serum is tested for its ability
to lyse target lymphocytes is relatively insensitive. Antihu- Clinical Approaches to Sensitized
man globulin (T cell AHG) can be added to enhance the Patients
ability to detect class I anti-HLA antibodies, but this assay
is still considered the least sensitive of antibody detection Ideally alloantibody would be avoided at the time of trans-
techniques. Flow cytometric crossmatch methods are semi- plantation, but that is not always possible. The next section
quantitative and are more sensitive for detecting anti–class describes the options for patients with preformed anti-HLA
I and II antibodies. antibodies before transplantation.
Solid-phase assays have revolutionized this field because
they are the most sensitive, but also provide information
DECEASED DONOR TRANSPLANTATION
regarding the specificity of the HLA antibody. For this test,
single HLAs are bound to microspheres or enzyme-linked If sensitized patients do not have any prospective living
immunosorbent assay (ELISA) plates. For solid-phase donors, their only transplant option is to be placed on the
assays such as the commonly used LABscreen, the level of deceased donor waiting list. Approximately 30,000 patients
alloantibody is usually expressed as the mean fluorescence on the Organ Procurement and Transplantation Network/
intensity (MFI).4 MFI levels considered to be positive vary United Network for Organ Sharing (UNOS) cadaver donor
between laboratories and transplant programs, but an MFI kidney waiting list are “sensitized”13 and currently about
level of >1000 generally is accepted as positive. 7000 patients have a cPRA of 99% or 100% in the in US.1
Commercially available modified solid-phase assay tests In 2014 the deceased donor kidney allocation system
have also been developed to detect complement binding (KAS) changed, and it now offers renewed hope for sensi-
(C1q or C3d). DSA with C1q and/or C3d binding positivity tized transplant candidates. This system increases the allo-
is associated with the development of antibody-mediated cation priority for sensitized candidates on a sliding scale
rejection (AMR) and allograft loss,5–7 and some groups starting with a cPRA of 20%. Although all patients with a
routinely use these tests to distinguish the most deleterious cPRA of greater than 80% received allocation points in the
DSA. However, it remains unclear whether C1q positivity old US system, now patients with a cPRA of 80% to 84%
is superior to using MFI or antibody titer to predict AMR or receive 2.5 points and those with a cPRA of 100% receive
allograft loss,8–10 and thus testing for C1q or C3d binding is 202 points. Patients with 99% and 100% cPRA have even
not universally performed. greater access to the available donor pool because they
In practice, a solid-phase assay is commonly used as an also now have regional and national priority. Indeed, these
initial screening tool to determine the presence of alloan- changes have increased the number of sensitized patients
tibody at the time the transplant candidate is being evalu- who have been transplanted.1 Within the first year of
ated. This test is used to determine which HLA antigens implementation, nearly 1000 patients with cPRA of 99%
to avoid at the time of transplantation and can be used to or 100% received a transplant.
determine the breadth of sensitization against HLA by cal- The Acceptable Mismatch Program of Eurotransplant
culating the panel-reactive antibody (cPRA). The cPRA is another approach to transplanting sensitized candi-
provides information regarding the probability of finding dates with a deceased donor. In this program, anti-HLA
22 • Transplantation in the Sensitized Recipient and Across ABO Blood Groups 357

antibodies are identified using a CDC assay. Highly sen- The survival advantage of an incompatible transplant
sitized patients (PRA >85%) are placed at the top of the compared with being on the waiting list in the US was
kidney match run and organs are allocated based on the not found in a recent study.32 A matched cohort analy-
absence of DSA against the donor HLA (i.e., “acceptable sis was performed in the United Kingdom comparing 213
mismatch” for HLA). In this program, approximately 60% patients who underwent positive crossmatch transplan-
of the highly sensitized patients are transplanted within 2 tation and 852 matched controls.32 The patient survival
years after inclusion in this acceptable mismatch program was similar among patients who received a positive
and the short-term graft survival appears similar to that of crossmatch transplant or remained on dialysis at 3 and 7
unsensitized patients. Low levels of DSA that are present years.32 In this cohort, the 5-year patient survival after
at the time of transplant are not considered at the time of incompatible transplant was 68% after HLA-incompat-
allocation, and it is unclear what the long-term outcomes ible transplantation, 89% after compatible living donor
will be in this program. transplant, and 77% for compatible deceased donor
transplant.32 The differences in the results between the
US and United Kingdom cohort are likely related to the
KIDNEY PAIRED DONATION
increased mortality on dialysis in the US and high-risk
If a sensitized candidate has potential living donors, these transplant group from the United Kingdom (all patients
donors should be HLA-typed to find a crossmatch-negative received positive crossmatch transplants in the United
donor. If no such donor can be found, sensitized candidates Kingdom whereas 185 patients in the US cohort had pos-
may opt to enter into one of the growing number of paired itive DSA by solid-phase single-antigen bead assay, but
living donor programs.14–19 These “exchange” schemes had a negative crossmatch).
have been shown to increase the transplantation rate of Regardless of the patient survival advantage after incom-
ABO-incompatible and sensitized patients20 (Fig. 22.1). patible transplantation, allograft survival is reduced.33
Although these programs increase the number of potential These transplants are also associated with higher rates of
donors for sensitized individuals, patients with antibodies hospital readmission in the first year posttransplant and
against a wide variety of HLA types still may not be able to are more expensive.31–34 The potential disadvantages of
find a crossmatch-negative donor, even if the donor pool is incompatible transplantation should not be used to dis-
very large. One of the central questions in paired donation is: courage this practice, but should be used for clinical deci-
How long should a sensitized patient wait for a crossmatch- sion making.
negative donor versus proceeding to a transplant using a Whereas avoiding DSA is ideal, many highly sensitized
donor against whom the recipient has DSA? Because many patients remain on the waiting list despite paired donor
sensitized patients may not find a DSA-negative donor, programs and increased priority for deceased donors.
most paired donor programs employ “optimization” pro- When considering a transplant in the setting of DSA, the
tocols that seek to identify donors for sensitized patients major risk to consider is that of graft loss. Some incompat-
with lower levels of DSA than their original donor. Thus ible transplants are relatively low risk whereas others are
transplantation of a sensitized patient in this setting might prohibitively high risk for early graft loss. Thus before we
employ both paired donation and living donor incompatible discuss the various options for incompatible transplanta-
transplant protocols.21 tion, we need to examine the concept of immunologic risk
associated with DSA.
KIDNEY TRANSPLANTATION IN
THE PRESENCE OF DSA
Immunologic Risk
A viable option for sensitized candidates is to perform a
kidney transplant despite the presence of DSA.22–28 Over- Clinicians now have the ability to semiquantitatively esti-
all, data suggests that patient survival may be improved mate the level DSA from very low to very high. Although
after incompatible transplant compared with remain- DSA detected with solid-phase assays and/or crossmatch
ing on dialysis29,30 (Fig. 22.2). This is despite reduced testing is no longer considered an absolute contraindication
allograft survival as will be discussed in the next section. to transplant, it continues to represent an immunologic risk.
In a multicenter trial involving 22 centers in the US, the This concept of immunologic risk has emerged as one of the
survival of 1025 patients who received an incompatible core principles in the transplantation of sensitized patients.
living donor kidney transplant were matched with con- Quantifying this risk is an important aspect of designing
trols who remained on the waitlist or received a deceased protocols to enable successful kidney transplantation in
donor transplant.31 At 1, 3, and 5 years patients who sensitized patients. A combination of the various previously
received an incompatible kidney transplant had improved described assays allows clinicians to better determine the risk
survival (see Fig. 22.2). This patient survival benefit was of antibody-mediated graft damage in sensitized patients.
significant at 8 years across all levels of incompatibility However, current assays cannot completely determine the
(DSA positive only and positive flow cytometric cross- entire immunologic risk of all patients. Even when all DSA
match). Specifically, at 5 years the patient survival was testing is negative, sensitized patients are at increased risk
86.0% in those patients who received an incompatible for T cell mediated rejection and or may possess antibodies
living donor transplant, 74.4% in those who received a against antigens not detected by current assays.
deceased donor transplant and/or remained on the wait- AMR can be categorized broadly as hyperacute rejection,
list, and 59.2% in those patients who remained on the early active AMR, acute active AMR, and chronic active
waitlist and did not receive a transplant.29 AMR.
358 Kidney Transplantation: Principles and Practice

Two-Way Exchange Three-Way Exchange List Exchange


Recipient 1
Deceased donor
Donor 1 (hard-to-match)
waitlist
A B
A
O A
Donor 1 Recipient 1

Waitlist
priority
B A
A Donor 3
O
Donor 2 Recipient 2
Recipient 2 B
(Positive
crossmatch)
B B
O
A B Donor 2 Recipient 3

A
Domino-Paired Donation

Using Compatible Pairs


O Altruistic donor
Compatible
O A
A
O
Donor 1 Recipient 2
A Donor Recipient
O
(hard-to-match)
Donor 1 Recipient 1 C
A
O
A Donor 2 Recipient 2
(hard to match)
Waitlist
D candidate E

NEAD Chain

Segment 2
O Altruistic donor
B
O
Donor 1 Recipient 1
Donor 3 Recipient 3
A
O
A
O
Donor 2
(bridge donor) Recipient 2 Donor 4 Recipient 4

B A
B A

Donor 5 Recipient 5
Segment 1 (bridge donor)
F
Fig. 22.1 Paired donation schemas. (A) Two incompatible pairs swap kidneys in a two-way exchange. (B) A three-way exchange with no reciprocal
exchanges so hard-to-match pairs are better matched. (C) In a list exchange, an incompatible donor donates to a waitlist candidate in exchange for
waitlist priority for the recipient. (D) DPD pairs an altruistic donor with an incompatible recipient. The donor then continues the chain or donates to a
waitlist candidate. (E) A compatible pair can join an exchange helping hard-to-match pairs. (F) NEAD chains are like domino chains except the last donor
becomes a bridge donor and awaits more pairs to perpetuate the chain at a later time.38 (Taken from Wallis CB, Samy KP, Roth AE, Rees MA. Kidney paired
donation. Nephrol Dial Transplant 2011;26:2091–9.)

HYPERACUTE REJECTION level of DSA at the time of transplantation. The exact level
The primary goal of “desensitization” is the avoidance of of DSA that leads to this complication is unclear, but most
the catastrophic occurrence of hyperacute rejection, which programs consider a positive T cell AHG crossmatch a high
occurs minutes to hours posttransplant leading to almost risk for hyperacute rejection. Indeed, early in our experi-
immediate allograft loss. The incidence is related to the ence, we transplanted 10 patients who, despite multiple
22 • Transplantation in the Sensitized Recipient and Across ABO Blood Groups 359

100 cytometric crossmatch, to up to approximately 40% in patients


with preformed DSA and high positive flow cytometric cross-
80
match.22,28,36,37 In a series from Hôpital St. Louis in Paris, the
incidence of AMR was 36.4% with a baseline DSA MFI of 3001
to 6000 and 51.3% with a baseline DSA MFI of >6000.37
Survival (%)
60 The incidence of early active AMR appears to correlate
P < 0.001
with the development of high levels of DSA after transplanta-
40 P < 0.001
tion. Our group examined the natural history of DSA early
after transplantation and its relationship with early active
Recipients of incompatible transplants AMR in 70 positive crossmatch kidney transplant recipi-
20
Waiting-list-or-transplant control group ents.22 The overall incidence of AMR was 36% with the
Waiting-list-only control group mean time to diagnosis of approximately 10 days, and all
0 episodes occurred within 1 month of transplantation. All but
0 1 2 3 4 5 6 7 8 one AMR episode were associated with allograft dysfunction
Years (defined as an increase in serum creatinine over nadir >3.0
No. at Risk mg/dL in the first month), but in many instances evidence of
Recipients of incompatible 1025 958 832 584 327 histologic injury preceded the increase in serum creatinine.
transplants The time course of changes in DSA posttransplant cor-
Waiting-list-or-transplant 5125 4546 3673 2493 1414
control group related well with the development of AMR (Fig. 22.3). Over-
Waiting-list-only 5125 4141 3024 1810 916 all, mean DSA levels decreased by day 4 and remained low
control group in patients who did not develop AMR. By day 10, however,
Fig. 22.2 Overall comparison of survival between the group that DSA levels increased in patients developing AMR, with 92%
received kidney transplants from HLA incompatible live donors (23/25) of patients with a flow cytometric crossmatch of
and each group of matched controls. In one control group, the con-
trols remain on the waiting list or received a transplant from a deceased
>359 (molecules of equivalent soluble fluorochrome units of
donor. In the other control group, controls remained on the waitlist approximately 34,000) developing AMR. The B flow cytomet-
and did not receive a transplant from a deceased donor. (From Orandi ric crossmatch and the total DSA measured by single-antigen
BJ, Luo X, Massie JM, et al. Survival benefit from kidney transplants from beads with the solid-phase assay correlated well across a wide
HLA-incompatible live donors. N Engl J Med 2016;374;940–50.) spectrum, suggesting that either could be used for monitoring.
Importantly, when allografts demonstrated the cluster indica-
plasmapheresis treatments (mean of 10 treatments), were tive of AMR (high DSA, histologic injury, and graft dysfunc-
unable to achieve a negative T cell AHG crossmatch.26 tion), all but one (24/25) were also C4d-positive.
Given that these highly sensitized patients had almost no
other option for transplant at the time, we performed the
LATE ANTIBODY-MEDIATED REJECTION: ACTIVE
transplant despite the persistence of a low titer T cell AHG
crossmatch (undiluted to 1:8) on the day of transplantation. AND CHRONIC AMR
Of these 10 patients, two developed hyperacute rejection. Although classification systems have been developed to dif-
The incidence of early active AMR in this group was 70% ferentiate between active and chronic active AMR, these
and the 1-year allograft survival was only 50%. In today’s histologic lesions represent a continuum of injury.35 His-
transplant environment, almost no kidney transplants are tologically, active AMR usually presents with glomerulitis
performed in the setting of high levels of DSA as defined by a and peritubular capillaritis with or without C4d positivity
positive T cell AHG crossmatch or MFI >10,000. (whether it occurs early or late posttransplant).35 The timing
and clinical relevance of active AMR is variable. As described
EARLY ACTIVE ANTIBODY-MEDIATED previously, patients can present with an active AMR that
REJECTION presents early within the first few weeks posttransplant. If the
early active AMR is averted, many patients with DSA pres-
Even if hyperacute rejection is avoided, patients with DSA ent at the time of transplantation develop active AMR at a
still have a high incidence of active AMR within the first few later time point. The onset is often insidious, subclinical, and
weeks after transplantation typically during an amnestic unrecognized without surveillance biopsy protocols.
response. Although the incidence of hyperacute rejection When the microvascular inflammation (glomerulitis and
has decreased, active AMR that occurs early posttransplant peritubular capillaritis) and/or C4d positivity is accompa-
has emerged as one of the major complications when trans- nied by duplication of glomerular basement membranes or
planting highly sensitized recipients. transplant glomerulopathy, criteria for chronic active AMR
From a histologic perspective the kidney biopsy usually has been reached. Chronic antibody-mediated allograft
shows evidence of C4d deposition in the peritubular capil- injury has become increasingly implicated as a major cause
laries and microvascular inflammation (peritubular and/ of late renal allograft loss.33,38–43 This is especially common
or glomerular capillaritis). Depending on the severity, in patients with DSA at the time of transplantation, but actu-
thrombosis and evidence of acute tubular injury may also ally most of the chronic active AMR that is encountered in
be present.35 Usually these rejections are associated with clinical practice occurs in the setting of de novo DSA.44–46
an abrupt reduction in glomerular filtration rate (GFR), but Chronic active AMR generally carries one of the worst
these rejections can also be subclinical. prognoses of any histologic lesion found on surveillance
The reported incidence of early active AMR is based largely biopsy. In one study, 60% of grafts with chronic active AMR
on single-center reports and ranges from as low as approxi- on a 1-year biopsy either failed or lost >50% of their func-
mately 1% in patients with preformed DSA and negative flow tion by 5 years after transplantation.38 Many patients with
360 Kidney Transplantation: Principles and Practice

600 600

500 500

BFXM (channel shift)

BFXM (channel shift)


400 400

300 300

200 200

100 100

0 0
A Baseline Day 0 Day 4 Day 10 Day 28 B Baseline Day 0 Day 4 Day 10 Day 28

600 600

500 500
BFXM (channel shift)

BFXM (channel shift)


400 400

300 300

200 200

100 100

0 0
C Baseline Day 4 Day 10 Day 28 D Baseline Day 4 Day 10 Day 28

Fig. 22.3 Correlation between DSA levels and early acute clinical antibody-mediated rejection (AMR). (A) DSA levels from baseline to 28 days
posttransplant for the high DSA patients without AMR; (B) high DSA patients with AMR; (C) low DSA patients without AMR; and (D) low DSA patients
with AMR. BFXM, B flow cytometric crossmatch. (From Burns JM, Cornell LD, Perry DK, et al. Alloantibody levels and antibody mediated rejection early after
positive crossmatch kidney transplantation. Am J Transplant 2008;8:2684.)

1.0
Key: –DSA–XM
P < 0.01 +DSA/-XM
0.8
+DSA/low + XM
chronic AMR
% allografts

+DSA/high + XM
0.6 –DSA/+XM

0.4

0.2

0.0
0 1 2 3 4 5
Years posttransplant
Fig. 22.4 Incidence of chronic AMR. The overall incidence of chronic AMR was 11.7% and was determined by examining both surveillance and
indication biopsies. The incidence correlated with increasing DSA at baseline and was the following: 8.2% in DSA negative/negative flow cytometric
crossmatch patients (−DSA/−XM); 17.0% in positive DSA/negative flow crossmatch patients (+DSA/−XM); 30.6% in positive DSA/low positive flow cross-
match (+DSA/low+XM); and 51.2% in the +DSA/high+XM group, P < 0.01 (Cochran test for trend). The incidence of chronic AMR was similar in patients
with negative DSA/negative flow crossmatch and negative DSA/positive flow crossmatch groups (8.2% vs. 15.4%, P = 0.19) (From: Schinstock CA, Gandhi
M, Cheungpasitporn W, et al. Kidney transplant with low levels of DSA or low positive B-flow crossmatch: an underappreciated option for highly sensitized
transplant candidates. Transplantation 2017;101(10):2429–39.)

chronic active AMR have minimal or no interstitial fibrosis testing but negative with flow cytometric crossmatch tests
suggesting that it is its own distinct process separate from (+DSA/-XM), 30.6% in patients with positive DSA based
other forms of chronic injury.40 on solid-phase tests and low positive flow cytometric cross-
Like early acute, active AMR, the incidence of chronic match testing (+DSA/low+XM), and 51.2% in patients with
active AMR depends on the DSA and crossmatch status positive solid-phase tests and high positive flow cytometric
at the time of transplantation. We performed a retrospec- crossmatch testing (+DSA/high+XM)2 (see Fig. 22.4).
tive observational study to examine the allograft histology The renal allograft survival after incompatible transplan-
and allograft survival after transplantation in the setting tation also correlates with the amount of DSA at the time of
of various levels of DSA and flow cytometric crossmatch. transplantation.2,47,48 A multicenter observational study of
We found that the incidence of chronic active AMR corre- the living donor transplants performed at 22 centers in the
lated with increasing DSA at baseline (Fig. 22.4) and was US showed that the 1-year unadjusted all-cause graft loss was
as follows at a mean follow-up of 4.1 ± 1.9 years: 8.2% in 3.9%, 3.8%, 6.9%, and 19.4% for compatible, positive DSA by
patients who had negative DSA and negative flow cyto- SAB and negative flow cytometric crossmatch, positive flow
metric crossmatch at the time of transplant (−DSA/−XM), cytometric crossmatch and negative cytotoxic crossmatch,
17.0% in patients who were positive for DSA by solid-phase and positive cytotoxic crossmatch transplant recipients,
22 • Transplantation in the Sensitized Recipient and Across ABO Blood Groups 361

40
PCC
35 PFNC
PLNF
30

Percent graft loss


Compatible
25

20

15

10

0 P < 0.001

0 1 2 3 4 5
Years since transplantation

Number at
Risk
PCC 304 245 226 210 186 157
PFNC 536 499 469 419 337 262
PLNF 185 178 163 140 105 80
Compatible 9669 9291 8503 7132 5819 4720
Fig. 22.5 All-cause allograft loss by crossmatch and DSA testing at the time of transplant. PCC, positive cytotoxic crossmatch; PFNC, positive flow cyto-
metric crossmatch and negative cytotoxic crossmatch; PLNF, positive Luminex single-antigen bead and negative flow cytometric crossmatch.(From Orandi
BJ, Garonzik-Wang JM, Massie AB, et al. Quantifying the risk of incompatible kidney transplantation: a multicenter study. Am J Transplant 2014;14(7):1573–80.)

1.0 DESENSITIZATION
0.9 For many years, the mainstay of “desensitization” (i.e., anti-
-XMKTx
body reduction before transplantation) involved either mul-
Allograft survival

Class I only tiple plasma exchange treatments or high-dose intravenous


0.8
immunoglobulin either alone or in combination with other
agents such as rituximab. All of these studies except one were
0.7 nonrandomized single-center experiences. The lack of stan-
Class II only
P = 0.0003 dardization made interpretation and comparison of these vari-
0.6
Class I + II
ous approaches difficult. What became clear from this era was
that it is possible to transplant patients with relatively high lev-
0.5 els of DSA and achieve acceptable 1-year graft survival rates
0 365 730 1095 1460 1825
(i.e., acceptable to Centers for Medicare & Medicaid Services
Days after transplant (CMS) under the conditions of participation). Although paired
Fig. 22.6 5-Year outcomes after positive crossmatch kidney trans- donation and increased deceased donor waiting list priority
plant. Allograft survival is significantly lower in positive crossmatch has led to decreased demand for desensitization, it is likely that
recipients compared with negative crossmatch recipients, particularly
in patients with anti–Class II DSA at baseline. (Taken from Bentall A, Cor-
this trend will change. Many highly sensitized patients who
nell LD, Gloor JM, et al. Five-year outcomes in living donor kidney trans- have been on dialysis for a prolonged period of time have a
plants with a positive crossmatch. Am J Transplant 2013;13(1):76–85.) high mortality rate and are still waiting for an organ49; thus
the demand for desensitization will likely increase—especially
respectively. At 5 years, the graft loss was 16.6%, 20.2%, if new technology emerges.
28.8%, and 39.9% in these groups48 (Fig. 22.5). One promising technology is IgG endopeptidase (IdeS).
The type of antibody at the time of transplantation also This bacterial-derived enzyme rapidly degrades all IgG sub-
influences allograft survival. The presence of anti–class II types resulting in a rapid temporary reduction of serum IgG
antibody at the time of transplant has a higher correlation levels. In a recent study, 24 of 25 highly sensitized patients
with chronic AMR and allograft loss33,43 (Fig. 22.6). from the US and Sweden received IdeS and subsequently
underwent successful deceased donor kidney transplanta-
tion.50 One patient had hyperacute rejection likely from
Therapeutic Approaches to unrecognized IgM, IgA, or non-HLA antibody; but this ther-
Overcome Alloantibody apy may become an important tool in the future to desensi-
tize patients with limited transplant options. Further studies
Three specific problems that need therapy are: desensitization, with this drug are likely to be forthcoming.
prevention and/or treatment of acute AMR, and prevention/ Proteasome inhibitors have been shown to cause
treatment of chronic AMR. We discuss each individually while plasma cell (PC) death in vitro and in mouse models lead-
acknowledging that there can be significant overlap. ing to reductions in antibody production. Bortezomib,
362 Kidney Transplantation: Principles and Practice

a first-in-class proteasome inhibitor, has been used for Research interest in this area appears to be at an all-time
desensitization and AMR with mixed results at the expense high and involves a variety of agents. One long-term ret-
of adverse events.51–57 Recently a small study suggested rospective study from two centers suggested that an anti-
promising results with carfilzomib, a second-generation body against the interleukin (IL)-6 receptor (expressed
proteasome inhibitor.58 New therapies aimed to interrupt on PCs) might decrease progression of ongoing antibody
the survival signals of the long-lived plasma cell are also injury.64 Two phase III randomized trials, both involving
being developed. C1 esterase inhibitors, are now underway. Both involve
patients with biopsy-proven acute AMR and use surro-
PREVENTION OR TREATMENT OF EARLY gate endpoints to obtain accelerated approval. One study
(NCT02547220) uses the lack of progression to transplant
ACTIVE AMR
glomerulopathy as the surrogate endpoint, and the other
Active AMR can be a major complication in the first few study (NCT03221842) uses a lack of decline in eGFR and
weeks after transplantation in patients with DSA at the time graft loss as a composite endpoint. Interestingly, surrogate
of transplantation. Many of these are severe clinical rejec- endpoints have not been used for drug approval in trans-
tions with rapidly rising serum DSA levels accompanied plant studies. Therefore this is a new area for regulators and
by significant graft dysfunction. The incidence of active investigators in addition to a new area of therapy.
AMR that occurs early posttransplant depends on the level
of DSA at baseline, but, despite desensitization, may be as THE PATH TO NEW DRUG APPROVAL IN HIGH-
high as 40% to 50%27,28 in patients with enough antibody RISK PATIENTS
to cause a strongly positive crossmatch.
Blockade of the terminal complement component C5 with It important to reemphasize that there is not an effective US
the anti-C5 monoclonal antibody eculizumab was shown to Food and Drug Administration (FDA)-approved therapy for
decrease early active AMR (defined as increased DSA, C4d any of the problems that face renal transplant candidates or
positivity, and increased creatinine) in positive crossmatch recipients with DSA. A persistent barrier to drug approval is
living donor kidney transplants. The rate of AMR decreased the lack of expert consensus regarding important features
from 41.2% in historical controls to 7.7% in this single-cen- of study design such as inclusion criteria and endpoints.65
ter study.27 Another single-armed study involving deceased Fortunately, the progress made in the characterization of
donor kidney transplant recipients with C1q-positive DSA alloantibody, increased experience and data regarding the
also suggested a decrease in AMR compared with historical correlation between histologic findings and clinical out-
controls.59 A multicenter, phase III randomized clinical trial comes have increased our ability to construct studies that
failed to validate this finding, but the major reason for fail- are more likely to show efficacy of potential therapeutic
ure was likely a result of the study design (ClinicalTrials.gov agents. For example, a decrease in cPRA or an increased
identifier: NCT01399593). The multicenter study included renal transplantation rate may be appropriate endpoints
many patients with relatively low levels of DSA, and thus for desensitization. A reduced incidence of AMR or lack of
the rejection rate in the control group was too low to show progression to transplant glomerulopathy may also corre-
an effect. Additionally, the reduction in early active AMR late with treatment response and subsequent improvement
did not translate into reduced chronic active AMR at 1 year in renal allograft survival. Although is it unclear at present
posttransplant.60 what constitutes the ideal pathway forward in demonstrat-
More proximal blockade of complement with C1 esterase ing efficacy of therapy, attempts are being made to identify
inhibitors has also been used in small single-center studies more effective means of desensitization and treatment of
for the prevention and treatment of refractory AMR with AMR.
favorable results,61–63 but larger multicenter randomized
trials are ongoing for confirmation.
Mechanistic View of Antibody
PREVENTION OR TREATMENT OF LATE AMR AND Production and Injury
CHRONIC AMR The pathway to antibody production has been clearly
As mentioned earlier, the incidence of chronic active AMR delineated in numerous animal and human studies.66–68
is increased in kidney transplant recipients with DSA, but The bone marrow continuously generates a large variety
evidence suggests that 5-year allograft survival is only of naïve B cells expressing cell surface immunoglobulin.
reduced in patients with DSA and a high B flow cytomet- Each naïve B cell’s immunoglobulin is unique and capa-
ric crossmatch.2 Allograft survival appears to be similar ble of interacting with an enormous variety of antigens,
in patients with no preformed DSA and low level DSA at including all types of class I and class II HLA molecules.
baseline (positive DSA on solid-phase assays only or low B These mature, but naïve, B cells remain in a quiescent state
flow cytometric crossmatch).2 Nevertheless, the detection until they encounter antigen in secondary lymphoid tis-
of active or chronic AMR on protocol biopsies early after sue, such as the spleen. Upon antigen exposure and sub-
transplantation is associated with shortened allograft sur- sequent activation by T cells that have encountered the
vival. Therapy to prevent or treat chronic active AMR is also same antigen, the B cell antigen-specific population rap-
a major unmet need for patients who develop de novo DSA idly proliferates and differentiates into other populations to
after transplantation. In fact, antibody-mediated injury is a more efficiently secrete antibody. Specifically, this leads to
major contributor to 29% of allograft losses over the first 10 the development of plasma cells (either short-lived or long-
years posttransplant.64 lived) and memory B cells. Memory B cells also express cell
22 • Transplantation in the Sensitized Recipient and Across ABO Blood Groups 363

surface immunoglobulin and are capable of rapid conver- Patient Survival After Live Donor Kidney Transplant
sion to plasma cells within hours of reexposure to antigen, According to ABO Compatibility
but they do not secrete antibody themselves. In contrast, 100%
plasma cells become terminally differentiated and secrete 90%
large amounts of antibody.
80% Compatible
Long-lived plasma cells can persist for years in special
microenvironments of the marrow and spleen, continu- 70%
A2i

% Survival
ously producing antibody even in the absence of antigenic 60%
stimulation. Because of their terminal differentiation, they 50% ABOi *
are resistant to most pharmacologic agents. Most of the 40%
anti-HLA antibody detected in sensitized patients is likely 30%
produced by long-lived plasma cells. Plasma cells seem to be
20%
resistant to most immunomodulatory agents commonly in
use in clinical transplantation.66,69 They do not use IL-2 for 10% *P = 0.0009 vs. compatible
their function and are not significantly inhibited by either 0%
calcineurin inhibitors or antibodies against IL-2 receptors. 0 1 2 3 4 5
They do not expression CD52, the target for alemtuzumab, Years after transplant
and they are resistant to desensitization with intravenous
immunoglobulin (IVIG) and thymoglobulin. Additionally, Death-Censored Live Donor Allograft Survival
because they do not express CD20, they are essentially According to Blood Type Compatibility
unaffected by rituximab. We and others have attempted
100%
to find agents that might deplete plasma cells in sensitized
90%
recipients. In vitro, bortezomib caused apoptosis of plasma
cells, but this was not observed with rituximab, IVIG, or 80% Compatible
thymoglobulin.66,69 Bortezomib led to a significant deple- 70%
% Survival A2i
tion of bone marrow-derived plasma cells in some patients 60%
in a dose escalation study70 (see Fig. 22.4). 50% ABOi †
The way in which DSA ultimately leads to rejection is 40%
complex and incompletely understood. Three main patho- 30%
logic processes are likely involved and include: direct bind-
20%
ing of DSA to the endothelium and subsequent endothelial †P
10% < 0.05 vs. compatible
cell activation, recruitment of an inflammatory infiltrate,
and complement activation. One of the histologic hallmarks 0%
of AMR is microvascular inflammation, specifically glo- 0 1 2 3 4 5
merular and peritubular capillaritis. Direct activation of the Years after transplant
endothelium and the complement system leads to recruit- Fig. 22.7 Posttransplant patient and death-censored allograft survival
ment of an inflammatory infiltrate, but evidence suggests after ABO incompatible transplantation in the United States. Patients
that natural killer (NK) cells play a role in antibody-medi- transplanted from 2000 to 2011. (Taken from Axelrod D, Segev D, Xiao H,
Schnitzler M, et al. Economic impacts of ABO-incompatible live donor kid-
ated allograft injury.71,72 Increased NK cell transcripts are ney transplantation: a national study of Medicare-insured recipients. Am J
found in renal allograft biopsies during AMR. In renal and Transplant 2016;16:1465–73.)
cardiac transplant models, the severity and frequency of
AMR was reduced when NK cells were depleted with mono- with hyperacute and early AMR. However, like sensitized
clonal antibody.73–75 patients, several options now exist for patients who are ABO
The potential role of complement in AMR has been recog- incompatible with their living donors. These include (1)
nized for decades. In the early 1990s, C4d staining of renal being placed on the deceased donor waiting list, (2) entering
tissue became reliable and increasingly utilized. An inactive a paired living donor program, or (3) undergoing a desensi-
split product resulting from the cleavage of C4b, C4d cova- tization protocol and receiving the ABO-incompatible living
lently binds to the site of complement activation, which donor kidney despite the presence of antidonor antibody.
is usually vascular endothelium in the renal allograft. Being placed on the deceased donor waiting list is the most
With advances in tissue typing techniques, an association commonly used option. Because most ABO-­incompatible
between DSA and C4d staining was made. Subsequently, candidates are blood group O (78% in our series),76 their
several clinical studies have shown that C4d staining of current mean waiting time for a deceased donor kidney is
the peritubular capillaries is associated with AMR and graft approximately 5 years in the US. This long waiting time
failure. translates into increased morbidity and mortality pre-
and posttransplantation, especially in older patients and
diabetics.
ABO-Incompatible In countries with fewer deceased donor kidney trans-
Transplantation plants, ABO-incompatible kidney transplantation may be
the only option.77,78 Numerous studies have shown that
Historically, the presence of antibody against donor blood ABO-incompatible living donor kidney transplantation has
group also has been considered a contraindication to kid- higher early graft loss rates than ABO-compatible grafts,
ney transplantation, with high antibody levels associated but long-term survival is comparable (Fig. 22.7), ranging
364 Kidney Transplantation: Principles and Practice

from 79% to 98% at 5 years.33,68,77,79–84 The reasons for Conclusion


the improved long-term survival of ABO incompatible
compared with DSA-positive transplants is unclear. The The treatment options for renal transplant candidates who
development of accommodation or even tolerance to blood have DSA or who are blood group incompatible with their
group antibody may occur despite the continued presence living donor continue to evolve. In many instances, paired
of anti–blood group antibody.85–87 A recent histologic study donation or acceptable mismatch programs can avoid these
suggests that ABO-incompatible renal allografts have less antibody barriers. However, for some patients, a DSA-pos-
peritubular capillaritis and less transplant glomerulopathy itive or ABO-incompatible kidney transplant may be the
compared with DSA-positive grafts at 5 years. best treatment option. The immunologic risk of patients
The level of anti–blood group antibody that causes hyper- varies from very low to prohibitively high, and protocols
acute rejection has not been determined exactly and may can be tailored to reduce the risk of antibody-mediated
vary. Several groups have shown that anti–blood group damage. Novel new therapies such as the use of eculizumab
isohemagglutination titers of less than 1:8 or 1:16 seems to to prevent early AMR in DSA-positive transplants suggest
be “safe” at the time of transplantation because evidence of that this major complication is becoming more manage-
antibody deposition is not seen on 30-minute postreperfu- able. However, chronic antibody-mediated injury remains
sion surveillance biopsy specimens. Achieving these “safe” a major unsolved problem in patients who receive HLA-
levels of antibody can be difficult in some patients, however. incompatible transplants. Future research efforts focusing
Patients who at baseline (before any therapy) have high on the mechanisms of antibody production and its effect on
levels of anti–blood group antibody (e.g., >1:512) rarely the graft should provide for continued progress in this chal-
achieve adequate reduction of anti–blood group antibody lenging field.
using current protocols. Performing splenectomy either
before or at the time of transplantation might allow success- References
ful ABO-incompatible transplantation even in patients with 1. Stewart DE, Kucheryavaya AY, Klassen DK, Turgeon NA, Formica RN,
very high anti–blood group antibody levels. Aeder MI. Changes in deceased donor kidney transplantation one year
Protocols have evolved to include preemptive plas- after kas implementation. Am J Transplant 2016;16(6):1834–47.
mapheresis treatments, and antibody monitoring aimed 2. Schinstock CA, Gandhi M, Cheungpasitporn W, et al. Kidney trans-
at maintaining low levels of antidonor antibody in the plant with low levels of DSA or low positive B-flow crossmatch: an
underappreciated option for highly sensitized transplant candidates.
first 2 weeks after transplantation. Most protocols aim Transplantation 2017;101(10):2429–39.
to maintain the isoagglutination anti–blood group anti- 3. Patel R, Terasaki PI. Significance of the positive crossmatch test in kid-
body titer less than 1:16 for 2 weeks.23,77,80,81,83,84 As a ney transplantation. N Engl J Med 1969;280(14):735–9.
result of the lack of prospective randomized trials, many 4. Pei R, Lee JH, Shih NJ, Chen M, Terasaki PI. Single human leukocyte anti-
gen flow cytometry beads for accurate identification of human leukocyte
important questions regarding ABO-incompatible kid- antigen antibody specificities. Transplantation 2003;75(1):43–9.
ney transplantation remain unanswered, including: (1) 5. Loupy A, Lefaucheur C, Vernerey D, et al. Complement-binding
What is the maximum blood group titer that is accept- anti-HLA antibodies and kidney-allograft survival. N Engl J Med
able at the time of transplant? (2) Do immunoadsorption 2013;369(13):1215–26.
columns that specifically remove anti–blood group anti- 6. Sicard A, Ducreux S, Rabeyrin M, et al. Detection of C3d-binding
donor-specific anti-HLA antibodies at diagnosis of humoral rejection
body have less morbidity and better outcomes than stan- predicts renal graft loss. J Am Soc Nephrol 2015;26(2):457–67.
dard plasmapheresis?81 (3) What is the role of rituximab 7. Baid-Agrawal S, Lachmann N, Budde K. Complement-binding
in these protocols? (4) Is steroid-free immunosuppression anti-HLA antibodies and kidney transplantation. N Engl J Med
possible in ABO-incompatible transplantation?79 and (5) 2014;370(1):84.
8. Zeevi A, Lunz J, Feingold B, et al. Persistent strong anti-HLA antibody
How does one generate and measure accommodation at high titer is complement binding and associated with increased risk
and tolerance to ABO-incompatible transplantation in of antibody-mediated rejection in heart transplant recipients. J Heart
both adults and children?56,87 Lung Transplant 2013;32(1):98–105.
A major barrier to the widespread application of ABO- 9. Gandhi M, Degoey S, Henderson N, Voit L, Krueter J. C1q single anti-
incompatible kidney transplantation is the increased cost gen bead assay only detects high titer/avidity class-I anti HLA anti-
bodies detected by single antigen beads. Hum Immunol 2014;75:73.
compared with conventional transplants.79,88 A recent 10. Tambur AR, Herrera ND, Haarberg KM, et al. Assessing antibody
economic analysis was performed comparing the cost of the strength: comparison of MFI, C1q, and titer information. Am J Trans-
transplant episode of ABO compatible versus-incompatible plant 2015;15(9):2421–30.
transplants performed from 2000 through 2011. The aver- 11. Tambur AR, Ramon DS, Kaufman DB, et al. Perception versus real-
ity?: virtual crossmatch—how to overcome some of the technical and
age overall cost of the ABO-incompatible transplant episode logistic limitations. Am J Transplant 2009;9(8):1886–93.
was $65,080 versus $32,039. This analysis also showed 12. Tait BD, Susal C, Gebel HM, et al. Consensus guidelines on the testing
that posttransplant costs were higher. The marginal costs and clinical management issues associated with HLA and non-HLA
compared with ABO-compatible transplants during year 1 antibodies in transplantation. Transplantation 2013;95(1):19–47.
was $25,044, $10,496 during year 2, and $7307 at year 13. Hart A, Smith JM, Skeans MA, et al. OPTN/SRTR 2016 Annual Data
Report: kidney. Am J Transplant 2018;18(Suppl 1):18–113 https://
3.79 Although ABO-incompatible transplantation comes doi.org/10.111/ajt.14557.
with increased cost, it remains cost effective compared 14. de Klerk M, Witvliet MD, Haase-Kromwijk BJ, Claas FH, Weimar W. A
with maintaining the patient on dialysis while waiting for a highly efficient living donor kidney exchange program for both blood
blood group compatible deceased donor kidney. type and crossmatch incompatible donor-recipient combinations.
Transplantation 2006;82(12):1616–20.
Although paired donation has significantly decreased the 15. Li H, Stegall MD, Dean PG, et al. Assessing the efficacy of kidney paired
need for ABO-incompatible living donor kidney transplan- donation—performance of an integrated three-site program. Trans-
tation, it remains a viable option for some patients. plantation 2014;98(3):300–5.
22 • Transplantation in the Sensitized Recipient and Across ABO Blood Groups 365

16. Gentry SE, Montgomery RA, Segev DL. Kidney paired dona- 41. Loupy A, Suberbielle-Boissel C, Hill GS, et al. Outcome of subclini-
tion: fundamentals, limitations, and expansions. Am J Kidney Dis cal antibody-mediated rejection in kidney transplant recipients with
2011;57(1):144–51. preformed donor-specific antibodies. Am J Transplant 2009;9(11):
17. Montgomery RA, Zachary AA, Ratner LE, et al. Clinical results from 2561–70.
transplanting incompatible live kidney donor/recipient pairs using 42. Worthington JE, Martin S, Al-Husseini DM, Dyer PA, Johnson RW.
kidney paired donation. JAMA 2005;294(13):1655–63. Posttransplantation production of donor HLA-specific antibod-
18. Rees MA, Kopke JE, Pelletier RP, et al. A nonsimultaneous, extended, ies as a predictor of renal transplant outcome. Transplantation
altruistic-donor chain. N Engl J Med 2009;360(11):1096–101. 2003;75(7):1034–40.
19. Segev DL, Gentry SE, Warren DS, Reeb B, Montgomery RA. Kidney 43. Issa N, Cosio FG, Gloor JM, et al. Transplant glomerulopathy: risk and
paired donation and optimizing the use of live donor organs. JAMA prognosis related to anti-human leukocyte antigen class II antibody
2005;293(15):1883–90. levels. Transplantation 2008;86(5):681–5.
20. Wallis CB, Samy KP, Roth AE, Rees MA. Kidney paired donation. 44. Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Evolution and clinical
Nephrol Dial Transplant 2011;26:2091–9. pathologic correlations of de novo donor-specific HLA antibody post
21. Schinstock C, Dean PG, Li H, et al. Desensitization in the era of kidney kidney transplant. Am J Transplant 2012;12(5):1157–67.
paired donation: the Mayo Foundation 3-site experience. Clin Trans- 45. Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Rates and determinants
plant 2013:235–9. of progression to graft failure in kidney allograft recipients with
22. Burns JM, Cornell LD, Perry DK, et al. Alloantibody levels and acute de novo donor-specific antibody. Am J Transplant 2015;15(11):
humoral rejection early after positive crossmatch kidney transplanta- 2921–30.
tion. Am J Transplant 2008;8(12):2684–94. 46. Schinstock CA, Cosio F, Cheungpasitporn W, et al. The value of proto-
23. Gloor JM, DeGoey SR, Pineda AA, et al. Overcoming a positive cross- col biopsies to identify patients with de novo donor-specific antibody
match in living-donor kidney transplantation. Am J Transplant at high risk for allograft loss. Am J Transplant 2017;17(6):1574–84.
2003;3(8):1017–23. 47. Amrouche L, Aubert O, Suberbielle C, et al. Long-term outcomes of
24. Glotz D, Antoine C, Julia P, et al. Desensitization and subsequent kid- kidney transplantation in patients with high levels of preformed
ney transplantation of patients using intravenous immunoglobulins DSA: the Necker high-risk transplant program. Transplantation
(IVIg). Am J Transplant 2002;2(8):758–60. 2017;101(10):2440–8.
25. Montgomery RA, Zachary AA. Transplanting patients with a posi- 48. Orandi BJ, Garonzik-Wang JM, Massie AB, et al. Quantifying the risk
tive donor-specific crossmatch: a single center’s perspective. Pediatr of incompatible kidney transplantation: a multicenter study. Am J
Transplant 2004;8(6):535–42. Transplant 2014;14(7):1573–80.
26. Stegall MD, Gloor J, Winters JL, Moore SB, Degoey S. A comparison 49. Stewart DE, Kucheryavaya AY, Klassen DK, Turgeon NA, Formica RN,
of plasmapheresis versus high-dose IVIG desensitization in renal Aeder MI. Changes in deceased donor kidney transplantation one year
allograft recipients with high levels of donor specific alloantibody. Am after KAS implementation. Am J Transplant 2016;16(6):1834–47.
J Transplant 2006;6(2):346–51. 50. Jordan SC, Lorant T, Choi J, et al. IgG endopeptidase in highly sensitized
27. Stegall MD, Diwan T, Raghavaiah S, et al. Terminal complement inhi- patients undergoing transplantation. N Engl J Med 2017;377(5):442–53.
bition decreases antibody-mediated rejection in sensitized renal trans- 51. Wong W, Lee RA, Saidman SL, Smith RN, Zorn E. Bortezomib in kid-
plant recipients. Am J Transplant 2011;11(11):2405–13. ney transplant recipients with antibody mediated rejection: three case
28. Vo AA, Peng A, Toyoda M, et al. Use of intravenous immune globulin and reports. Clin Transplant 2009:401–5.
rituximab for desensitization of highly HLA-sensitized patients awaiting 52. Sberro-Soussan R, Zuber J, Suberbielle-Boissel C, Legendre C. Bortezo-
kidney transplantation. Transplantation 2010;89(9):1095–102. mib alone fails to decrease donor specific anti-HLA antibodies: 4 case
29. Orandi BJ, Luo X, Massie AB, et al. Survival benefit with kidney reports. Clin Transplant 2009:433–8.
transplants from HLA-incompatible live donors. N Engl J Med 53. Sberro-Soussan R, Zuber J, Suberbielle-Boissel C, et al. Bortezomib
2016;374(10):940–50. as the sole post-renal transplantation desensitization agent does
30. Montgomery RA, Lonze BE, King KE, et al. Desensitization in not decrease donor-specific anti-HLA antibodies. Am J Transplant
HLA-incompatible kidney recipients and survival. N Engl J Med 2010;10(3):681–6.
2011;365(4):318–26. 54. Sberro-Soussan R, Zuberl J, Suberbielle-Boissel C, Legendre C. Bortezo-
31. Orandi BJ, Luo X, King EA, et al. Hospital readmissions following HLA- mib alone fails to decrease donor specific anti-HLA antibodies: even
incompatible live donor kidney transplantation: a multi-center study. after one year post-treatment. Clin Transplant 2010:409–14.
Am J Transplant 2017;18(3):650–8. 55. Woodle ES, Shields AR, Ejaz NS, et al. Prospective iterative trial
32. Manook M, Koeser L, Ahmed Z, et al. Post-listing survival for highly of proteasome inhibitor-based desensitization. Am J Transplant
sensitised patients on the UK kidney transplant waiting list: a matched 2015;15(1):101–18.
cohort analysis. Lancet 2017;389(10070):727–34. 56. Moreno Gonzales MA, Gandhi MJ, Schinstock CA, et al. 32 doses of
33. Bentall A, Cornell LD, Gloor JM, et al. Five-year outcomes in living bortezomib for desensitization is not well tolerated and is associated
donor kidney transplants with a positive crossmatch. Am J Transplant with only modest reductions in anti-HLA antibody. Transplantation
2013;13(1):76–85. 2017;101(6):1222–7.
34. Axelrod D, Lentine KL, Schnitzler MA, et al. The incremental cost of 57. Everly MJ, Everly JJ, Susskind B, et al. Bortezomib provides effective
incompatible living donor kidney transplantation: a national cohort therapy for antibody- and cell-mediated acute rejection. Transplanta-
analysis. Am J Transplant 2017;17(12):3123–30. tion 2008;86(12):1754–61.
35. Haas M, Sis B, Racusen LC, et al. Banff 2013 meeting report: inclusion 58. Tremblay S, Shields AR, Alloway RR, et al. A prospective carfilzomib-
of c4d-negative antibody-mediated rejection and antibody-­associated based desensitization trial: phase 1 results [abstract]. Am J Transplant
arterial lesions. Am J Transplant 2014;14(2):272–83. 2016;16(Suppl. 3) Available online at: https://atcmeetingabstracts.
36. Schinstock CA, Gandhi M, Cheungpasitporn W, et al. Kidney trans- com/abstract/a-prospective-carfilzomib-based-desensitization-trial-
plant with low levels of DSA or low positive B-flow crossmatch: an phase-1-results/ (accessed 25.02.19).
underappreciated option for highly sensitized transplant candidates. 59. Lefaucheur C, Viglietti D, Hidalgo LG, et al. Complement-­activating
Transplantation 2017;101(10):2429–39. https://doi.org/10.1097/ anti-HLA antibodies in kidney transplantation: allograft gene
TP.0000000000001619. expression profiling and response to treatment. J Am Soc Nephrol
37. Lefaucheur C, Loupy A, Hill GS, et al. Preexisting donor-specific HLA 2018;29(2):620–35. https://doi.org/10.1681/ASN.2017050589.
antibodies predict outcome in kidney transplantation. J Am Soc 60. Cornell LD, Schinstock CA, Gandhi MJ, Kremers WK, Stegall MD. Posi-
Nephrol 2010;21(8):1398–406. tive crossmatch kidney transplant recipients treated with eculizumab:
38. Cosio FG, Grande JP, Wadei H, Larson TS, Griffin MD, Stegall MD. outcomes beyond 1 year. Am J Transplant 2015;15(5):1293–302.
Predicting subsequent decline in kidney allograft function from early 61. Vo AA, Zeevi A, Choi J, et al. A phase I/II placebo-controlled trial of
surveillance biopsies. Am J Transplant 2005;5(10):2464–72. C1-inhibitor for prevention of antibody-mediated rejection in HLA
39. El-Zoghby ZM, Stegall MD, Lager DJ, et al. Identifying specific causes of sensitized patients. Transplantation 2015;99(2):299–308.
kidney allograft loss. Am J Transplant 2009;9(3):527–35. 62. Montgomery RA, Orandi BJ, Racusen L, et al. Plasma-derived C1
40. Gloor JM, Cosio FG, Rea DJ, et al. Histologic findings one year after esterase inhibitor for acute antibody-mediated rejection following
positive crossmatch or ABO blood group incompatible living donor kidney transplantation: results of a randomized double-blind placebo-
kidney transplantation. Am J Transplant 2006;6(8):1841–7. controlled pilot study. Am J Transplant 2016;16(12):3468–78.
366 Kidney Transplantation: Principles and Practice

63. Viglietti D, Gosset C, Loupy A, et al. C1 Inhibitor in acute antibody-medi- 76. Stegall MD, Dean PG, Gloor JM. ABO-incompatible kidney transplan-
ated rejection nonresponsive to conventional therapy in kidney trans- tation. Transplantation 2004;78(5):635–40.
plant recipients: a pilot study. Am J Transplant 2016;16(5):1596–603. 77. Fuchinoue S, Ishii Y, Sawada T, et al. The 5-year outcome of ABO-
64. Stegall M, Cornell L, Park MJ, Kremers W, Smith B. Cosio F. Renal incompatible kidney transplantation with rituximab induction.
allograft histology at 10 years after transplantation in the tacro- Transplantation 2011;91(8):853–7.
limus era: evidence of pervasive chronic injury. Am J Transplant 78. Shimmura H, Tanabe K, Ishida H, et al. Lack of correlation between
2018;18(1):180–8. https://doi.org/10.1111/ajt.14431. results of ABO-incompatible living kidney transplantation and anti-
65. Stegall MD, Morris RE, Alloway RR, Mannon RB. Developing new ABO blood type antibody titers under our current immunosuppres-
immunosuppression for the next generation of transplant recipients: sion. Transplantation 2005;80(7):985–8.
the path forward. Am J Transplant 2016;16(4):1094–101. 79. Axelrod D, Segev DL, Xiao H, et al. Economic impacts of ABO-­
66. Perry DK, Pollinger HS, Burns JM, et al. Two novel assays of alloanti- incompatible live donor kidney transplantation: a national study of
body-secreting cells demonstrating resistance to desensitization with medicare-insured recipients. Am J Transplant 2016;16(5):1465–73.
IVIG and rATG. Am J Transplant 2008;8(1):133–43. 80. Galliford J, Charif R, Chan KK, et al. ABO incompatible living renal
67. Slifka MK, Antia R, Whitmire JK, Ahmed R. Humoral immunity due to transplantation with a steroid sparing protocol. Transplantation
long-lived plasma cells. Immunity 1998;8(3):363–72. 2008;86(7):901–6.
68. Stegall MD, Dean PG, Gloor J. Mechanisms of alloantibody pro- 81. Genberg H, Kumlien G, Wennberg L, Berg U, Tyden G. ABO-incom-
duction in sensitized renal allograft recipients. Am J Transplant patible kidney transplantation using antigen-specific immunoad-
2009;9(5):998–1005. sorption and rituximab: a 3-year follow-up. Transplantation 2008;
69. Perry DK, Burns JM, Pollinger HS, et al. Proteasome inhibition causes 85(12):1745–54.
apoptosis of normal human plasma cells preventing alloantibody pro- 82. Gloor JM, Lager DJ, Moore SB, et al. ABO-incompatible kidney trans-
duction. Am J Transplant 2009;9(1):201–9. plantation using both A2 and non-A2 living donors. Transplantation
70. Diwan TS, Raghavaiah S, Burns JM, Kremers WK, Gloor JM, Stegall 2003;75(7):971–7.
MD. The impact of proteasome inhibition on alloantibody-producing 83. Montgomery JR, Berger JC, Warren DS, James NT, Montgomery RA,
plasma cells in vivo. Transplantation 2011;91(5):536–41. Segev DL. Outcomes of ABO-incompatible kidney transplantation in
71. Venner JM, Hidalgo LG, Famulski KS, Chang J, Halloran PF. The the United States. Transplantation 2012;93(6):603–9.
molecular landscape of antibody-mediated kidney transplant rejec- 84. Tanabe K, Takahashi K, Sonda K, et al. Long-term results of ABO-
tion: evidence for NK involvement through CD16a Fc receptors. Am J incompatible living kidney transplantation: a single-center experi-
Transplant 2015;15(5):1336–48. ence. Transplantation 1998;65(2):224–8.
72. Hidalgo LG, Sis B, Sellares J, et al. NK cell transcripts and NK cells in 85. Park WD, Grande JP, Ninova D, et al. Accommodation in ABO-
kidney biopsies from patients with donor-specific antibodies: evidence incompatible kidney allografts, a novel mechanism of self-protec-
for NK cell involvement in antibody-mediated rejection. Am J Trans- tion against antibody-mediated injury. Am J Transplant 2003;
plant 2010;10(8):1812–22. 3(8):952–60.
73. Kohei N, Tanaka T, Tanabe K, et al. Natural killer cells play a critical 86. Salama AD, Delikouras A, Pusey CD, et al. Transplant accommoda-
role in mediating inflammation and graft failure during antibody-medi- tion in highly sensitized patients: a potential role for Bcl-xL and allo-
ated rejection of kidney allografts. Kidney Int 2016;89(6):1293–306. antibody. Am J Transplant 2001;1(3):260–9.
74. Akiyoshi T, Hirohashi T, Alessandrini A, et al. Role of comple- 87. West LJ. ABO-incompatible hearts for infant transplantation. Curr
ment and NK cells in antibody mediated rejection. Hum Immunol Opin Organ Transplant 2011;16(5):548–54.
2012;73(12):1226–32. 88. Schwartz J, Stegall MD, Kremers WK, Gloor J. Complications, resource
75. Hirohashi T, Chase CM, Della Pelle P, et al. A novel pathway of chronic utilization, and cost of ABO-incompatible living donor kidney trans-
allograft rejection mediated by NK cells and alloantibody. Am J Trans- plantation. Transplantation 2006;82(2):155–63.
plant 2012;12(2):313–21.
23 Paired Exchange Programs
for Living Donors
SOMMER ELIZABETH GENTRY and DORRY L. SEGEV

CHAPTER OUTLINE Introduction Nondirected Donors and Kidney Donor


Finding Exchange Opportunities Chains
Legality, Anonymity, Travel, and Expenses Outlook
Matching Algorithms

Introduction exchanges comprised more than 10% of living kidney dona-


tion at one Korean center.13 Korea’s approach emphasized
There are far more eligible kidney transplant candidates world- close human leukocyte antigen (HLA) matching between
wide than there are organs available. Kidney exchange, also donors and recipients. Living donors were only accepted if
referred to as paired kidney donation, has become strongly they shared more than one DR antigen or two of four A/B anti-
established over the past decade as a modality for facilitating gens with their recipients. Of pairs deemed incompatible, 30%
living kidney donation by donors who are not compatible with had HLA mismatching beyond these criteria, 65% were blood
their intended recipients.1–3 Instead of donating directly to the type incompatible, and only 5% had a positive crossmatch
intended recipient, candidate-donor pairs are matched together test.14 Compared with other programs that allow more HLA
to participate in a mutually beneficial exchange of organs. This mismatch, this restriction may have directed more candidate/
requires a coordinating effort to collect data about incompat- donor pairs into the exchange group, but may have ruled out
ible pairs and about other types of participants like nondirected more potential exchanges.
(altruistic) donors and compatible pairs, and to select appropri-
ate matches4 by a manual or algorithmic process. Legality, Anonymity, Travel, and
In this chapter, we examine the common features and the
idiosyncrasies of several successful paired exchange efforts Expenses
around the world5: in Korea, the Netherlands, Canada, Aus-
tralia, India, the UK, the US, and other countries. Review- Legal restrictions on living organ donation have played a
ing these differences may be reassuring for groups working significant role in the evolution of kidney exchange in many
to implement paired exchange, because for each potential countries. For a time in the US, people took care to refer to
snag that might have delayed or limited one program’s “kidney paired donation” rather than kidney exchange
efforts, there are counterexamples in which that aspect of because of concerns about whether the National Organ
paired exchange was not an impediment.6 Transplantation Act’s (NOTA’s) prohibition on giving valu-
able consideration to living donors encompassed a ban on
exchanging one kidney for another. In 2007 the US enacted
Finding Exchange Opportunities a new law clarifying that paired exchange had never been
illegal under NOTA.15 Similar restrictions in Australia and
In every kidney exchange program, some incompatible pairs Canada have also been a concern.15 In the UK only relatives
will fail to find an exchange opportunity. Because blood group and those with strong emotional ties to the candidate were
O donors are most likely to be compatible with their recipients, permitted to be living organ donors, until legislation allow-
there will be a blood group imbalance, with too few O donors ing kidney exchange was enacted in 2006.16 In India, some
for too many O recipients in any population of incompatible legal restrictions have been streamlined but paired donors
pairs.7 Populations of incompatible pairs also have larger num- must be first-degree relatives.17
bers of highly sensitized candidates, who are difficult to match The usual practice in the US is to maintain strict anonym-
in an exchange. This imbalance limits exchange opportunities ity among the exchange participants before surgery, with
and leads to innovations like the inclusion of compatible pairs8 meetings possible after surgery if desired by all parties.18 In
and nondirected donors,9 extending match arrangements to the Netherlands, patients in a pilot study expressed a strong
three or four or more pairs,10 and desensitization as part of the preference for anonymity, so that country’s exchange pro-
exchange program,11 all of which can enhance the proportion gram maintains anonymity.19 In Germany, anonymous
of incompatible pairs successfully transplanted.12 paired kidney exchange is prohibited, so exchanges can
Paired exchange may have the longest history in Korea, only proceed after incompatible pairs meet one another.20
which established an exchange program in 1991. By 2003 In Romania, a single-center program transplanted 56 pairs
367
368 Kidney Transplantation: Principles and Practice

between 2001 and 2005.21 This group felt that maintaining D1 R1 NDD
anonymity among pairs involved in a paired donation was
unnecessary and would be too logistically difficult, so they D2 R2
D1 R1
encouraged open interaction before and after the transplants.
In the Netherlands, a compact country that is densely D2 R2
Two-way kidney paired donation
populated, donors travel to the transplant center where their
paired recipients are being cared for.5 In the US, which has Time passes
a much larger geographic footprint, donor organs are rou- D1 R1 D3 R3
tinely transported to the recipient center.22 US physicians
became more comfortable with transporting organs for kid- D2 R2 D4 R4
ney paired donations after a study evaluating delayed trans-
plants of other live donor kidneys showed that up to 8 hours D3 R3
Time passes
of cold ischemia time did not have negative repercussions Open chain
for a live donor kidney.23 In Canada, transporting live donor Three-way kidney paired
kidneys would be a logical response to far-flung paired dona- donation NDD
tion matches, but regulations and the risky logistics of travel
there have limited such transports.24 In Australia, most kid- D1 R1
neys are transported despite the challenges of coordinating NDD D2 R2
the transports and of prolonged cold ischemia time.25
In the US financial arrangements are frequently a compli- D1 R1 Time passes
cating factor in kidney paired donation. There is no obvious
D3 R3
party responsible for paying the expenses incurred in donor D2 R2
evaluations for incompatible donors, many of whom never D4 R4
donate, nor for the tissue typing and administrative costs Waitlist
Waitlist
of maintaining a paired donation registry to locate suitable
matches. Some have called for extending the concept of Domino paired donation Closed chain
the standard acquisition charge, used in the US to recoup Fig. 23.1 Terminology for various forms of paired donation. Pictured
expenses related to acquiring deceased donor organs, to are nondirected, or altruistic donors (NDD), recipients (R) who are
kidney paired donation.26 paired with their incompatible donors (D), and waitlist recipients, in a
variety of paired donation arrangements.

Matching Algorithms decisions. For instance, it is not optimal to rank all possible
matches and then choose the highest ranked match first, the
Deciding which incompatible pairs, compatible pairs, and next highest ranked available match second, and so on. This
nondirected donors should be matched together for a kid- match-rank heuristic, which has been used in some opera-
ney exchange is critically important for any program. Each tional registries,30 might transplant fewer people than would
donor should usually be blood group and tissue type com- be possible with better matching algorithms.
patible with the candidate to whom they will donate. Char- There are only two correct approaches to find a set of
acterizing recipient unacceptable antigens and allowing exchanges with the largest number of transplants for two-
centers to define their own criteria for unacceptability helps and three-way exchanges among a specific list of participants:
decrease the number of proposed matches that fail because exhaustive search or integer programming (optimization). In
of an unexpected positive crossmatch.27 Some transplant exhaustive search, a computer generates every possible set
centers have experience with desensitization protocols for of matches and then compares them to select the best set of
ABO-incompatible or HLA-incompatible transplants, or matches, as in the Korean registry.31 Exhaustive search is only
both. These centers may wish to combine desensitization possible for small registries, because the number of different
with kidney exchange to obtain transplants for their hard- sets of matches possible grows incredibly quickly. Integer pro-
est-to-match patients.11,28 gramming models, including many algorithms specialized for
Beyond simply understanding which donors could suc- the paired donation problem,32 use mathematical techniques
cessfully donate to which candidates in a kidney paired to locate and verify the best set of matches without having to
donation registry, matching participants together so that explicitly describe all possible sets of matches.
the largest number of people can achieve the greatest ben- However, recent evidence suggests that in actual reg-
efit requires sophisticated mathematics and specialized opti- istries more matches can be made by matching with very
mization algorithms.4,29 We refer to a “match” as a single high frequency than by improving the match algorithm.33
two-way, three-way, or chain of transplants, as in Fig. The advantage of optimization is only relevant when regis-
23.1. For a fixed pool of participants in a paired donation tries wait for many incompatible pairs to accumulate and
registry, “matching” means selecting a nonoverlapping set perform matching infrequently. Ashlagi et al.33 find that
of matches to proceed to transplant. frequent matching is the best strategy for matching, when
The decisions in a paired exchange registry are necessar- pairs might depart after being asked to wait too long.
ily interconnected, because choosing one match means that By far the most significant factor in increasing the fraction
other potentially useful matches involving any of those same of incompatible pairs matched is increasing the number of
participants will not be possible. Some seemingly effective participants in a registry. All candidates benefit from increas-
matching heuristics neglect the interconnectedness of these ing pool size, but the benefit is particularly pronounced for
23 • Paired Exchange Programs for Living Donors 369

sensitized candidates. For instance, a nationwide registry in 100% AB AB AB


the US would create a sixfold increase in kidney exchange B B
opportunities for sensitized candidates.4 B
The advantage of large-scale programs over small ones 75% AB
has implications for the organization of registries around A
A
the world. Canada, the UK, and the Netherlands, each with
population in the tens of millions, have established national A
kidney exchange registries.16,34,35 In South Korea and Tur- 50%
key, kidney exchange is conducted within single-center B

programs.5,36 One single-center program in the US, by rely- O


ing on compatible pairs, has grown its exchange program 25% O
to be more than one-third of its live donor kidney volume.37 A
O
In the US there are many models competing: there are sin-
gle-center programs, several multicenter consortia, and an 0%
effort by the United Network for Organ Sharing (UNOS) to Recipients NDD Paired donors Bridge donors
establish a national kidney exchange registry integrated
Fig. 23.2 Distribution of blood groups among recipients of incompat-
with the national deceased donor allocation system.38 The ible pairs, nondirected donors (NDD), the donors of incompatible pairs,
various registries in the US serve different but overlapping and bridge donors in extended chains. (Reprinted with permission from
populations, because many incompatible pairs are reg- American Journal of Transplantation.)
istered for more than one of these. Unfortunately, fewer
incompatible pairs and many fewer sensitized candidates
will find matches if participants are divided into smaller
pools than if the participants were in one larger pool. Nondirected donors are particularly valuable in paired
donation because nondirected donors are not paired with
one particular candidate who must receive a kidney, eas-
Nondirected Donors and Kidney ing the reciprocal compatibility requirement. Also, blood
group O donors are more prevalent among nondirected
Donor Chains donors than paired donors (see Fig. 23.2). In programs
where nondirected donors have been incorporated into
In arrangements referred to as chains or a domino paired KPD, nondirected donor chains often comprise a majority of
donations, a nondirected or altruistic donor starts a the transplants arranged. In the Netherlands, nondirected
sequence of kidney paired donations by donating to a paired donors are only matched to incompatible pairs after the
recipient whose donor then becomes available for the next pairs have tried to find a traditional kidney exchange with
recipient (see Fig. 23.1). At the end of the sequence, one other pairs. Also, nondirected donors in the Netherlands
incompatible pair’s donor is left unmatched. This donor are allocated on a center basis rather than by the national
might donate immediately to a recipient on the deceased program.42
donor waiting list, creating a closed chain (domino paired
donation). The last donor might instead wait between a
few months and a year as a bridge donor, before donating Outlook
to another incompatible pair to begin another sequence
of donations (open chain, or nonsimultaneous extended For small European countries, an encouraging develop-
altruistic donor chain). ment is cross-border exchange.43 In June of 2012, a couple
There has been some debate in the literature about in Greece became part of a kidney paired donation chain
whether open chains or closed chains will facilitate more in the US, after Greek law was amended to allow unrelated
transplants. Open chains hold a promise of indefinite con- organ donors and permit kidney paired donation for the
tinuation that is intuitively appealing, but in practice, first time. There have been calls for establishing interna-
bridge donors have a blood group distribution (Fig. 23.2) tional kidney paired donation systems to facilitate the flow
that makes them hard to match to recipients.39 Also, in one of organs around the world.44 Another innovation that has
study of a large US registry, about 5% of bridge donors did yet to be explored is transorgan paired exchange, in which
not in fact donate.40 Most bridge donors who did not donate one donor would donate a kidney and the other donor
had a medical issue that prevented donation or their kid- would donate part of a liver.45 We are optimistic that inno-
ney was declined by the recipient’s surgeon, whereas about vations in paired donation will allow even more candidates
1.5% of bridge donors elected not to proceed with donation with incompatible donors to benefit from transplantation.
after their recipients had been transplanted.
The National Kidney Registry in the US also has an References
advanced donation program, in which a donor donates 1. Segev DL, Kucirka LM, Gentry SE, Montgomery RA. Utilization and
their kidney before any match has been identified for their outcomes of kidney paired donation in the United States. Transplanta-
intended recipient.41 This might be necessary if the donor’s tion 2008;86(4):502–10.
time window for donating is limited or if the recipient is not 2. Gentry SE, Montgomery RA, Segev DL. Kidney paired dona-
tion: fundamentals, limitations, and expansions. Am J Kidney Dis
yet ready for donation, for instance an older donor with 2011;57(1):144–51.
a young recipient who is expected to need a kidney one 3. Gentry S, Segev DL. Living donor kidney exchange. Clin Transplant
decade later. 2011:279–86.
370 Kidney Transplantation: Principles and Practice

4. Segev DL, Gentry SE, Warren DS, Reeb B, Montgomery RA. Kidney 26. Rees MA, Schnitzler MA, Zavala EY, et al. Call to develop a standard
paired donation and optimizing the use of live donor organs. JAMA acquisition charge model for kidney paired donation. Am J Transplant
2005;293(15):1883–90. 2012;12(6):1392–7.
5. Ferrari P, de Klerk M. Paired kidney donations to expand the living 27. Baxter-Lowe LA, Cecka M, Kamoun M, Sinacore J, Melcher ML.
donor pool. J Nephrol 2009;22(6):699–707. Center-defined unacceptable HLA antigens facilitate transplants for
6. Melcher ML, Blosser CD, Baxter-Lowe LA, et al. Dynamic challenges sensitized patients in a multi-center kidney exchange program. Am J
inhibiting optimal adoption of kidney paired donation: findings of a Transplant 2014;14(7):1592–8.
consensus conference. Am J Transplant 2013;13(4):851–60. 28. Ferrari P, Hughes PD, Cohney SJ, Woodroffe C, Fidler S, D’Orsogna L.
7. Gentry SE, Segev DL, Montgomery RA. A comparison of popula- ABO-incompatible matching significantly enhances transplant rates
tions served by kidney paired donation and list paired donation. Am in kidney paired donation. Transplantation 2013;96(9):821–6.
J Transplant 2005;5(8):1914–21. 29. Gentry SE. Optimization over graphs for kidney paired donation. In:
8. Gentry SE, Segev DL, Simmerling M, Montgomery RA. Expanding kid- Lim G, editor. Optimization in medicine and biology. Milton Park, UK:
ney paired donation through participation by compatible pairs. Am J Taylor & Francis/CRC Press; 2008:177–96.
Transplant 2007;7(10):2361–70. 30. Keizer KM, de Klerk M, Haase-Kromwijk BJ, Weimar W. The Dutch
9. Rees MA, Kopke JE, Pelletier RP, et al. A nonsimultaneous, extended, algorithm for allocation in living donor kidney exchange. Transplant
altruistic-donor chain. N Engl J Med 2009;360(11):1096–101. Proc 2005;37(2):589–91.
10. Saidman SL, Roth AE, Sonmez T, Unver MU, Delmonico FL. Increasing 31. Kim BS, Kim YS, Kim SI, et al. Outcome of multipair donor kidney
the opportunity of live kidney donation by matching for two and three exchange by a web-based algorithm. J Am Soc Nephrol 2007;18(3):
way exchanges. Transplantation 2006;81(5):773–82. 1000–6.
11. Montgomery RA. Living donor exchange programs: theory and prac- 32. Abraham DJ, Blum A, Sandholm T. Clearing algorithms for barter
tice. Br Med Bull 2011;98:21–30. exchange markets: enabling nationwide kidney exchanges. Proceed-
12. Flechner SM, Thomas AG, Ronin M, et al. The first 9 years of kidney ings of the 8th ACM conference on electronic commerce. San Diego,
paired donation through the National Kidney Registry: characteris- CA: ACM; 2007.
tics of donors and recipients compared with national live donor trans- 33. Ashlagi I, Bingaman A, Burq M, et al. Effect of match-run frequencies
plant registries. Am J Transplant 2018;18(11):2730–8. https://doi. on the number of transplants and waiting times in kidney exchange.
org/10.1111/ajt.14744. Am J Transplant 2018;18(5):1177–86.
13. Park K, Lee JH, Huh KH, Kim SI, Kim YS. Exchange living-donor kid- 34. de Klerk M, Keizer KM, Claas FH, Witvliet M, Haase-Kromwijk BJ, Wei-
ney transplantation: diminution of donor organ shortage. Transplant mar W. The Dutch national living donor kidney exchange program.
Proc 2004;36(10):2949–51. Am J Transplant 2005;5(9):2302–5.
14. Huh KH, Kim MS, Ju MK, et al. Exchange living-donor kidney transplan- 35. Cole EH, Nickerson P, Campbell P, et al. The Canadian kidney paired
tation: merits and limitations. Transplantation 2008;86(3):430–5. donation program: a national program to increase living donor trans-
15. Toews M, Giancaspro M, Richards B, Ferrari P. Kidney paired plantation. Transplantation 2015;99(5):985–90.
donation and the “valuable consideration” problem: the experi- 36. Yucetin L, Tilif S, Kececioglu N, et al. Paired exchange kidney trans-
ences of Australia, Canada, and the United States. Transplantation plantation experience of Turkey. Transplant Proc 2013;45(3):860–3.
2017;101(9):1996–2002. 37. Bingaman AW, Wright Jr FH, Kapturczak M, Shen L, Vick S, Murphey
16. Johnson RJ, Allen JE, Fuggle SV, Bradley JA, Rudge C. Early experience CL. Single-center kidney paired donation: the Methodist San Antonio
of paired living kidney donation in the United Kingdom. Transplan- experience. Am J Transplant 2012;12(8):2125–32.
tation 2008;86(12):1672–7. Available online at: https://www.doi. 38. Akkina SK, Muster H, Steffens E, Kim SJ, Kasiske BL, Israni AK. Donor
org/10.1097/TP.0b013e3181901a3d. exchange programs in kidney transplantation: rationale and opera-
17. Kute VB, Agarwal SK, Sahay M, et al. Kidney-paired donation to tional details from the north central donor exchange cooperative. Am
increase living donor kidney transplantation in India: guidelines of J Kidney Dis 2011;57(1):152–8.
Indian Society of Organ Transplantation 2017. Indian J Transplant 39. Gentry SE, Montgomery RA, Swihart BJ, Segev DL. The roles of domi-
2017;12(1):67–74. nos and nonsimultaneous chains in kidney paired donation. Am J
18. Montgomery RA, Zachary AA, Ratner LE, et al. Clinical results from Transplant 2009;9(6):1330–6.
transplanting incompatible live kidney donor/recipient pairs using 40. Cowan N, Gritsch HA, Nassiri N, Sinacore J, Veale J. Broken chains
kidney paired donation. JAMA 2005;294(13):1655–63. and reneging: a review of 1748 kidney paired donation transplants.
19. Kranenburg LW, Visak T, Weimar W, et al. Starting a crossover kid- Am J Transplant 2017;17(9):2451–7.
ney transplantation program in the Netherlands: ethical and psycho- 41. Flechner SM, Leeser D, Pelletier R, et al. The incorporation of an
logical considerations. Transplantation 2004;78(2):194–7. advanced donation program into kidney paired exchange: ini-
20. Giessing M, Deger S, Roigas J, et al. Cross-over kidney transplantation tial experience of the National Kidney Registry. Am J Transplant
with simultaneous laparoscopic living donor nephrectomy: initial 2015;15(10):2712–7.
experience. Eur Urol 2008;53(5):1074–8. 42. Roodnat JI, Zuidema W, van de Wetering J, et al. Altruistic donor trig-
21. Lucan M. Five years of single-center experience with paired kidney gered domino-paired kidney donation for unsuccessful couples from
exchange transplantation. Transplant Proc 2007;39(5):1371–5. the kidney-exchange program. Am J Transplant 2010;10(4):821–7.
22. Segev DL, Veale JL, Berger JC, et al. Transporting live donor kidneys 43. Bohmig GA, Fronek J, Slavcev A, Fischer GF, Berlakovich G, Viklicky O.
for kidney paired donation: initial national results. Am J Transplant Czech-Austrian kidney paired donation: first European cross-border
2011;11(2):356–60. living donor kidney exchange. Transpl Int 2017;30(6):638–9.
23. Simpkins CE, Montgomery RA, Hawxby AM, et al. Cold ischemia time 44. Connolly JS, Terasaki PI, Veale JL. Kidney paired donation—the next
and allograft outcomes in live donor renal transplantation: is live step. N Engl J Med 2011;365(9):868–9.
donor organ transport feasible? Am J Transplant 2007;7(1):99–107. 45. Samstein B, de Melo-Martin I, Kapur S, Ratner L, Emond J. A liver
24. McGregor T, Sener A, Paraskevas S, Reikie B. Kidney paired donation for a kidney: ethics of trans-organ paired exchange. Am J Transplant
and the unique challenges of kidney shipment in Canada. Can J Surg 2018;18(5):1077–82.
2018;61(2):139–40.
25. Allen RDM, Pleass HCC, Woodroffe C, Clayton PA, Ferrari P. Challenges
of kidney paired donation transplants involving multiple donor and
recipient surgeons across Australia. ANZ J Surg 2018;88(3):167–71.
24 Kidney Allocation
DIANA A. WU, JAYME E. LOCKE, and JOHN L. R. FORSYTHE

CHAPTER OUTLINE Two-Step Process Other Variations


General Points of Allocation Eurotransplant
United Kingdom Australia
United States Summary

It is a favorite assertion of the authors that clinical trans- Although this is a clear principle, the secondary question of
plantation is an excellent example of “medical ethics in how to measure the benefit is much more difficult to answer.
practice.” Perhaps this is best demonstrated by the processes Should this be based on survival statistics (kidney or patient),
of organ allocation where a balance must be struck between comparative benefit of life years gained, or should we include
allocation that makes best use of a scarce resource and the quality of life for each of the possible patients? In addition,
principle of equitable access to all on the waiting list.1 an allocation system based purely on utility causes disad-
vantage to many patient groups—the elderly, diabetics, and
those who have waited for a longer time.4,5
Two-Step Process On the other hand, equity, the concept that each patient
should have an equal chance of receiving an offer, will
It is important to acknowledge that, for the patient to result in a system that does not maximize the precious gift
receive an offer of a deceased donor kidney, they must have of a donor organ. For instance, a “queuing” or “first-come
completed a two-step process. First, the patient must gain first-served” system gives priority to more elderly patients
access to the waiting list. The preparation for listing and and those who have been on dialysis longer with more
the criteria for this are covered elsewhere (see Chapter 4), comorbidity, which mitigates against the best results.6
but this is an important step and there is evidence of ineq- Most allocation systems around the world were built on
uity in clinical processes to screen patients for access to the human leukocyte antigen (HLA) matching between donor
waiting list.2,3 This chapter will concentrate on the second and recipient. This stands to reason as a better match pro-
step, which is allocation of a donor kidney to a patient who duces better survival.7,8 However, the unintended conse-
is already on the waiting list. quence of this allocation principle is that patients with rare
HLA types (who also happen to be ethnic minorities in any
community) and sensitized patients (who also happen to be
General Points of Allocation mainly female) are disadvantaged.9
It follows that allocation systems mature to be an amal-
As transplantation from deceased donors develops in gam between markers of utility and factors associated with
any particular country, the sophistication of allocation fairness (Table 24.1).
processes also matures. Early allocation plans may begin Age is also a controversial issue in allocation policies.
with a local clinician picking from a list of local patients, Few would deny priority for pediatric patients because of
a system that is subjective, open to challenge, and does the natural emotional response to help children, backed by
not maximize population size for matching. Allocation the clinical issues peculiar to children with renal failure,
algorithms then develop regionally, and eventually result including growth and developmental delay. Yet some sys-
in a complex process that is usually run on a national tems have a strict cutoff between designation of a patient as
basis by a designated organization. On occasion, an a pediatric or adult, which seems harsh on the young per-
international collaboration is set up—perhaps the most son who has only strayed 1 day into the adult sector, los-
notable example of this is Eurotransplant, which covers ing all priority in the process. In addition, any priority for
the nations of Germany, Belgium, Austria, Netherlands, younger patients (even though utility would suggest that
Luxembourg, Slovenia, Croatia, and Hungary. The more they will benefit to a greater extent) is often resisted and can
advanced allocation schemes are safeguarded by legisla- be characterized as age discrimination. Some have advo-
tive frameworks and appropriate governance processes. cated that the allocation of younger donor kidneys should
The ethical dilemma in the design of allocation policies faced be reserved for younger recipients (obviously accompanied
by all countries is the balance between utility and equity. by older donor kidneys allocated to older recipients) because
Utility-based allocation gives priority to those with the this treats all patients in an age-matched fashion.10 The
best chance of a favorable outcome—by this method gain- most modern schemes attempt to predict the likely longev-
ing the maximum benefit from every transplanted kidney. ity of the transplanted kidney, advocating better matched
371
372 Kidney Transplantation: Principles and Practice

TABLE 24.1 Allocation Criteria


Allocation Criteria UK US Eurotransplant Australia
HLA MM loci/importance DR > B DR only DR = B = A DR > B/A
Waiting time definition Listing date Start of dialysis Start of dialysis Start of dialysis
Definition of pediatric <18 years, retain pediatric <18 years <16 years or >16 yrs with <18 years, first dialysis <17
recipient priority until growth potential on years and on dialysis for
transplanted x-ray >1 yr
Recipient age (adults) + + − −
Donor-recipient age matching + − + −
Definition of highly sensitized CRF ≥85% Sliding scale for CPRA PRA >85% PRA >50% for 000 MM, >80%
recipient 20–100 for all other MM levels
Priority for HLA homozygous DR, B − DR, B, A −
recipients
Local allocation priority + + + +
Balance of exchange − − + +
Other allocation criteria/ Defaulting of rare HLA Priority for previous Medical urgency Around 80% of kidneys allo-
features antigens living organ donors AMP cated within donor state
HLA match and age EPTS ESP via state-based algorithms
combined KDPI

AMP, acceptable mismatch programme; CPRA, calculated panel reactive antibody; CRF, calculated reaction frequency; EPTS, estimated post transplant
survival score; ESP, Eurotransplant seniors program; HLA, human leukocyte antigen; KDPI, kidney donor profile index; MM, mismatch; PRA, panel reac-
tive antibody.
Sources: UK: NHS Blood and Transplant policy 186/9, effective 26/01/18. Available online at: https://nhsbtdbe.blob.core.windows.net/umbraco-assets-
corp/6522/pol186-kidney-transplantation-deceased-donor-organ-allocation.pdf.
US: Organ Procurement and Transplantation Network policy 8, effective 08/10/17. Available online at: https://optn.transplant.hrsa.gov/media/1200/optn_
policies.pdf#nameddest=Policy_08.
Eurotransplant: Eurotransplant foundation manual, Chapter 4, version 5.2, effective 11/11/16. Available online at: https://www.eurotransplant.org/cms/m
ediaobject.php?file=H4+ETKAS+November+11%2C+20161.pdf.
Australia: The Transplantation Society of Australia and New Zealand, version 1.1, effective 17/05/17. Available online at: https://www.tsanz.com.au/organa
llocationguidelines/documents/ClinicalGuidelinesV1.1May2017.pdf.

United Kingdom
The first UK national scheme began in 1989 with a
simple HLA match allocation.12 This was still a hybrid
scheme in that one kidney from each donor was allocated
nationally, whereas the paired kidney was allocated
locally, governed by individual center policies. This was
revised in 1998 after analysis of outcome had shown
three distinct tiers of HLA mismatch as factors in graft
survival.13 Both kidneys were allocated on a national
basis in the 1998 scheme, which also introduced a num-
ber of novel allocation tools including a points score to
differentiate equally well-matched (or equally poorly
matched) patients within each tier.5 The points were
based on donor recipient age difference, recipient age,
Fig. 24.1 Marginal donor kidney.
waiting time, matchability score, sensitization level,
and, as a compromise to those centers who wished to
kidneys for those who are younger, and accepting a more maintain a local organ allocation flavor, points for bal-
poorly matched kidney for those patients who, on balance, ance of organ exchange among centers. Matchability
have fewer years to survive.11 was a measure of the probability of a particular patient
Finally, the characteristics of donors have changed rap- being offered a well-matched kidney—this was a cor-
idly in recent years. More aged, higher body mass index rection factor for those patients who were “difficult to
(BMI), and more comorbidity in donors has been well match.”
demonstrated, resulting in terms such as “marginal” or This scheme was analyzed around 8 years later and
“extended criteria” donor (Figs. 24.1 and 24.2). The fair inequity of access, as an unintended consequence of the
method of allocation that also gives maximum benefit of dominance of HLA matching, was seen to be a significant
these donor kidneys provides a significant challenge for problem.13 As a result a new allocation scheme was derived
national transplant organizations. and introduced in 2006, which is still in place at the time of
Although these themes are universal, the national response this writing.14
has been varied depending on the prevailing clinical, patient, The new scheme placed less emphasis on HLA matching
and political views.11 We will describe here two notable although zero HLA mismatched patients retained top prior-
schemes and summarize the variation in some others. ity and well-matched patients (100, 10, and 110) also had
24 • Kidney Allocation 373

1600

1400

1200
70 or over
Number of donors

1000 60–69
50–59
800 18–49
0–17
600

400

200

0
2006/07 2007/08 2008/09 2009/10 2010/11 2011/12 2012/13 2013/14 2014/15 2015/16
Fig. 24.2 Age of deceased donors in the UK.

some level of priority. Highly sensitized patients were also as determined by a donor risk index16 and the perceived
seen to benefit. For each patient a “calculated reaction fre- risk associated with a particular recipient. In this man-
quency” was measured. This was defined as the percentage ner simulations suggest that the potential of a particular
of 10,000 recent donors to which the patient has preformed kidney can be better matched to the needs of the allo-
antibodies and when this rose to over 85%, the patient cated recipient.
was deemed highly sensitized and gained priority. A novel
approach was developed to tackle the inequity of access
experienced by patients with rare HLA types, whereby United States
rare HLA antigens (with a frequency of less than 2% in
the donor pool) were defaulted to close counterparts based Although the number of kidney transplants performed in
on serologic reactivity data. For instance, the rare antigen the US has increased over the years, there remains a critical
B82 was defaulted to B12, opening up 18% of the donor organ shortage. In fact, as of September 2017, more than
pool. The points score was also adapted to give much more 96,000 people were waiting for a deceased donor kidney in
weight to waiting time, as a result of patient input to the the US; yet fewer than 20,000 receive a kidney transplant
design of the scheme. Finally, points for recipient age were each year.17
combined with the HLA match to give the best matched kid- Efforts to address this critical shortage and improve
neys to younger patients. organ matching and placement resulted in the US
It is important to note that the cold ischemic time (CIT) was Congress passing the National Organ Transplant Act
also monitored as the new scheme came in. A surrogate for this (NOTA) in 1984. NOTA (1) established the Organ Pro-
factor—proximity of the donor to the recipient center—was curement and Transplantation Network (OPTN) to
included in the points score and, gratifyingly, the CIT actually maintain a national registry for organ matching, and
decreased over the introduction of the 2006 scheme.14 (2) required the OPTN to be operated by a private, non-
In 2014 the scheme was extended to kidneys donated profit organization under federal contract, effectively
from donors after circulatory death (DCD), using the same creating the United Network for Organ Sharing (UNOS;
algorithm but based on regional rather than fully national the OPTN contractor). The goal of US allocation policy
allocation, to minimize CIT.11,15 has been to balance utility and equity regarding the dis-
The 2006 scheme was reviewed and found to have tribution of deceased donor organs. Not surprisingly,
increased the number of transplants for long-waiting and the policy has changed over time to optimize alloca-
highly sensitized patients (Fig. 24.3). Although the percent- tion efforts aimed at balancing these often competing
age of difficult to match and minority ethnic patients trans- goals.18
planted had also increased, significant inequity remained, Initially, US allocation policy focused heavily on donor
reflecting the immense challenge of achieving a good bal- and recipient HLA matching; over time, however, advances
ance between utility and equity. in immunosuppression regimen resulted in lower acute
Work began on a new allocation algorithm in 2016 rejection rates, and in 1995 allocation points awarded
and it is hoped that the new scheme will be introduced based on HLA-A matching were eliminated and more
in 2019. It is designed to further improve inequities of emphasis was placed on waiting time. The concept of first
access, particularly to highly sensitized or difficult to come, first transplanted was introduced.18
match patients. In addition, it is planned that there will However, HLA subtype matching was not completely
be better matching between the risk of a donor kidney abandoned from the allocation policy. Secondary to
374 Kidney Transplantation: Principles and Practice

100
Pre 2006 scheme Post 2006 scheme
90 981

80
824
70
% DBD kidney transplants

60

50 538
512 497
441 429
40
392
370
30 326

236
20 207 205
195
165 159
143
10 96 95
53 48 53 70 43
11 31 2 6
0
Easy

Moderate

Difficult

CRF ≥85%

<1 year

1–3 years

3–5 years

5–7 years

≥7 years

White

Asian

Black

Other

Not reported
Matchability Highly Waiting time Recipient ethnicity
sensitized

Fig. 24.3 Effect of UK 2006 National Kidney Allocation scheme. DBD, donors after brain death; CRF, calculated reaction frequency. (Based on NHS Blood
and Transplant data from 2003 versus 2013.)

racial differences in locus allele frequency, HLA subtype ECD, and DCD. Within each category, several principles
matching was shown to be disadvantageous to minori- dictated allocation priority18,24:
ties, limiting their access to deceased donor transplanta-   

tion.19 As a result, in 2003 allocation points based on Candidates are listed for simultaneous kidney and non-
□ 

HLA-B matching were eliminated. Subsequent studies kidney organ transplants.


demonstrated that these policy changes were effective Candidates with zero antigen mismatch with the donor
□ 

in mitigating racial disparities with no adverse effect on kidney (accepting organ procurement organization
graft survival. Unfortunately, these policy changes did [OPO] must “pay back” a kidney to a common national
not eliminate racial disparities in deceased donor trans- pool).
plantation rates.20,21 Candidates are assigned points enabling ranking within
□ 

Other allocation policy changes (circa 2002) have each group:


focused on expanding the deceased donor pool to include Waiting time (begins at time of listing; listing require-
□ 

those kidneys previously deemed not suitable for trans- ment = maintenance dialysis or glomerular filtration
plant—the concept of expanded criteria donor (ECD) kid- rate ≤20 mL/min)
neys and DCD kidneys. ECD was defined as kidneys from Number of antigen mismatches between donor and
□ 

any brain dead donor ≥60 years or aged 50 to 59 years candidate at the DR locus
with at least two of the following: history of hypertension, Calculated panel reactive antibody (cPRA) ≥80%
□ 

serum creatinine >1.5 mg/dL, or cause of death from cere- Adherence to geography: local → regional → national;
□ 

brovascular accident. Receipt of an ECD kidney conferred at each geographic level candidates were rank ordered
a 1.7-fold higher risk for graft failure compared with a according to the point system
standard criteria donor (SCD) kidney; however, studies Pediatric candidates (<18 years) were given priority
□ 

consistently demonstrated ECD kidney transplantation ahead of adult candidates within each geographic level
was associated with a significant survival benefit over for nonzero mismatch kidney offers from donors <35
remaining on dialysis.22,23 These changes resulted in an years.
allocation system that categorized donors into four mutu- Kidneys were distributed to ABO identical candidates
□ 

ally exclusive groups: SCD <35 years, SCD ≥35 years, first, then to ABO-compatible candidates.

  
24 • Kidney Allocation 375

TABLE 24.2 The US Kidney Allocation System Allocation Sequence


Sequence A KDPI ≤20% Sequence B KDPI >20% but <35% Sequence C KDPI ≥35% but ≤85% Sequence Da KDPI >85%
Local CPRA 100 Local CPRA 100 Local CPRA 100 Local CPRA 100
Regional CPRA 100 Regional CPRA 100 Regional CPRA 100 Regional CPRA 100
National CPRA 100 National CPRA 100 National CPRA 100 National CPRA 100
Local CPRA 99 Local CPRA 99 Local CPRA 99 Local CPRA 99
Regional CPRA 99 Regional CPRA 99 Regional CPRA 99 Regional CPRA 99
Local CPRA 98 Local CPRA 98 Local CPRA 98 Local CPRA 98
Zero mismatch (top 20% EPTS) Zero mismatch Zero mismatch Zero mismatch
Prior living donor Prior living donor Prior living donor Local + regional
Local pediatrics Local pediatrics Local National
Local top 20% EPTS Local adults Regional
Zero mismatch (all) Regional pediatrics National
Local (all) Regional adults
Regional pediatrics National pediatrics
Regional (top 20%) National adults
Regional (all)
National pediatrics
National (top 20%)
National (all)

aAll categories in Sequence D are limited to adult candidates.

Despite the numerous allocation policy changes since the weight, ethnicity, history of hypertension, history of diabe-
OPTN’s inception in 1984, variability in access to transplanta- tes, cause of death, serum creatinine, hepatitis C virus, and
tion by candidate blood type, high kidney discard rates, unre- DCD.27 A lower KDPI is associated with better posttransplant
alized graft years, and high retransplant rates persisted. Thus survival.28 Similar to the previous allocation scheme, KDPI is
in December 2014 the new Kidney Allocation System (KAS) now utilized to categorize donor kidneys into four mutually
was implemented with the stated goals to make the most of exclusive sequences: (1) KDPI ≤20% (0.20), (2) KDPI >20%
every donated kidney without diminishing access, promote (0.20) but <35% (0.35), (3) KDPI ≥ 35% (0.35) but <85%
graft survival for those at highest risk for retransplant, mini- (0.85), and (4) KDPI ≥85% (0.85; Table 24.2). Sequence D
mize loss of potential graft function through better matching, (KDPI ≥85%) reflects a similar quality of kidney as the previ-
improve efficiency and utilization by providing better informa- ous ECD definition (higher risk for graft failure), and as such,
tion about kidney offers, provide comprehensive data to guide requires a separate approval and consent for eligibility.
transplant decision making, and reduce differences in access EPTS is used to estimate posttransplant survival of recipi-
for ethnic minorities, pediatric candidates, and sensitized can- ents and to risk-stratify patients into two broad groups (≤20%
didates.25 To accomplish these stated goals designed to bal- vs. 21%–100%) based on four variables: recipient age, diabetic
ance equity and utility, KAS resulted in system-wide changes: status, time on dialysis, and number of prior solid-organ trans-
plants.29 Because candidates with EPTS scores ≤20% have
  

Kidney donors scored by the Kidney Donor Profile Index


□ 
the longest estimated posttransplant survival, they are priori-
(KDPI)
tized to receive kidney offers from donors with KDPI ≤20%. In
Candidates classified by Estimated Post-Transplant Sur-
□ 
general, older age, longer dialysis duration, prior solid-organ
vival (EPTS) score
transplant, and the presence of diabetes are associated with
Waiting time calculation begins at initiation of dialysis
□ 
higher EPTS scores.26 KAS also revised candidate alloca-
(not time of listing) or at time of preemptive listing
tion points to reflect updated goals. Specifically, waiting-time
cPRA priority points allocated using sliding scale
□ 
points now begin at dialysis or listing with glomerular filtra-
Pediatric (<18 years) allocation priority for kidneys with
□ 
tion rate (GFR) ≤20 mL/min and cPRA priority awards addi-
KDPI <0.35
tional points to candidates with highest level of sensitization.
Points for pediatric candidates when offered a zero anti-
□ 
All candidates are ranked by points within their EPTS group.
gen mismatch
Data from the first 18 months since implementation of KAS
Blood type B candidates eligible to accept kidneys from
□ 
were reviewed. Post-KAS there has been a 6.9% increase in sol-
A2 and A2B donors
itary deceased donor kidney transplants observed nationally;
Elimination of kidney “pay back” policy
□ 
however, the geographic distribution of this increased volume
Elimination of kidney variances
□ 
   was not consistent across regions.30,31 In fact, most UNOS
To improve efficiency and organ utilization, KAS uses KDPI regions saw only modest changes in volume with the largest
to risk-stratify donors and EPTS is used to risk-stratify relative increase occurring in region 9 and the largest relative
recipients.26 KDPI replaced SCD and ECD definitions and decrease in region 6 (Fig. 24.4). The proportion of transplants
provides increased precision in determining organ qual- occurring in patients with cPRA 99% to 100% initially rose
ity.27,28 To maximize matching between graft and recipi- from 2.4% of all deceased donor transplants performed each
ent longevity, KAS prioritizes recipients with EPTS scores month to over 17% immediately after KAS implementation.
<20% with donor kidneys with KDPI <20%. By 18 months post-KAS the proportion had decreased and
KDPI is a measure of relative risk derived from the Kid- plateaued to approximately10% of all transplants.31
ney Donor Risk Index (KDRI) on a cumulative percentage In addition, KAS improved patient access to deceased
scale, and is calculated using 10 donor criteria—age, height, donor transplantation for historically disadvantaged
376 Kidney Transplantation: Principles and Practice

1
20 6
7
10 9
8 2
17.518.0
5
18 17.2
11
3
4

16
Percentage of kidney transplants

6
3
14.2
13.3 13.9
14 13.0 Pre KAS (1 year)
12.6 12.9
Post KAS (12 months)
11.3
12 Post KAS (18 months) 10.9
10.8
9.7
10 9.6
9.4
7.8
7.0 6.9 7.6
8 7.4
7.0 6.46.7 6.4 6.7
6.4
6.2
6
4.5
3.9 3.8
4 3.4 3.6 3.8

0
1 2 3 4 5 6 7 8 9 10 11
OPTN region
Fig. 24.4 Geographic distribution of deceased donor kidney transplants in the US pre- and post-KAS implementation.

80
Pre KAS (1 year)
6 months post KAS
1 year post KAS 59.9 59.3
60
54.8 56.6
18 months post KAS
Discard rates (%)

40

20.3 20.7
18.2 20.0
.17.0 17.8 18.5 18.5
20

6.4 7.5 5.8.5.4


2.6
2.0 2.5 2.2
0
0-20 21-34 35-85 86-100 Overall
KDPI (%)
Fig. 24.5 US deceased donor kidney discard rates pre- and post-KAS implementation.

groups. Specifically, in the first 18 months since implemen- Whereas implementation of KAS has resulted in sev-
tation there has been a 16.6% relative increase in trans- eral “big wins,” it is important to note several unin-
plants among African American candidates. Moreover, tended consequences. Specifically, the percentage of
there was a 33% increase in transplantation among candi- kidneys distributed outside the recovering OPO’s donor
dates with ≥5 and <10 years dialysis time and 138% increase service area increased in the first year post-KAS, and as
among candidates with ≥10 years dialysis time. There has such, there was a significant increase in the frequency of
also been a 449% increase in ABO-compatible (e.g., A2 and CIT exceeding 24 hours.30 Rates of delayed graft func-
A2B donor kidneys to B recipients) transplants.30 tion (DGF) also increased in the first 18 months post-
Longevity mismatches have also decreased in the first year KAS from 24.4% to 29.5%.30,31 Despite increases in CIT
post-KAS implementation. The percentage of recipients aged and DGF, graft survival rates in the first 6 months post-
≥65 years receiving KDPI <20% kidneys decreased from 3.2% KAS have remained similar. Finally, although trans-
to 1.1%, and concurrently the number of recipients aged <40 plant volume increased with implementation of KAS
years receiving KDPI <20% kidneys increased by 81.7%.30,31 so too have discard rates, and discard has been most
Not surprisingly, the gap between donor and recipient age pronounced among donor kidneys with KDPI ≥85%
decreased significantly in the first year post-KAS. (Fig. 24.5).30,31
24 • Kidney Allocation 377

Without question the full effect of KAS has yet to be increased use of points systems brings objectivity and gives
appreciated. Longer follow-up will be needed to better reassurance to patients. It also allows the use of computer
assess patient and graft outcomes, transplant volumes, and simulations as new iterations of the allocation algorithm
discard rates, and to ensure that the stated goals of improv- are designed. These simulations have become a valuable
ing access for vulnerable populations persists. tool that allows the designers to see the predicted conse-
quences of minor alterations in the points scoring schemes.
It is quite possible that artificial intelligence will plan
allocation systems in the future. In the meantime we must
Other Variations continue to monitor present schemes and fine tune them
to improve the balance between conflicting principles that
EUROTRANSPLANT govern allocation.

This scheme, one of the longest running, has a number References


of distinctive features. First, it has medical urgency as
1. Gutmann T, Land W. The ethics of organ allocation: the state of
a factor in the allocation score.32 Second, rather than debate. Transplant Rev 1997;11(4):191–207.
being defined by a strict age cutoff, pediatric status (and 2. Oniscu GC, Schalkwijk AAH, Johnson RJ, Brown H, Forsythe JLR.
therefore priority) is granted to any patient demonstrat- Equity of access to renal transplant waiting list and renal transplanta-
ing growth potential on x-ray.32 Third, an acceptable tion in Scotland: cohort study. BMJ 2003;327(7426):1261.
mismatch program was introduced as early as 1996 for 3. Akolekar D, Oniscu GC, Forsythe JL. Variations in the assessment
practice for renal transplantation across the United Kingdom. Trans-
those patients who are highly sensitized.33 Finally, in the plantation 2008;85(3):407–10.
Eurotransplant Senior program, running since 1999 and 4. Meier-Kriesche H-U, Port FK, Ojo AO, et al. Effect of waiting time on
colloquially referred to as the “old for old” scheme, non- renal transplant outcome. Kidney Int 2000;58(3):1311–7.
sensitized patients aged over 65 have priority for donors 5. Johnson RJ, Fuggle SV, O’Neill J, et al. Factors influencing outcome
after deceased heart beating donor kidney transplantation in the
aged over 65 irrespective of HLA matching.34 Overall the United Kingdom: an evidence base for a new national kidney alloca-
Eurotransplant scheme has a good record in achieving tion policy. Transplantation 2010;89(4):379–86.
transplantation for long waiting, highly sensitized, and 6. Morris PJ, Johnson RJ, Fuggle SV, Belger MA, Briggs JD. Analysis of
difficult to match patients. This may be, in part, because factors that affect outcome of primary cadaveric renal transplantation
of the international nature of the organ allocation in the UK. HLA task force of the kidney advisory group of the United
Kingdom Transplant Support Service Authority (UKTSSA). Lancet
organization.35,36 1999;354(9185):1147–52.
7. Gilks WR, Bradley BA, Gore SM, Klouda PT. Substantial ben-
efits of tissue matching in renal transplantation. Transplantation
AUSTRALIA 1987;43(5):669–74.
8. Takemoto S, Terasaki PI, Cecka JM, Cho YW, Gjertson DW. Survival
Australia is notable for strict entry criteria to the waiting list of nationally shared, HLA-matched kidney transplants from cadaveric
with national agreement for patients to have an estimated donors. N Engl J Med 1992;327(12):834–9.
5-year survival of >80% post transplantation. Only well- 9. Rudge C, Johnson RJ, Fuggle SV, Forsythe JL. Renal transplantation in
matched grafts are shared nationally and approximately the United Kingdom for patients from ethnic minorities. Transplanta-
80% are allocated by state protocols, which are encouraged tion 2007;83(9):1169–73.
10. Ross LF, Parker W, Veatch RM, Gentry SE, Thistlethwaite Jr JR. Equal
to weight waiting time heavily to improve access for those opportunity supplemented by fair innings: equity and efficiency in
who have been on the list for a long time.37 allocating deceased donor kidneys. Am J Transplant 2012;12(8):
2115–24.
11. Wu DA, Watson CJ, Bradley JA, et al. Global trends and challenges in
Summary deceased donor kidney allocation. Kidney Int 2017;91(6):1287–99.
12. Fuggle SV, Johnson RJ, Rudge CJ, Forsythe JL. Human leukocyte anti-
gen and the allocation of kidneys from cadaver donors in the United
The uplifting effect of being chosen to receive a kidney, Kingdom. Transplantation 2004;77(4):618–20.
or the polar opposite effect of living on the list for years 13. Fuggle SV, Johnson RJ, Bradley JA, Rudge CJ. Impact of the 1998 UK
with no call, define the need for transparent, objective national allocation scheme for deceased heartbeating donor kidneys.
Transplantation 2010;89(4):372–8.
policies of allocation that are monitored diligently and 14. Johnson RJ, Fuggle SV, Mumford L, et al. A new UK 2006 national
reviewed regularly. The World Health Organization and kidney allocation scheme for deceased heart-beating donor kidneys.
the Declaration of Istanbul have set out principles to be Transplantation 2010;89(4):387–94.
followed.38,39 The latter reminds us that clear programs 15. NHS Blood and Transplant. Kidney Advisory Group. Policy POL186/7.
Kidney transplantation: deceased donor organ allocation Bristol 2017.
with visible accountability are needed, otherwise the Available online at:https://nhsbtdbe.blob.core.windows.net/umbraco-
vacuum is filled by illegal organ trafficking and trans- assets-corp/4368/kidney_allocation_policy.pdf [accessed 30.08.17].
plant tourism. 16. Watson CJ, Johnson RJ, Birch R, Collett D, Bradley JA. A simplified
The strong effect of HLA matching, especially in the earlier donor risk index for predicting outcome after deceased donor kidney
eras when immunosuppression was less effective, means that transplantation. Transplantation 2012;93(3):314–18.
17. Organ Procurement and Transplantation Network Database. Avail-
allocation algorithms have been built around this central able online at: https://optn.transplant.hrsa.gove/data/view-data-
factor. But this has caused inequity, especially where ethnic reports/national-data/# [accessed 26.09.17].
minorities with rare HLA types make up a significant part of 18. Smith JM, Biggins SW, Haselby DG, et al. Kidney, pancreas and liver
the population. This is compounded when those same groups allocation and distribution in the United States. Am J Transplant
2012;12(12):3191–212.
have a predilection for renal disease and a cultural back- 19. Roberts JP, Wolfe RA, Bragg-Gresham JL, et al. Effect of changing
ground that makes deceased organ donation less frequent. the priority for HLA matching on the rates and outcomes of kid-
Different nations balance utility and equity in ways that ney transplantation in minority groups. N Engl J Med 2004;350:
feel comfortable to their medical and social context. The 545–51.
378 Kidney Transplantation: Principles and Practice

20. Ashby VB, Port FK, Wolfe RA, et al. Transplanting kidneys without 31. The new kidney allocation system (KAS): the first 18 months. UNOS
points for HLA-B matching: consequences of the policy change. Am J Transplant Pro website. Available online at: https://www.transplant
Transplant 2011;11:1712–8. pro.org/news/kidney/18-month-analysis-shows-progress-in-kidney-
21. Hall EC, Massie AB, James NT, et al. Effect of eliminating priority allocation-system [accessed 26.09.17].
points for HLA-B matching on racial disparities in kidney transplant 32. Eurotransplant. Eurotransplant Manual Version 5.2. Chapter 4 Kidney
rates. Am J Kidney Dis 2011;58:813–6. (ETKAS and ESP) Leiden 2016. Available online at: https://www.eur
22. Ojo AO, Hanson JA, Meier-Kriesche H, et al. Survival in recipients of otransplant.org/cms/mediaobject.php?file=H4+ETKAS+November+
marginal cadaveric donor kidneys compared with other recipients and 11%2C+20161.pdf [accessed 30.08.17].
wait-listed transplant candidates. J Am Soc Nephrol 2001;12:589–97. 33. Claas FH, Rahmel A, Doxiadis II . Enhanced kidney allocation to highly
23. Stratta RJ, Rohr MS, Sundberg AK, et al. Increased kidney trans- sensitized patients by the acceptable mismatch program. Transplanta-
plantation utilizing expanded criteria deceased donors with results tion 2009;88(4):447–52.
comparable to standard criteria donor transplant. Ann Surg 34. Frei U, Noeldeke J, Machold-Fabrizii V, et al. Prospective age-match-
2004;239:688–97. ing in elderly kidney transplant recipients—a 5-year analysis of the
24. Organ Procurement and Transplantation Network website. Avail- Eurotransplant Senior Program. Am J Transplant 2008;8(1):50–7.
able online at: https://optn.transplant.hrsa.gov/governance/policies/ 35. Persijn GG, Smits JM, Smith M, Frei U. Five-year experience with the
[accessed 2018]. new eurotransplant kidney allocation system, 1996 to 2001. Trans-
25. Kidney Allocation System (KAS) Clarifications and Clean Up. Avail- plant Proc 2002;34(8):3072–4.
able online at:https://optn.transplant.hrsa.gov/media/1239/04_kas 36. Persijn GG, Smits JM, Frei U. Three-year experience with the new
-clarifications.pdf [accessed 26.09.17]. eurotransplant kidney allocation system. Nephrol Dial Transplant
26. Israni AK, Salkowski N, Gustafson S, et al. New national allocation 2001;16(Suppl 6):144–6.
policy for deceased donor kidneys in the United States and possible 37. The Transplantation Society of Australia and New Zealand. Clinical
effect on patient outcomes. J Am Soc Nephrol 2014;25(8):1842–8. Guidelines for Organ Transplantation from Deceased Donors. Chapter 5
27. Guide to calculating and interpreting KDPI. Available online at:https:// Kidney; 2017. Available online at: https://www.tsanz.com.au/organal
optn.transplant.hrsa.gov/media/1512/guide_to_caculating_interpr locationguidelines/documents/ClinicalGuidelinesV1.1May2017.pdf
eting_kdpi.pdf [accessed 26.09.17]. [accessed 30.08.17].
28. Stewart D, Samana C, Cherikh W, et al. Has displaying KDPI in Donor- 38. Steering Committee of the Istanbul Summit. Organ trafficking and
Net® led to a spike in discard rates for lower quality kidneys? Am J transplant tourism and commercialism: the declaration of Istanbul.
Transplant 2013;13:(S5). Lancet 2008;372(9632):5–6.
29. A guide to calculating and interpreting the Estimated Post-Transplant 39. World Health Organization guiding principles on human cell, tissue and
Survival (EPTS) score used in the kidney allocation system (KAS). organ transplantation. 2010. Available online at:http://www.who.
Available online at:https://optn.transplant.hrsa.gov/media/1511/ int/transplantation/Guiding_PrinciplesTransplantation_WHA63.22
guide_to_calculating_interpreting_epts.pdf [accessed 26.09.17]. en.pdf?ua=1.
30. Stewart DE, Kucheryavaya AY, Klassen DK, Turgeon NA, Formica RN,
Aeder MI. Changes in deceased donor kidney transplantation one year
after KAS implementation. Am J Transplant 2016;16(6):1834–47.
25 Pathology of Kidney
Transplantation
ALTON B. FARRIS III, LYNN D. CORNELL and ROBERT B. COLVIN

CHAPTER OUTLINE Renal Allograft Biopsy Protocol Biopsies


Optimal Tissue Acute Tubular Necrosis
Microscopy Calcineurin Inhibitor Nephrotoxicity
Classification of Pathologic Diagnoses in the Acute Calcineurin Inhibitor Toxicity
Renal Allograft Toxic Tubulopathy
Donor Kidney Biopsy Acute Arteriolar Toxicity and Thrombotic
Hyperacute Rejection Microangiopathy
Acute Renal Allograft Rejection Differential Diagnosis
Acute T-Cell-Mediated Rejection Chronic Calcineurin Inhibitor Toxicity
Tubulointerstitial Rejection (Type I) CNI-Arteriolopathy
Endarteritis (Type II Rejection) Glomerular Lesions
Glomerular Lesions Tubules and Interstitium
Atypical Rejection Syndromes Differential Diagnosis
Differential Diagnosis Target of Rapamycin Inhibitor Toxicity
Acute Antibody-Mediated Rejection Drug-Induced Acute Tubulointerstitial
Diagnostic Criteria Nephritis
Pathologic Features Infections
C4d Interpretation Polyomavirus Tubulointerstitial Nephritis
C4d Negative Antibody-Mediated Adenovirus
Rejection Acute Pyelonephritis
Differential Diagnosis Major Renal Vascular Disease
Accommodation De Novo Glomerular Disease
Complement Inhibition Membranous Glomerulonephritis
Classification Systems ANTI-GBM Nephritis
Late Graft Diseases De Novo Podocytopathy in Congenital
Chronic Antibody-Mediated-Rejection Nephrosis
Transplant Glomerulopathy Focal Segmental Glomerulosclerosis
Peritubular Capillary and Tubulointerstitial Recurrent Renal Disease
Lesions Posttransplant Malignancy and
Transplant Arteriopathy Posttransplant Lymphoproliferative
Chronic T-Cell-Mediated Rejection Disease
Other Specific Diagnoses Future Directions in Biopsy Assessment
Interstitial Fibrosis and Tubular Atrophy

ABBREVIATIONS AMR antibody-mediated rejection EM electron microscopy


ATN acute tubular necrosis FSGS focal segmental glomerulosclerosis
CAN chronic allograft nephropathy GBM glomerular basement membrane
CMV cytomegalovirus HLA human leukocyte antigen
CNI calcineurin inhibitor IFTA interstitial fibrosis and tubular atrophy
CNIT calcineurin inhibitor toxicity MGN membranous glomerulonephritis
DGF delayed graft function MPGN membranoproliferative glomerulone-
DSA donor-specific antibody phritis

379
380 Kidney Transplantation: Principles and Practice

PAS periodic acid-Schiff TCMR T cell-mediated rejection


PTC peritubular capillary TG transplant glomerulopathy
PTLD posttransplant lymphoproliferative disease TBM tubular basement membrane
PTN polyomavirus tubulointerstitial nephritis TMA thrombotic microangiopathy

Renal Allograft Biopsy with paraffin sections.13 Rapid (2-hour) formalin/paraffin


processing is used at Massachusetts General Hospital for
Renal biopsy remains the “gold standard” for the diagno- urgent and weekend biopsies.
sis of episodes of graft dysfunction that occur commonly
in patients after transplantation. Studies have indicated
MICROSCOPY
that the results of a renal allograft biopsy change the clini-
cal diagnosis in 30% to 42% and therapy in 38% to 83% The biopsy is examined for glomerular, tubular, vascu-
of patients, even after the first year.1–4 Most important, lar, and interstitial pathology including: (1) transplant
unnecessary immunosuppression was avoided in 19% of glomerulitis, glomerulopathy, and de novo or recurrent
patients.3 The biopsy is also a gold mine of information on glomerulonephritis; (2) tubular injury, isometric vacuol-
pathogenetic mechanisms, a generator of hypotheses that ization, tubulitis, atrophy, or intranuclear viral inclusions;
can be tested in experimental animal studies and in clini- (3) endarteritis, fibrinoid necrosis, thrombi, myocyte necro-
cal trials. Finally, the biopsy serves, in turn, to validate the sis, nodular medial hyalinosis, or chronic allograft arteri-
hypothesis tested in such trials. Renal biopsy interpretation opathy; (4) interstitial infiltrates of activated mononuclear
currently relies primarily on histopathology complemented cells, edema, or neutrophils, fibrosis, and scarring. Arter-
by immunologic molecular probes. Quantitative gene ies and arterioles are particularly scrutinized, because the
expression analysis methods may be implemented more diagnostic lesion often lies there.
in the future as those techniques are further validated and A typical immunofluorescence panel (used at Massachu-
approved for clinical use.5–8 setts General Hospital) detects IgG, IgA, IgM, C3, kappa and
This chapter describes the relevant light, immunofluores- lambda light chains, C4d, albumin, and fibrin in cryostat
cence, and electron microscopy (EM) findings of the most com- sections. C4d, a complement fragment, is used to identify
mon lesions affecting the renal allograft and their differential AMR; the other stains are primarily for recurrent or de
diagnosis, citing references largely limited to human patho- novo glomerulonephritis.14 Immunohistochemistry (IHC)
logic studies after 1990. The discussion is broadly divided in paraffin sections is indicated in the differential diagnosis
into allograft rejection and nonrejection pathology, with of lymphoproliferative or viral diseases and may be used for
an emphasis on differential diagnosis of acute and chronic C4d. EM is valuable when de novo or recurrent glomerular
allograft dysfunction. Grading systems of acute and chronic disease is suspected and to evaluate peritubular capillary
rejection are discussed further in those sections. Additional ref- (PTC) basement membranes.15
erences and details are available in a comprehensive review.9
CLASSIFICATION OF PATHOLOGIC DIAGNOSES IN
OPTIMAL TISSUE THE RENAL ALLOGRAFT
At least seven nonsclerotic glomeruli and two arteries (big- The ideal diagnostic classification of renal allograft pathology
ger than arterioles) must be present in a renal allograft should be based on pathogenesis, have therapeutic relevance,
biopsy for adequate evaluation.10,11 Using these criteria, and be reproducible. The current classification based on Banff
the sensitivity of a single core is approximately 90%, and and other systems (Table 25.1), meets these criteria.16,17
the predicted sensitivity of two cores is about 99%.11 How-
ever, adequacy depends entirely on the lesions seen in the
biopsy: one artery with endarteritis is sufficient for the diag- Donor Kidney Biopsy
nosis of acute cellular rejection (TCMR), even if no glomeru-
lus is present; similarly, immunofluorescence or EM of one Biopsy of the cadaveric donor kidney is sometimes used to
glomerulus is adequate to diagnose membranous glomer- determine the suitability of the kidney for transplantation.
ulonephritis (MGN). In contrast, a large portion of cortex Objective pathologic criteria based on outcome that could
with a minimal infiltrate does not exclude rejection. Sub- be applied to the renal biopsy as a screening test have not
capsular cortex often shows inflammation and fibrosis and been fully established, as donor biopsies are not always
is not representative. Diagnosis of certain diseases is even performed and controlled trials have not been done. One
possible with only medulla (acute humoral rejection [acute of the major problems in assessing the donor kidney is that
AMR], polyomavirus tubulointerstitial nephritis [PTN]). this is usually done with cryostat sections, often by local
However, a normal medulla does not rule out rejection.12 pathologists in the middle of the night. Using arbitrary
Frozen sections for light microscopy are of limited value, criteria risks that kidneys will be discarded needlessly.
because frozen artifacts preclude accurate evaluation. The In several large studies, the outcome at 1 to 5 years has
diagnostic accuracy of frozen sections was 89% compared not measurably correlated with pathologic lesions.18–20
25 • Pathology of Kidney Transplantation 381

TABLE 25.1 Pathologic Classification of Renal Allograft strikingly diminished in recipients of grafts with >20% glo-
Disease16 merulosclerosis compared with those 0% sclerosis (35% vs.
80%).24 However, other large studies have failed to detect a
I. Immunologic rejection major effect of glomerulosclerosis >20%, if adjusted for the
A. Hyperacute rejection
B. Acute rejection
age of the donor25 or renal function.26 At least 25 glomeruli
a. Acute T-cell-mediated rejection (acute cellular rejection, C4d−) are needed to correlate with outcome.27 A wedge biopsy
i. Tubulointerstitial (Banff type I) may not be representative, because it includes mostly outer
ii. Endarteritis (Banff type II) cortex, the zone where glomerulosclerosis and fibrosis due
iii. Arterial fibrinoid necrosis/transmural inflammation (Banff to vascular disease is most severe, therefore a needle biopsy
type III)
iv.  Glomerular (transplant glomerulitis; no Banff type) is recommended. Even though many other studies try to
b. Acute AMR (acute humoral rejection, C4d+) correlate fibrosis or vascular disease, reproducibility of
i. Tubular injury scoring these lesions, even on permanent sections in broad
ii. Capillaritis/thrombotic microangiopathy daylight, is notoriously poor.28 At this time histologic evalu-
iii. Arterial fibrinoid necrosis
C. Chronic rejection
ation is recommended in donors with any evidence of renal
a. Chronic T-cell-mediated rejection (with T cell activity) dysfunction, a family history of renal disease, or whose age
b. Chronic antibody-mediated rejection (with antibody is >60 years. Histologic selection of optimal kidneys from
activity, C4d+) donors over age 60 years can result in a graft survival rate
II. Allo-/autoantibody-mediated diseases of allografts similar to that of grafts from younger patients.29
A. Anti-GBM disease in Alport’s syndrome
B. Nephrotic syndrome in nephrin-deficient recipients Other lesions may cause the transplant surgeon or pathol-
C. Anti-TBM disease in TBM antigen-deficient recipients ogist to argue against use of the graft. Arterial intimal fibro-
D. De novo membranous glomerulonephritis sis increases the risk of delayed graft function (DGF)30 and
E. Antiangiotensin II receptor autoantibody syndrome has a slight effect on 2-year graft survival (6% decrease).31
III. Nonrejection injury
A. Acute ischemic injury or other causes of acute tubular injury/
Thrombotic microangiopathy (TMA) with widespread but
necrosis (ATN) less than 50% glomerular thrombi increases the likelihood
B. Drug toxicity of DGF and primary nonfunction,25 but unaltered 2-year
□ Calcineurin inhibitor (cyclosporine, tacrolimus) graft survival can still be observed.32 Likewise, deceased
□ mTOR inhibitors (sirolimus, everolimus, rapamycin)
donor kidneys with fibrin thrombi in up to essentially 100%
□ Drug-induced interstitial nephritis

C. Acute tubulointerstitial nephritis (drug allergy) of glomeruli due to presumed disseminated intravascular
D. Infection (viral, bacterial, fungal) (e.g., BK virus nephropathy, coagulation have been transplanted successfully with ini-
pyelonephritis) tial DGF but eventual stable allograft function.33 Despite
E. Major artery/vein thrombosis initial DGF, It has been shown that donor-derived glomeru-
F. Mechanical
□ Obstruction
lar fibrin thrombi can resolve after donor kidney transplan-
□ Urine leak
tation,34 sometimes quite rapidly.35
G. Renal artery stenosis Reversal of diabetic glomerulosclerosis36 and IgA nephrop-
H. Arteriosclerosis athy have been reported,37 as well as membranous glomer-
I. De novo glomerular disease/glomerulopathy (other than trans- ulonephritis,38 lupus nephritis,39 membranoproliferative
plant glomerulopathy)
J. Posttransplant lymphoproliferative disease (PTLD) glomerulonephritis (MPGN),40 and endotheliosis due to pre-
K. Chronic allograft nephropathy, not otherwise classified (intersti- eclampsia (personal observation).
tial fibrosis and tubular atrophy)
IV. Recurrent primary disease
A. Immunologic (e.g., IgA nephropathy, lupus nephritis, anti-GBM
disease)
Hyperacute Rejection
B. Metabolic (e.g., amyloidosis, diabetes, oxalosis)
C. Unknown (e.g., dense deposit disease, focal segmental glo- Hyperacute rejection refers to immediate rejection (typi-
merulosclerosis) cally within minutes to hours) of the kidney upon perfusion
with recipient blood, where the recipient is presensitized to
alloantigens on the surface of the graft endothelium. Dur-
ing surgery, the graft kidney becomes soft and flabby; and
As rejection and patient death from complications dimin- livid, mottled, purple, or cyanotic in color; and urine output
ish, the influence of the quality of the graft is likely to ceases. The kidney subsequently swells, and widespread
increase. Both donor biopsies and reperfusion biopsies can hemorrhagic cortical necrosis and medullary congestion
be quite helpful in assessing the baseline status of the graft, appears. The large vessels are sometimes thrombosed.
although reperfusion biopsies do not provide aid in donor Early lesions show marked accumulation of platelets in
selection.21 glomerular capillaries’ lumina that appear as amorphous,
Glomerulosclerosis is one feature that is readily assessed pale pink, finely granular masses in hematoxylin and eosin
in frozen section, by the most casual observation. Glomeru- (H&E) stained slides (negative on periodic acid Schiff [PAS]
losclerosis >20% correlates with poor graft outcome In sev- stains). Neutrophil and platelet margination then occur over
eral studies, donor serum creatinine did not distinguish the the next hour or so along damaged endothelium of small
different degrees of glomerulosclerosis found on biopsy,22–24 arteries, arterioles, glomeruli, and PTCs, and the capillar-
although that has been demonstrated by other studies.20 In ies fill with a sludge of compacted red cells and fibrin.41 The
one study, the odds ratio for poor outcome remained sig- larger arteries are usually spared. The neutrophils do not
nificant after adjustment for donor age, rejection episodes, infiltrate initially but form “chain-like” figures in the PTCs
or panel reactive antibody.23 Five-year graft survival was without obvious thrombi.41 The endothelium is stripped off
382 Kidney Transplantation: Principles and Practice

the underlying basal lamina, and the interstitium becomes TABLE 25.2 Summary of Banff/CCTT Types of Acute
edematous and hemorrhagic. Intravascular coagulation T-Cell-Mediated Rejection1,16
occurs and cortical necrosis ensues over 12 to 24 hours.
The medulla is relatively spared, but is ultimately affected Suspicious/ Tubulitis + infiltrate
borderline2 Tubulitis (t1, t2, or t3) with minor interstitial infiltrate
as the whole kidney becomes necrotic.42 Widespread micro- (i0 or i1) or infiltrate (i2, i3) with mild tubulitis (t1)
thrombi are usually found in the arterioles and glomeruli Type I3 Tubulitis >4 cells/tubule + infiltrate >25%
and can be detected even in totally necrotic samples. The A: with 5–10 cells/tubule (t2), or
small arteries may show fibrinoid necrosis. Mononuclear B: with >10 cells/tubule (t3)
Type II Mononuclear cells under arterial endothelium
infiltrates are typically sparse. One case showed CD3+ cells A: <25% luminal area, or
in the adventitia of small arteries and in the surrounding B: ≥25% luminal area
interstitium.43 By EM, neutrophils attach to injured glo- Type III Transmural arterial inflammation, or fibrinoid
merular endothelial cells.41 The endothelium is swollen— ­arterial necrosis with accompanying lymphocytic
separated from the glomerular basement membrane (GBM) inflammation4
by a lucent space. Capillary loops and PTCs are often bare of
endothelium. Platelet, fibrin thrombi, and trapped erythro-
cytes occlude capillaries.16 histocompatibility antigens in the kidney and is much
The site of antibody and complement deposition is deter- more common than acute humoral rejection, due to donor-
mined by the site of the target endothelial alloantigens. specific antibodies (i.e., acute antibody-mediated rejec-
Hyperacute rejection due to preexisting anti-HLA class I tion), although the latter is now recognized with greater
antibodies may show C3, C4d, and fibrin throughout the frequency and has a worse prognosis. Only since 1999 has
microvasculature.44 ABO antibodies (primarily IgM) also the distinction between the two been clearly made in the
deposit in all vascular endothelium. Cases with anticlass literature.
II antibodies may have IgG/IgM primarily in glomerular
capillaries and peritubular capillaries, where class II is nor-
ACUTE T-CELL-MEDIATED REJECTION
mally conspicuous.45 In antiendothelial-monocyte antigen
cases, IgG is primarily in PTCs, rather than glomeruli or T cells react to donor histocompatibility antigens expressed
arteries.46 Often antibodies cannot be detected in the ves- in the tubules, interstitium, vessels, and glomeruli, sepa-
sels,47 even though they can be eluted from the kidney.48,49 rately or in combination (Table 25.2). The donor ureter is
In these cases C4d should be positive in PTCs14 and more also affected but rarely sampled.52
useful than immunoglobulin stains. Occasional cases, par-
ticularly intraoperative biopsies, may be negative for C4d Tubulointerstitial Rejection (Type I)
(A. H. Cohen, Cedar Sinai Hospital, Los Angeles, personal The prominent microscopic feature of TCMR is a pleomor-
communication), perhaps related to focally decreased per- phic interstitial infiltrate of mononuclear cells, accompa-
fusion or insufficient time to generate substantial C4d nied by interstitial edema and sometimes hemorrhage (Fig.
amounts.16 25.1). The infiltrate is typically patchy, both in the cortex
The differential diagnosis of hyperacute rejection includes and medulla. The infiltrating cells are primarily T cells
ischemia and major vascular thrombosis.16 The major diag- and macrophages. Activated T cells (lymphoblasts) with
nostic feature of hyperacute rejection is C4d deposition in increased basophilic cytoplasm, nucleoli, and occasional
PTCs and the prominence of neutrophils in capillaries. mitotic figures indicate increased synthetic and proliferative
Although the finding of antibody and C4d deposition in activity.53 Granulocytes are not uncommonly present but
PTCs is diagnostic when present, negative immunofluo- rarely prominent. When neutrophils are conspicuous, the
rescence stains do not exclude hyperacute rejection. Exog- possibility of AMR or pyelonephritis should be considered.
enous antibody (rabbit or horse antilymphocyte serum) Eosinophils are present in about 30% of biopsies with rejec-
can cause severe endothelial injury sometimes with C4d tion and can be abundant, but are rarely more than 2% to
deposition mimicking hyperacute rejection.50 Hyperacute 3% of the infiltrate.54,55 Abundant eosinophils (10% of infil-
rejection typically has more hemorrhage, necrosis, and trate) are associated with endarteritis (Banff type II).56 Mast
neutrophil accumulation in glomeruli and PTCs than acute cells increase, as judged by tryptase content, and correlate
tubular necrosis (ATN), although glomerular neutrophils with edema.57 Acute rejection with abundant plasma cells
alone are associated with ischemia.51 Major arterial throm- has been described as early as the first month associated
bosis has predominant necrosis with little hemorrhage or with poor graft survival.58–60 Infiltrating T cells express
microthrombi and PTC neutrophils are not prominent. cytotoxic molecules, namely perforin,61,62 FasL,62,63 gran-
Renal vein thrombosis shows marked congestion and rela- zyme A and B,62,64–66 and TIA-1/GMP-17,66,67 and tumor
tively little neutrophil response. necrosis factor-β (lymphotoxin).68 Apoptosis of the infiltrat-
ing T cells can be demonstrated with the TdT-uridine-nick
end label (TUNEL) technique, probably as a result of acti-
Acute Renal Allograft Rejection vation-induced cell death, and would thereby serve to limit
the immune reaction.67
Acute rejection typically develops in the first 2 to 6 weeks Mononuclear cells invade tubules and insinuate between
after transplantation, but can arise in a normally func- tubular epithelial cells, a process termed “tubulitis” (see Fig.
tioning kidney from 3 days to 10 years or more, or in a 25.1 inset), which is best appreciated in sections stained
graft affected by other conditions, such as ATN, calcineu- with PAS or a silver stain to delineate the tubular basement
rin inhibitor toxicity, or chronic rejection. Acute rejection membrane (TBM). All cortical tubules (proximal and distal)
may be cell mediated, humoral, or both (see Table 25.1). as well as the medullary tubules and the collecting ducts
TCMR is mediated primarily by T cells reacting to donor may be affected. Tubular cell apoptosis occurs,67,69–71 which
25 • Pathology of Kidney Transplantation 383

A B
Fig. 25.1 Acute cellular rejection type I. (A) Mononuclear cells, composed of activated lymphocytes and macrophages, infiltrate the edematous
interstitium and invade tubules. Tubulitis affects proximal and other tubules, where mononuclear cells are interposed between the tubular epithelial
cells (inset). The invading mononuclear cells appear dark with scant cytoplasm, which distinguishes them from tubular epithelial cells. The tubular base-
ment membranes are stained red by the periodic acid-Schiff stain, which is useful to delineate the boundary between the tubule and the interstitium.

correlates with the number of cytotoxic cells and macro- comparable to that in the normal thymus (1.8% of cells).67
phages in the infiltrate.67,70 Tubular epithelial cells express Others have described occasional TUNEL+ lymphocytes.69
human leukocyte antigen–DR (HLA-DR), intercellular Apoptosis probably occurs in infiltrating T cells as a result
adhesion molecule 1 (ICAM 1), and vascular cell adhesion of activation-induced cell death and would thereby serve to
molecule 1 (VCAM-1) in increased amounts in TCMR72–82 limit the immune reaction.67 Little, if any, immunoglobu-
and express the costimulatory molecules CD80 and CD86.83 lin deposition is found by immunofluorescence in TCMR,
Tubules also synthesize tumor necrosis factor-α,84 transform- which is characterized primarily by extravascular fibrin
ing growth factor-β1, IL-15, osteopontin, and vascular endo- accumulation in the interstitium and not uncommonly
thelial growth factor (VEGF).85–87 Increased expression of increased C3 along the TBM. The C3 is largely derived from
S100A4 may signal the process of epithelial to mesenchymal tubular cells.95 C3 may have a role in the pathogenesis of
transition (EMT),88 which may actually consist of an in situ acute rejection, because C3-deficient mouse kidneys have
epithelial response rather than a true emigration of tubular prolonged survival.96 C4d deposition in PTCs indicates an
epithelial cells into the interstitium.89 Thus, as suggested by antibody-mediated component.
proceedings at a Banff conference on allograft pathology,17 Gene expression studies of graft tissue have revealed that
EMT may be better thought of as an epithelial to mesenchy- transcripts for proteins of cytotoxic T lymphocytes (CTLs),
mal phenotype (EMP).90 Some tubular cell-derived molecules such as granzyme B, perforin, and Fas ligand97–103 and the
have the potential to inhibit acute rejection, such as protease master transcription factor for CTLs, T-bet, are characteristic
inhibitor-9 (PI-9), the only known inhibitor of granzyme B65 of TCMR.101 Graft CTL-associated transcripts (CATs) precede
and IL-15, which inhibits expression of perforin.85 tubulitis in mouse kidney grafts.104 Treatment of rejection
CD8+ and CD4+ cells invade tubules.91 Intratubular is followed by a measurable decrease of CATs.100 However,
T cells with cytotoxic granules,67 and CD4+FOXP3+ cells92 knockout of either granzyme or perforin does not prevent
accumulate selectively in the tubules, compared with the acute rejection, suggesting they are not essential.105 IFNγ
interstitial infiltrate. T cells proliferate once inside the tubule, mRNA is detectable in fine needle aspirates 1 week before
as judged by the marker Ki67 (MIB-1), which contributes to the clinical onset of rejection.106 Other genes associated with
their concentration within tubules, in addition to selective acute rejection are IFNγ, TNFβ, TNFα, RANTES (regulated
invasion.67,93 Increased tubular HLA-DR,72,73 tumor necro- on activation, normal T cell expressed and secreted, also
sis factor (TNF)α,84 IFNγ receptor,68 IL-2 receptor,94 and known as also Chemokine (C-C motif) ligand 5 [CCL5]), and
IL-8 are detectable by immunoperoxidase study in TCMR. macrophage inflammatory protein 1-alpha (MIP-1-alpha,
Several adhesion molecules are increased on tubular cells also known as Chemokine [C-C motif] ligand 3 [CCL3]); no
during rejection, including ICAM-1 (CD54) and VCAM-1, elevation of TGFβ or IL-10 is detected.101
and correlate with the degree of T cell infiltration.82
Signs of tubular cell injury can be detected by TUNEL Endarteritis (Type II Rejection)
apoptosis assay. Increased numbers of TUNEL+ tubular Infiltration of mononuclear cells under arterial and arte-
cells are present in acute rejection, compared with normal riolar endothelium is the pathognomic lesion of TCMR
kidneys.67,69 The frequency was significantly lower in cyclo- (Fig. 25.2). Many terms have been used for this process,
sporin A (CsA) toxicity or ATN.67 The degree of apoptosis including “endothelialitis,” “endothelitis,” “endovasculi-
correlates with the cytotoxic cells in the infiltrate, consistent tis,” “intimal arteritis,” or “endarteritis.” We prefer the last
with a pathogenetic relationship.67 Prominent apoptosis of term, which emphasizes the type of vessel (artery vs. vein)
the infiltrating T cells has also been detected at a frequency involved and the site of inflammation. Mononuclear cells
384 Kidney Transplantation: Principles and Practice

A B
Fig. 25.2 Acute cellular rejection type II. (A) Endarteritis in a medium sized artery. The endothelium is lifted by undermining mononuclear cells, with-
out involvement of the media. (B) Subendothelial infiltration in a small artery with underlying arteriosclerosis (donor disease). This acute process should
be distinguished from chronic transplant arteriopathy.

that are sometimes attached to the endothelial surface are little change. In severe cases a transmural mononuclear infil-
insufficient for the diagnosis of endarteritis; however, they trate may be seen (termed “type III rejection”). The cells infil-
probably represent the early phase of this lesion. Endarteri- trating the endothelium and intima are T cells and monocytes,
tis in TCMR must not be confused with fibrinoid necrosis of but not B cells.112 Both CD8+ and CD4+ cells invade the intima
arteries. The latter is characteristic of acute AMR and can in early grafts, but later CD8+ cells predominate,91 suggesting
also be seen in thrombotic vasculopathy. Regrettably, some that class I antigens are the primary target.67 Vascular endo-
still do not separate these lesions, regarding all “vascular thelial cell apoptosis can be detected in sites of endarteritis.67,69
rejection” as predominately humoral. Normal arterial endothelial cells express class I antigens,
Endarteritis has been reported in 35% to 56% of renal weak ICAM-1, and little or no class II antigens, or VCAM-1.
biopsies with TCMR.11,55,107–109 Many do not find the lesion During acute rejection the endothelium of arteries expresses
as often, which may possibly be ascribed to inadequate sam- increased HLA-DR74,91 and ICAM-1 and VCAM-1.79,81
pling, overdiagnosis of rejection (increasing the denomina- This adhesion molecule upregulation occurs in association
tor), patient population with respect to medication adherence with CD3+82 and CD25+80 infiltrating mononuclear cells.
(severity of rejection), or the timing of the biopsy with respect Endothelial cells also have decreased endothelin expression
to antirejection therapy. Endarteritis lesions affect arteries of in rejection with endarteritis, but not in tubulointerstitial
all sizes including the arteriole, although the lesions affect rejection.114
larger vessels preferentially. For example, in a detailed analy-
sis, 27% of the artery cross sections were affected, vs. 13% Glomerular Lesions
of the arterioles.55 A sample of four arteries would have an In most TCMR cases, glomeruli are spared or show minor
estimated sensitivity of about 75% in the detection of type II changes, typically a few scattered mononuclear cells
rejection.55 Thus a sample may not be considered adequate (T cells and monocytes) and occasionally segmental endo-
to rule out endarteritis unless several arteries are included. thelial damage (Fig. 25.3).115 A severe form of this glo-
“Arteriolitis” has the same significance as endarteritis.110 merular injury, termed “transplant glomerulitis” or “acute
Endarteritis can occur in cases with little or no interstitial allograft glomerulopathy,” develops in a minority of cases
infiltrate or tubulitis, arguing that it has a distinct patho- (<5%), manifested by hypercellularity, injury, and enlarge-
genetic mechanism,16,111 and even in cases with “isolated ment of endothelial cells; infiltration of glomeruli by mono-
endarteritis,” that finding is an independent risk factor for nuclear cells; and by webs of PAS-positive material.116
kidney transplant failure.111 In severe cases, a transmural Crescents and thrombi are rare. Endarteritis often accompa-
mononuclear infiltrate affects the media, with focal necro- nies the transplant glomerulitis.117 The glomeruli contain
sis of the myocytes, features that constitute type III rejection numerous CD3+ and CD8+ T cells and monocytes.91,118
(transmural inflammation or fibrinoid necrosis). Although Fibrin and scant immunoglobulin and complement depos-
this occasionally occurs in the absence of demonstrable anti- its are found in glomeruli. This variant of cellular rejection
bodies, it is more typical of AMR. has been associated with certain viral infections, such as
Endothelial cells are typically reactive with increased cyto- cytomegalovirus (CMV) infection and hepatitis C virus,119
plasmic volume and basophilia. The endothelium shows although viral antigens are not in the glomerular lesions.
disruption and lifting from supporting stroma by infiltrat-
ing inflammatory cells.112 Occasionally endothelial cells are Atypical Rejection Syndromes
necrotic or absent, however, thrombosis is rare. Endothelial Unique patterns of rejection have been observed under
apoptosis occurs67,69 and increased numbers of endothelial novel immunosuppression regimens. For example, follow-
cells appear in the circulation.113 The media usually shows ing pronounced lymphocyte depletion from alemtuzumab
25 • Pathology of Kidney Transplantation 385

A B

C D
Fig. 25.3 Acute humoral rejection. (A) Low power shows mild interstitial inflammation, focal hemorrhage, neutrophils and thrombi in glomerular
capillaries, and dilated peritubular capillaries with leukocytes. (B) At high power neutrophils can be seen in the peritubular capillaries with little tubulitis.
PAS stain. (C) Acute transplant glomerulitis is prominent in this case of acute humoral rejection. Glomerular endothelial cells are swollen and the capil-
laries are filled with mononuclear cells, probably mostly macrophages. PAS stain. (D) C4d stain of a case of acute humoral rejection, shows prominent,
diffuse staining of dilated peritubular capillaries, sometimes containing inflammatory cells, and linear staining along the glomerular basement mem-
brane. Immunohistochemistry with a polyclonal anti-C4d rabbit antibody.

(CAMPATH-1H),53,120,121 TCMR with a prominent mono- modifications in the combined kidney and bone marrow
cyte population (i.e., an acute monocytic rejection) has transplantation protocol, it is possible that the “engraft-
been described. In these cases, much of the interstitial rejec- ment syndrome” can be eliminated or at least attenuated;
tion infiltrate stains for CD68, correlating with renal dys- this suggests that “engraftment syndrome” may not be an
function and tubular stress, shown by HLA-DR staining of accurate term for what may actually just be a form of tran-
the tubules. Under these conditions, T cells did not correlate sient acute kidney injury.125
with renal dysfunction or HLA-DR staining.120
Studies have included simultaneous bone marrow and Differential Diagnosis
kidney transplantation protocols in attempt to induce toler- TCMR typically has a diffuse, interstitial mononuclear cell
ance to the transplanted organ. In these studies, human leu- infiltrate, whereas patients with CNI toxicity (CNIT) and
kocyte antigen (HLA)-mismatched renal transplants have those with stable function have only focal mononuclear
been performed; withdrawal of maintenance immunosup- cell infiltrates (Table 25.3). Endarteritis or C4d+ is found
pression has been accomplished in some of the patients with extremely rarely, if ever, in CNIT and if either is present, is
relatively preserved renal function.122 In several of these the most discriminating feature for acute rejection.126–128
patients, a capillary leak or engraftment syndrome has been Prominent tubulitis favors acute rejection, because it is less
observed around 10 days after a simultaneous kidney/bone prominent in acute tubular necrosis, particularly in the
marrow transplant preceded by a nonmyeloablative con- proximal tubules.129 However, tubulitis has been docu-
ditioning regimen. In this “engraftment syndrome,” acute mented in renal transplants with dysfunction due to lym-
tubular injury is accompanied by congested PTCs contain- phoceles (obstruction) and in urine leaks, possibilities that
ing mononuclear cells and red blood cells. IHC shows that need to be considered and excluded by other techniques.130
the cells are primarily CD68+MPO+ mononuclear cells Acute obstruction typically has some dilation of the collect-
and CD3+CD8+ T cells, the latter with a high proliferation ing tubules, especially in the outer cortex. Edema and a mild
index (Ki67+). XY chromosome fluorescence in situ hybrid- mononuclear infiltrate are also common.11,129,130
ization (FISH) has been used to demonstrate that the PTC Interstitial mononuclear inflammation and tubulitis
cells are recipient derived, correlating with chimerism stud- occur in a variety of diseases other than acute rejection,
ies showing a simultaneous decline in circulating donor such as drug-induced (allergic) or infectious tubuloint-
cells and recovery of recipient circulating cells. PTC endo- erstitial nephritis. When eosinophils are more abundant
thelial injury can also be seen on EM in these cases.122,123 than usual for rejection and eosinophils invading tubules
The etiology of the syndrome remains undefined, and oth- are identified, then drug allergy may be favored over
ers have performed combined kidney and bone marrow rejection. The presence of endarteritis permits a definitive
transplants without observing this phenomenon.124 With diagnosis of active rejection.55 Lymphocytes commonly
386 Kidney Transplantation: Principles and Practice

TABLE 25.3 Banff Classification (Updated 2017)135,157


(A) CLASSIFICATION CATEGORIES

CATEGORY 1: NONSPECIFIC CHANGES OR NORMAL BIOPSY

CATEGORY 2: ANTIBODY-MEDIATED CHANGES


Acute/active ABMR If all three features are present, they are considered diagnostic. If 1 and 2 or 1 and 3 below are present, a “suspicious”
designation can be made.a
1. Histologic evidence of injury:
□ Microvascular inflammation (g >0 in the absence of recurrent or de novo glomerulonephritis, and/or ptc >0)
□ Intimal or transmural arteritis (v >0)1
□ Acute thrombotic microangiopathy (TMA) in the absence of other causes
□ Acute tubular injury in the absence of other causes
2. Evidence of antibody interaction with vascular endothelium:
□ Linear C4d staining in peritubular capillariesb
□ ≥ moderate microvascular inflammation ([g + ptc] ≥ 2)c
□ Increased gene transcript/classifier expressiond
3. Serologic evidence of DSAs (HLA or other antigens)e:
Chronic active ABMR If all three features are present, they are considered diagnostic. If 1 and 2 or 1 and 3 below are present, a “suspicious”
designation can be made.a
1. Histologic evidence of injury:
□ TG (cg >0), if not evidence of chronic TMA [even if by EM only (cg1a)]
□ Severe peritubular capillary basement membrane multilayering2
□ Arterial intimal fibrosis of new onset, excluding other causes
2. Evidence of antibody interaction with vascular endothelium:
□ Linear C4d staining in peritubular capillariesb
□ ≥ moderate microvascular inflammation ([g + ptc] ≥ 2)c
□ Increased gene transcript/classifier expressiond
3. Serologic evidence of DSAs (HLA or other antigens)e
C4d staining without evi- Only if three features are present:
dence of rejection 1. Linear peritubular capillary C4d stainingb
2. Criterion 1 for active or chronic, active ABMR not met
3. No molecular evidence for ABMR (i.e., in criterion 2 for active and chronic, active ABMR
4. No acute or chronic active acute TCMR, or borderline changes
CATEGORY 3: BORDERLINE CHANGES SUSPICIOUS FOR ACUTE TCMR
□ Foci of tubulitis (t1, t2, or t3), with minor interstitial inflammation (i0 or i1) or interstitial inflammation (i2, i3) with mild
(t1) tubulitis; if designated in a report or publication, retaining the i1 threshold from Banff 2005 is permitted
□ No intimal arteritis (v = 0)

CATEGORY 4: TCMR
Acute TCMR (grade/type) 1A. Interstitial inflammation (>25% of nonsclerotic cortical parenchyma, i2 or i3) and foci of moderate tubulitis (t2)
1B. Interstitial inflammation (>25% of nonsclerotic cortical parenchyma, i2 or i3) and foci of severe tubulitis (t3)
2A. Mild to moderate intimal arteritis (v1) with or without interstitial inflammation and tubulitis
2B. Severe intimal arteritis comprising >25% of the luminal area (v2) with or without interstitial inflammation and
tubulitis
3. Transmural arteritis and/or arterial fibrinoid change and necrosis of the medial smooth muscle cells with accompa-
nying lymphocytic inflammation (v3)
Chronic active TCMR 1A. Interstitial inflammation [>25% of total cortex (ti score 2 or 3) and >25% of sclerotic cortical parenchyma (i-IFTA
(grade) score 2 or 3)] with moderate tubulitis (t2) involving ≥1 tubules, not including severely atrophic tubules3
1B. Interstitial inflammation [>25% of total cortex (ti score 2 or 3) and >25% of the sclerotic parenchyma (i-IFTA score 2
or 3)] with severe tubulitis (t3) involving ≥1 tubules, not including severely atrophic tubules3
2. Chronic allograft arteriopathy (arterial neointima, intimal fibrosis with mononuclear cell infiltration)1
Interstitial fibrosis and I. Mild IFTA (≤25% of cortical area)
tubular atrophy (IFTA, II. Moderate IFTA (26%–50% of cortical area)
grade) III. Severe IFTA (>50% of cortical area)
CATEGORY 6: CHANGES NOT CONSIDERED TO BE CAUSED BY CHRONIC OR ACUTE REJECTION (SEE “NONREJECTION INJURY” IN TABLE 25.1)
aDesignate if C4d positive or C4d negative.
bC4d2 or C4d3 by immunofluorescence on frozen sections or C4d0 >0 by immunohistochemistry on paraffin sections.
cAt least moderate (≥ moderate ) microvascular inflammation ([g + ptc] ≥2) can be sufficient for this requirement; however, in the presence of acute TCMR,

borderline infiltrate, or infection, ptc ≥2 alone is not sufficient, and g must be ≥1.
dIncreased gene transcript/classifier expression is considered sufficient for this requirement “if thoroughly validated” in biopsy tissue.
eDSAs can be substituted by C4d staining or expression of validated transcripts/classifiers in criteria 2; however, extensive DSA testing (including non-HLA anti-

bodies if HLA antibody testing is negative) is still advised if criteria 1 and 2 are not met.
ABMR, antibody-mediated rejection; DSA, donor-specific antibody; EM, electron microscopy; HLA, human leukocyte antigen; IFTA, interstitial fibrosis and tubular
atrophy; TCMR, T-cell-mediated rejection; TG, transplant glomerulopathy.
1These arterial lesions may be indicative of ABMR, TCMR, or mixed ABMR/TCMR.
2≥ seven layers in one cortical peritubular capillary and five or more in two additional capillaries, avoiding portions cut tangentially.
3Severely atrophic tubules have three features: (1) diameter <25% of unaffected or minimally affected tubules; (2) undifferentiated-appearing, flattened,

or cuboidal epithelium; and (3) pronounced wrinkling and/or thickening of the tubular basement membrane. Other known causes of i-IFTA should be
excluded.
25 • Pathology of Kidney Transplantation 387

TABLE 25.3 Banff Classification (Updated 2017)135,157—cont’d


(B) QUANTITATIVE CRITERIA
Quantitative Criteria
(Lesion) 0 1 2 3
i i0: none or trivial (<10% of i1: 10%–25% of unscarred i2: 26%–50% of unscarred i3: >50% of unscarred cortex
(interstitial Inflammation) unscarred cortex) cortex inflamed cortex inflamed inflamed
ti ti0: none or trivial (<10% ti1: 10%–25% of scarred and ti2: 26%–50% of scarred and ti3: >50% of scarred and
(total interstitial inflam- of cortex) unscarred cortex unscarred cortex unscarred cortex
mation)
i-IFTA i-IFTA0: no inflammation i-IFTA1: inflammation in i-IFTA2: inflammation in i-IFTA3: inflammation in
(inflammation in intersti- or <10% of scarred 10%–25% of scarred corti- 26%–50% of scarred >50% of scarred cortical
tial fibrosis and tubular cortical parenchyma cal parenchyma cortical parenchyma parenchyma
atrophy)
t t0: no tubular mono- t1: 1–4 cells/tubular cross t2: 5–10 cells/tubular cross > 10 cells/tubular cross
(tubulitis) nuclear cells section1 section1 section2
V v0: no arteritis v1: mild to moderate v2: severe arteritis with ≥25% v3: transmural arteritis and/
(arteritis) arteritis in ≥1 arterial cross luminal area lost in ≥1 arte- or fibrinoid change and
section rial cross section medial smooth muscle
necrosis with vascular
lymphocytic infiltrate
g g0: none g1: <25% of glomeruli g2: segmental or global in g3: mostly global in >75% of
(glomerulitis)3 25%–75% of glomeruli glomeruli
ptc4 ptc0: Absent or < 10% of ptc1: 3–4 luminal inflamma- ptc2: 5–10 luminal inflamma- ptc3: >10 luminal inflamma-
(peritubular capillaritis) cortical PTCs tory cells3 tory cells3 tory cells
ci ci0: ≤5% of cortical area ci1: 6%–25% of cortical area ci2: 26%–50% of cortical area ci3: >50% of cortical area
(interstitial fibrosis)
ct ct0: none ct1: ≤ tubular atrophy ct2: 26%–50% tubular ct3: >50% tubular atrophy
(tubular atrophy) atrophy
cg cg0: no GBM double cg1: GBM double contours cg2: Double contours in cg3: Double contours in
(transplant glomerulopa- contours in ≤25% of capillary loops5 26%–50% of capillary loops >50% of capillary loops
thy)
mm mm0: none mm1: ≤25% of nonsclerotic mm2: 26%–50% of nonscle- mm3: 50% of nonsclerotic
(mesangial matrix glomeruli rotic glomeruli glomeruli
increase)
cv cv0: arterial fibrous inti- cv1: arterial fibrous intimal cv2: arterial fibrous intimal cv3: arterial fibrous intimal
(arterial fibrous intimal mal thickening thickening with 1%–25% thickening with 26%–50% thickening with >50%
thickening)6 luminal narrowing luminal narrowing luminal narrowing
ah ah0: none ah1: mild-moderate in ≤1 ah2: moderate to severe in >1 ah3: Severe in many arteri-
(arteriolar hyalinosis) arteriole arteriole oles
aah aah0: no lesions typical of aah1: 1 arteriole, not circum- aah2: >1 arteriole, not circum- aah3: Any number of arteri-
(arteriolar hyaline CNI arteriolopathy ferential ferential oles, circumferential
thickening)7
C4d IF by immunofluores- C4d0: 0% of biopsy area, C4d1: 1 to <10% of biopsy C4d2: 10%–50% of biopsy C4d3: >50% of biopsy area,
cence considered negative area, considered minimal/ area, considered focal considered diffuse positive
negative unknown
C4d IHC by immunohisto- C4d0: 0% of biopsy area, C4d1: 1% to <10% of biopsy C4d2: 10%–50% of biopsy area, C4d3: >50% of biopsy area,
chemistry considered negative area, considered minimal/ considered focal positive considered diffuse positive
unknown
1Tubulitis can be considered per tubular cross section or per 10 tubular cells.
2t3 can also be diagnosed if or ≥ two areas of tubular basement membrane destruction accompanied by i2/i3 inflammation and t2 tubulitis elsewhere in the
biopsy.
3Complete or partial occlusion of ≥1 glomerular capillary by leukocyte infiltration and endothelial cell enlargement.
4Comment on extent (focal ≤ 50%; diffuse >50%) and composition (neutrophils and mononuclear cells).
5In the severely affected glomerulus; also note number and percent sclerotic. Furthermore, cg1a denotes no GBM double contours by light microscopy but GBM

double contours by electron microcopy (EM) with endothelial swelling and/or subendothelial electron lucent widening, and cg1b can denote ≥1 double
contours in ≥1 non-sclerotic glomerulus, confirmed by EM if available.
6Characterized by features of chronic rejection (fibrointimal thickening/neointima formation ± breach of internal elastic lamina or presence of occasional mono-

nuclear or foam cells, ± breaks in elastic lamina).


7Alternate scoring for hyaline arteriolar thickening (not always used diagnostically) due to calcineurin inhibitors (CNI).

Adapted from Farris AB. Histopathological syndromes of kidney allograft rejection and recurrent disease. In: Kirk A, Larsen C, Madsen J, Pearson T, Webber S, edi-
tors. Textbook of Organ Transplantation. Hoboken, NJ: John Wiley and Sons, Ltd; 2014. Loupy A, Haas M, Solez K, et al. The Banff 2015 kidney meeting report:
current challenges in rejection classification and prospects for adopting molecular pathology. Am J Transplant. 2017;17(1):28–41.

surround vessels (without medial involvement), a non- positive urine culture will also separate infection from
specific feature, and must not be confused with endar- rejection.131
teritis. Tubulitis is often present in atrophic tubules and Polyoma virus interstitial nephritis (BK virus) is often
does not indicate acute rejection. The diagnosis of acute diagnosed by the presence of the enlarged, hyperchro-
pyelonephritis should be raised when active inflamma- matic tubular nuclei with lavender viral nuclear inclu-
tion and abundant intratubular neutrophils are present. sions, often in collecting ducts. However, these may be
A note of caution though because in acute AMR, neu- inconspicuous, and diligent study of multiple sections
trophilic tubulitis with neutrophil casts can be seen; a may be required. Other clues are prominent apoptosis of
C4d stain will help in distinguishing between these. A tubular cells and abundant plasma cells, which invade
388 Kidney Transplantation: Principles and Practice

tubules. IHC for the polyoma SV40 large T antigen or in Diagnostic Criteria
situ hybridization for BK polyomavirus and EM (even of The three diagnostic criteria for acute AMR are (1) histo-
paraffin) will confirm the diagnosis. Sometimes BK virus logic evidence of acute injury (neutrophils in capillaries,
infection, with its exuberant plasmacytic infiltration and acute tubular injury, fibrinoid necrosis), (2) evidence of
activated immunoblasts may be confused with the plas- antibody interaction with tissue (typically C4d in PTCs), and
macytic hyperplasia form of posttransplant lymphopro- (3) serologic evidence of circulating antibodies to antigens
liferative disease,131 which also should be considered in expressed by donor endothelium (typically HLA).147,148,157
the differential diagnosis of acute cellular rejection. Rarer Criteria for the diagnosis of acute AMR have been refined
infections, including microsporidia, should also be con- over the years. Generally speaking, if only two of the three
sidered in biopsies with interstitial inflammation.132–134 major criteria are established (e.g., when antibody is nega-
tive or not done), the diagnosis can be considered suspicious
ACUTE ANTIBODY-MEDIATED REJECTION for acute AMR. Biopsies meeting criteria for both acute AMR
and TCMR type I or II are considered to have both forms of
Acute antibody-mediated rejection (also known as acute rejection. Biopsies with C4d and no pathology are likely a
humoral rejection, acute AMR, or, as referred to in the manifestation of “accommodation” (see later).131
latest Banff criteria: “active” AMR135) is a form of renal
allograft rejection due to damage by circulating antibod- Pathologic Features
ies that react to donor alloantigens on endothelium. These Histologic findings are typically scant to moderate mono-
antigens include HLA class I and class II antigens,14,136,137 nuclear interstitial infiltrates, sometimes with prominent
ABO blood group antigens,138 and other non-major his- neutrophils115,147,158,159 and increased numbers of macro-
tocompatibility complex (MHC) antigens,108,139 even in phages160 (see Fig. 25.3). The extent of mononuclear infil-
HLA-identical grafts.140 The main risk factors for donor- tration often does not meet the criteria for TCMR.159 PTCs
specific antibody (DSA; this term typically refers to anti- have neutrophils in about 50% of cases and are classically
HLA antibody) are blood transfusion, pregnancy, and prior dilated (Fig. 25.4A). Interstitial edema and hemorrhage
transplant.141 DSA may arise de novo in the posttransplant can be prominent. Glomeruli have accumulations of mac-
period, or alloantibody may be present before transplanta- rophages (∼50% of cases) and neutrophils (∼25% of cases;
tion in the case of positive crossmatch (+XM) or ABO blood see Fig. 25.3)115,147,159,161 and occasionally fibrin thrombi
group incompatible transplants with preconditioning regi- or segmental necrosis.115,136,147 Acute tubular injury,
mens to lower the alloantibody level before transplantation. sometimes severe, can be identified in many cases and
Hyperacute rejection is an immediate rejection that occurs may be the only initial manifestation of acute AMR. Focal
with high levels of preformed alloantibody directed against necrosis of whole tubular cross sections, similar to cortical
the graft. necrosis has been reported; 38% to 70% of acute AMR cases
Traditionally, identification of acute AMR in biopsies is may have patchy infarction.115,162 Little mononuclear cell
difficult because none of the histologic features is diagnostic, tubulitis is found, although a neutrophilic tubulitis with or
and immunoglobulin deposition was usually not detectable without neutrophil casts may be prominent,115 resembling
in the graft.142–144 Techniques for demonstrating C4d in acute pyelonephritis. Plasma cells can be abundant in acute
PTCs, pioneered by Feucht,145 have substantially improved AMR, either early59 or late163,164 after transplantation,
detection of this condition.14,144,146–148 Acute AMR may sometimes associated with severe edema and increased
occur in the absence of evidence for T-cell-mediated injury, IFNγ production in the graft.164 B cells can be also present,
particularly in +XM transplants;149–151 however, it is not but have no apparent diagnostic value.131
uncommon for both to be present, particularly in the later In about 15% of cases small arteries shows fibrinoid
posttransplant period (months to years).14 necrosis, with little mononuclear infiltrate in the intima or
Acute AMR typically presents with clinically severe acute adventitia but with neutrophils and karyorrhectic debris
rejection136 1 to 3 weeks after transplantation, but also (Fig. 25.5).115,162 Arterial thrombosis can be found in 10%,
can arise months to years later, associated with decreased and a pattern resembling TMA has also been reported.162
immunosuppression or noncompliance.152 With current Around 75% of cases with fibrinoid necrosis are C4d posi-
therapy, approximately 5% to 7% of recipients develop an tive.115,147,158,161 Presumably the C4d-negative cases had
episode of acute AMR, and about 25% of biopsies taken for T-cell-mediated rejection or TMA. Antibodies to the angio-
acute rejection have pathologic evidence of an acute AMR tensin II type 1 receptor have been detected in a few cases
component.16 The main risk factor is presensitization by with arterial fibrinoid necrosis, in the absence of capillary
blood transfusion, pregnancy, or prior transplant,141 how- C4d deposition.165 The presence of mononuclear endarteri-
ever, the majority have a negative crossmatch at the time of tis in cases of acute AMR strongly suggests a component of
transplantation.131 T-cell-mediated rejection.115
Serologic testing for DSA has become more sensitive in By EM the PTCs are dilated, containing neutrophils. The
the past decade due to the widespread use of solid-phase endothelium is reactive and shows loss of fenestrations.
assays rather than the older cell-based assays.153,154 The glomerular endothelium is separated from the GBM
These assays can be used before transplantation and for by a widened lucent space with endothelial cell swelling115
posttransplant monitoring for DSA. These more sensi- and loss of endothelial fenestrations, indicative of injury.
tive methods of detecting DSA have brought to light the Platelets, fibrin, and neutrophils are found in glomerular
spectrum of alloantibody-mediated damage (e.g., capil- and PTCs. The small arteries with fibrinoid necrosis show
laritis) that may not have been recognized in previous marked endothelial injury and loss, smooth muscle necro-
studies.155,156 sis, and deposition of fibrin.131
25 • Pathology of Kidney Transplantation 389

A B

C D

E
Fig. 25.4 Chronic allograft glomerulopathy. (A) widespread duplication of the GBM with mild mesangial hypercellularity and increased mononuclear
cells in the glomerular capillaries. PAS stain. Inset shows GBM multilamination at high power in a silver stain. (B) EM, high power of a glomerular capillary
showing duplication of the GBM; the new or second layer of GBM (short arrow) forms underneath the endothelium (E) and is separated from the old
GBM layer (long arrow) by the cellular (mononuclear or mesangial cell) interposition (*). C, capillary lumen; U, urinary space. (C) Immunohistochemistry
stain for C4d in paraffin sections shows prominent C4d deposition in glomerular and peritubular capillaries. (D) EM, high magnification of a PTC with
multilamination (arrow) of the basement membrane. Inset is a higher magnification of the area marked by arrow. E, endothelium; I, interstitium.

C4d Interpretation Although immunoglobulin deposition is found in only


Feucht and colleagues first drew attention to C4d as a pos- a minority of cases, C4d is characteristically detected in a
sible marker of an antibody-mediated component of severe widespread, uniform ring-like distribution in the PTCs by
rejection.145 C4d, a fragment of complement component immunofluorescence in cryostat sections14,145 (see Fig.
C4, is released during activation of the classical comple- 25.4B). Deposition occurs in both the cortex and medulla.
ment pathway by antigen–antibody interaction. Because Using IHC in formalin-fixed, paraffin embedded tissue,
C4d contains a thioester bond, it binds covalently to tis- C4d has a similar pattern, although the intensity is vari-
sues at the local site of activation. The covalent linkage able. “Serum staining” is an artifact of C4d IHC, so PTCs
explains why C4d remains for several days after alloan- must show clear circumferential staining to be called
tibody disappears, because antibody binds to cell surface positive by this technique. Glomerular capillary staining
antigens that can be lost by modulation, shedding, or cell also occurs but is hard to distinguish from C4d normally
death. found in the mesangium in frozen sections stained by
390 Kidney Transplantation: Principles and Practice

Other components of the complement system have been


sought. C3d, a degradation product of C3, was found in PTCs in
39% to 60% of biopsies from HLA-mismatched grafts with dif-
fuse C4d.158,168,172,182 C3d was usually172 but not always182
associated with C4d. C3d correlated with acute AMR in all
studies, and was associated with increased risk of graft loss in
two series, compared with C3d-negative cases, but C3d pro-
vided no convincing additional risk compared with C4d+. The
interpretation of C3d stains is complicated by the common
presence of C3d along the TBM.172 Even though C3d should
indicate more complete complement activation, it added no
diagnostic value to C4d in grafts showing histologic features
of acute AMR, except in the setting of ABO-incompatible
grafts.172 Other complement components, such as C1q, C5b-
9, and C-reactive protein (CRP) are not conspicuous in PTCs
in acute rejection.183,184 Lectin pathway components, which
activate C4 by binding to microbial carbohydrates, are some-
Fig. 25.5 Fibrinoid arterial necrosis: an arteriole with destruction of the times detected.168,185 Among 18 biopsies with C4d, 16 had
medial wall smooth muscle cells by fibrinoid necrosis. Some neutro-
phils are present underneath the reactive and swollen endothelium.
diffuse H-ficolin deposition along the PTCs, whereas none of
This vascular change is distinctly different from endarteritis (compare the 42 cases without C4d had H-ficolin. No Mannan-binding
with Fig. 25.2) and can be seen in both acute humoral rejection and lectin serine protease 1 (MASP-1, also known as mannose-
type III acute rejection. This case had positive C4d. associated serine protease 1) or MASP-2 was detectable.185
The significance of this observation is not clear, because MASP
immunofluorescence. Formalin fixation eliminates this proteins are required to activate C4 via the ficolins or Mannose-
background staining and demonstrates glomerular C4d in binding lectin (MBL).185
about 30% of acute AMR cases.159 Natural killer (NK) cells have been the focus of recent
Grafts with focal C4d (<50% of PTC) are of uncertain research in graft injury, particularly regarding AMR.186,187
significance and the patient should be monitored closely Microarray analysis has indicated that several DSA-specific
for donor-reactive antibodies. Two of three studies have gene transcripts show high expression in NK cells, and IHC
failed to show any significant clinical or pathologic dif- also shows prominent numbers of peritubular capillary
ference between cases with focal and diffuse C4d stain- NK cells in these cases.188 Depletion of NK cells with anti-
ing.161,163,166 C4d deposition can precede histologic NK1.1 significantly reduced DSA-induced chronic allograft
evidence of acute AMR by 5 to 34 days.167 C4d in 1-week vasculopathy in a murine cardiac allograft model.186,189
protocol biopsies was followed by clinical acute rejection
in 82% of cases168 and was associated with donor-reactive
antibodies.169
C4d Negative Antibody-Mediated
In the setting of acute rejection, C4d is a specific (96%) Rejection
and sensitive (95%) marker of circulating antidonor HLA-
specific antibodies by the antihuman globulin cytotoxicity Attention has recently been drawn to “C4d-negative”
test.170 PTC C4d deposition is associated with concurrent AMR, and recent Banff allograft classification documents
circulating antibodies to donor HLA class I or II antigens encourage that cases be designated as “C4d positive” or
in 88% to 95% of recipients with acute rejection.147,171,172 “C4d negative.”157,190 These cases have DSA and vary-
Moreover, C4d deposition and the severity of histologic ing degrees of morphologic evidence of antibody-mediated
injury by antibody correlates with the serum DSA level in injury but lack detectable C4d deposition in PTC endothe-
acute humoral rejection.149 False negative antibody assays lium.191–193 Morphologic signs of injury with concurrent
may be due to absorption by the graft as shown by elu- DSA positivity have been identified, particularly in sensi-
tion from rejected grafts in patients who had no detectable tized patients early after transplantation.191,194 Negativity
circulating antibody,173 or it may be due to differences in for C4d in AMR can be explained by various mechanisms:
detection of antibody directed against different HLA alleles time-dependent degradation of C4d-deposits in the micro-
and sensitivity of solid-phase methods for particular allo- circulation, complement independent antibody-mediated
antigens. Alternatively, non-HLA antigens may be the tar- injury, lack of sensitivity and reproducibility of the stain-
get.16,174–179 C4d-negative acute rejection may show flow ing methods, arbitrary criteria for defining “positivity,”
cytometry evidence of antidonor reactive antibodies as fre- and acute tissue injury due to nonrejection causes with
quently as 50%,171 due in part to noncomplement fixing incidental chronic alloantibody-associated changes (capil-
antibodies.180 Cell based assays have a false positive rate of laritis; usually C4d negative).193 Molecular studies have
<10%.147 uncovered a subset of cases with morphologic features
In a comparison of methods for C4d, the triple layer of antibody-mediated injury and DSA showing increased
immunofluorescence technique14 proved the most sensi- endothelial cell associated transcript expression, indicative
tive, although the difference with IHC in paraffin embedded of endothelial cell activation and stress. These data suggest
tissue was small.181 With fixed tissue, plasma in the capil- that 50% to 60% of AMR cases are missed by current Banff
laries and interstitium may stain for C4d, which interferes criteria due to C4d negativity,195 although many of these
with interpretation.131 cases may be chronic alloantibody-mediated endothelial
25 • Pathology of Kidney Transplantation 391

TABLE 25.4 Differentiation Between Acute Rejection AMR group.147 However, some of those who recover from
and Acute Calcineurin Inhibitor Toxicity the acute episode of acute AMR have a similar long-term
outcome,115 suggesting that the pathogenetic humoral
Acute Rejection CNI Toxicity response can be transient if treated effectively.
INTERSTITIUM
Infiltrate Moderate–marked Absent–mild
Accommodation
Edema Usual Can be present The process termed “accommodation” is a peculiar scenario
TUBULES related to AMR. Accommodation refers to the presence of
Tubular injury Usual Usual PTC C4d deposition in the absence of other evidence of
Vacuoles Occasional Common antibody-mediated injury and in the presence of normal or
Tubulitis Prominent Minimal–absent stable graft function. Accommodation is thought to repre-
ARTERIOLES sent a process of endothelial cell adaptation to antibody and
Endothelialitis Can be present Absent complement over time. In accommodation, donor-specific
Smooth muscle degeneration Absent Sometimes present antibodies may be detectable; however, morphologic signs
Mucoid intimal thickening Absent Sometimes present of tissue injury are absent. There are no signs of acute or
with red cells (TMA)
chronic TCMR or AMR; more specifically, there is no ATN-
ARTERIES like minimal inflammation, no glomerulitis [g0], no chronic
Endothelialitis Common Absent (rare mono- transplant glomerulopathy [cg0], no peritubular capillari-
nuclear TMA) tis [ptc0], and no PTC basement membrane multilamina-
PERITUBULAR CAPILLARIES tion. Current Banff criteria refer to this situation as “C4d
C4d May be positive Negative deposition without evidence of active rejection.” If there
GLOMERULI are simultaneous borderline changes, the cases can be con-
Mononuclear cells Often Rare sidered to be indeterminate.157,190,200 Accommodation is
Thrombi Occasional Occasionally common in the setting of ABO-incompatible allografts, with
prominent (TMA) at least 80% of normal surveillance biopsies showing C4d
deposition in PTCs.172 It appears that antibodies against
CNI, calcineurin inhibitor; TMA, thrombotic microangiopathy.
blood group antigens (i.e., ABO-incompatible allografts)
are mostly not injurious to allografts with “accommoda-
injury and represent a different mechanism of injury from tion”; however, allografts with “accommodation” having
acute AMR, which is likely more complement mediated.196 anti-HLA antibodies may progress to chronic AMR, given
Eventually, C4d-negative AMR will likely be added to the enough follow-up surveillance.157 The long-term signifi-
Banff diagnostic armamentarium as a distinct category of cance of these relatively uncommon cases is still under
AMR; however, data are still being gathered by a respec- investigation.138,201–203
tive Banff Working Group regarding the significance of this
entity in an attempt to provide diagnostic criteria.17 Complement Inhibition
Although most approaches for treatment or prevention of
Differential Diagnosis acute AMR involve removing alloantibody from the circu-
For differential diagnosis, it is helpful that both ATN144,197 lation (by plasmapheresis) or decreasing production of allo-
and TMA in native kidneys are C4d negative. Among 26 antibody (e.g., by antiplasma cell drugs), another technique
cases of TMA/hemolytic-uremic syndrome in native kid- to prevent graft damage by antibody is by inhibiting com-
neys, none had positive C4d, including cases with lupus plement. Eculizumab, a humanized monoclonal antibody
anticoagulant and antiphospholipid antibodies.144 In five directed against the terminal complement component C5,
cases of recurrent hemolytic-uremic syndrome in trans- is now being applied in renal transplantation, particularly
plant recipients, C4d was also negative.198 Among native in sensitized (+XM) patients at a high risk for early acute
kidney diseases, only lupus nephritis144,199 and endocardi- AMR. C5 is downstream of C4d in the complement cascade;
tis199 have been reported to have PTC C4d. Glomerular C4d thus, with DSA activation of complement, diffuse C4d depo-
deposits are not specific because they occur in many forms sition would be expected even with effective C5 inhibition.
of immune complex glomerulonephritis in native kidneys. Early surveillance biopsies in eculizumab-treated patients
Arterial intimal fibrosis often stains for C4d, even in native showed diffuse C4d deposition but absent morphologic signs
kidneys and should not be taken as evidence of AMR.144 of acute AMR, including a lack of endothelial cell activation
The comparative features of “pure” humoral and TCMR by EM. The absence of respective pathology suggests endo-
are given in Table 25.4. In acute AMR neutrophils are thelial protection by eculizumab, and moreover supports
the predominant inflammatory cells in PTCs, glomeruli, the notion that most cases of early acute AMR are comple-
tubules, and the interstitium, with or without accompany- ment mediated. However, acute AMR has been observed
ing fibrinoid necrosis. The vascular lesion of acute AMR, if despite eculizumab therapy and may be due to IgM DSA
present, is fibrinoid necrosis of the wall; whereas, in TCMR, not detected by the usual DSA testing methods.204,205 Nota-
endarteritis is the usual lesion. C4d deposition in PTCs bly, a subset of patients still developed features of chronic
(immunofluorescence microscopy) is typically only present humoral rejection (chronic AMR), including transplant
in acute AMR but not in TCMR.131 glomerulopathy (TG).196 Although effective in preventing
The prognosis of acute AMR is uniformly worse than early acute AMR in +XM transplants, it appears that com-
TCMR.14,108,115,136,139,162 In one series, 75% of the 1-year plement inhibition alone does not entirely prevent chronic,
graft losses from acute rejection were in the C4d+ acute antibody-mediated microcirculation injury. Furthermore,
392 Kidney Transplantation: Principles and Practice

the diagnostic reliability for acute AMR of C4d and serum definitions are faulty. Despite these considerations, Banff is
DSA are apparent in this setting, suggesting that diagnostic fully accepted as a scoring system of drug trials and is used
criteria refinements are needed (See Chapter 22). widely in clinical practice (although not necessarily with an
individual score report).131
CLASSIFICATION SYSTEMS
The most widely used system currently is the Banff work- Late Graft Diseases
ing schema (Banff). Banff started as an international
collaborative effort led by Kim Solez, Lorraine Racusen, Although acute rejection has diminished in clinical impor-
and Philip Halloran to achieve a consensus that would tance in the past decade, allografts are still lost by slow,
be useful for drug trials and routine diagnosis.10,17,206 progressive diseases that cause a 3% to 5% annual attrition
Banff is still growing and remodeling, undergoing revi- rate. The specific causes of this are many and sometimes dif-
sions based on data presented, and debated at the bien- ficult to ascertain, particularly if only an end-stage kidney
nial Banff meeting. These include restructuring that is examined. Unfortunately, the two terms “chronic rejec-
separated the category of endarteritis, according to the tion” and “CAN” have been used in past literature to lump
National Institutes of Health (NIH) Cooperative Clinical together these myriad diseases. The role of the pathologist
Trials in Transplantation (CCTT) criteria,11,207 the addi- in interpreting the biopsy is to provide the most specific
tion of acute148 and chronic AMR,208 and the birth10 and diagnosis possible and indicate the activity of the pro-
death208 of chronic allograft nephropathy (CAN).209 cess. Although some have argued that the renal biopsy is
Banff scores three elements to assess acute rejection: not useful in analyzing graft dysfunction after 1 year, the
tubulitis (t), the extent of cortical mononuclear infiltrate data show that in 8% to 39% of patients the biopsy led to
(i), and vascular inflammation (intimal arteritis or trans- a change in management that improved renal function.2,3
mural inflammation) (v). Mononuclear cell glomerulitis Here we will discuss the criteria used to distinguish some of
(g) is scored but not yet part of the classification of rejec- these diseases and those that remain idiopathic. The term
tion. Banff recognizes three major categories of acute T-cell- “chronic rejection” is best defined as chronic injury primar-
mediated rejection (tubulointerstitial, endarteritis, and ily mediated by an immune reaction to donor alloantigens.
arterial fibrinoid necrosis; see Table 25.2). The threshold
for type I (tubulointerstitial) TCMR is >25% cortical mono- CHRONIC ANTIBODY-MEDIATED REJECTION
nuclear inflammation in the nonatrophic areas, provided
tubulitis of at least 5 to 10 cells/tubule is present.207 Cases Circulating anti-HLA antibodies have been associated
with no tubulitis, regardless of the extent of infiltrate, are with increased risk of late graft loss.216,217 Chronic, active
not considered TCMR. Biopsies with C4d+ PTCs are con- antibody-mediated rejection (chronic humoral rejection,
sidered to have an additional component of AMR, which chronic AMR) is now recognized as a separate category in
occurs in 20% to 30% of cases.210 Cases with tubulitis are the Banff schema. Chronic AMR differs from acute AMR in
termed “suspicious for rejection” or “borderline” in the cur- the usual lack of evidence of acute inflammation (thrombi,
rent Banff system. Many, but not all, of these cases are early necrosis, mostly neutrophilic capillaritis), and the presence
or mild acute rejection: 75% to 88% of patients with sus- of matrix synthesis (basement membrane multilamination,
picious/borderline category and graft dysfunction improve fibrosis in arterial intima and the interstitium). Chronic
renal function with increased immunosuppression,211,212 AMR commonly arises late (>6 months after transplanta-
comparable to the response rate in type I rejection (86%).211 tion) and may occur in patients with or without a history
A minority (28%) of untreated suspicious/borderline cases of acute AMR, although C4d in early biopsies is a risk fac-
progress to frank acute rejection in 40 days.213 Almost all tor for later TG with C4d.218–221 In the setting of de novo
with suspicious/borderline findings do well, provided there DSA, many patients have reduced levels of immunosup-
is no element of concurrent AMR, which commonly has pression (absorption, iatrogenic or noncompliance).222 In
a suspicious/borderline pattern, although care must be these cases, a combination of chronic AMR and acute AMR
taken not to misinterpret peritubular capillaritis as inter- may be seen, along with a component of T-cell-mediated
stitial inflammation.155,159 The suspicious category is not rejection.151
counted as acute rejection in many clinical trials, a major The criteria of chronic AMR are the triad of: (1) one of
omission in our opinion. the following morphologic features, TG (duplication or
The interobserver reproducibility of the present Banff “double contours” in glomerular basement membranes),
classification is sufficient but needs improvement. In a multilamination of the PTC basement membrane, PTC loss
Canadian study, the agreement rate for rejection was 74%, and interstitial fibrosis (IF), or chronic arteriopathy with
but there was only 43% agreement on the suspicious/bor- fibrous intimal thickening (without duplication of the inter-
derline cases,214 similar to a European series.215 Among nal elastica); (2) diffuse C4d deposition in PTCs; and (3) cir-
a group of 21 European pathologists, the agreement rate culating DSA. If only two elements of the triad are present,
was poor for all of the acute Banff scores (t, i, v, g) in trans- the diagnosis is considered “suspicious.” Although helpful
plant biopsy slides (all kappa scores <0.4).28 Agreement when positive, C4d deposition and serum DSA are particu-
for t and v scores improved significantly when participants larly problematic in the chronic setting. They are less sensi-
were asked to grade a lesion in a photograph (kappa scores tive markers due to serum DSA level variability with time
of 0.61 and 0.69, respectively), arguing that the challenge posttransplant. Two features point to ongoing immuno-
is primarily finding the lesion in the glass slide. Lack of logic activity: the presence of C4d and mononuclear cells in
improvement in the other categories (g, i) argues that the glomerular and PTCs. Scoring of multilamination requires
25 • Pathology of Kidney Transplantation 393

EM, not always available in transplant biopsies, and quan- slightly thickened, attributable to compensatory hyper-
titative assessment of the number of layers, because to dis- trophy. With immunohistochemical techniques in paraf-
tinguish from other common causes of lamination, more fin sections, C4d is present along the glomerular capillary
than approximately six layers have to be present.223,224 To walls in about 10% to 30% of cases.218,235 Extensive cres-
be specific for AMR, current Banff criteria recommend that cents or diffuse immunoglobulin deposits are unusual and
seven or more layers should be present in one peritubular suggest recurrent or de novo glomerulonephritis.236–238
capillary and five or more layers be present in two additional It is now recognized that approximately 30% of TG due to
capillaries,157,190 and this is largely based on one study.224 chronic AMR are C4d negative.239 Notably, although most
In assessing peritubular capillary basement membrane cases of TG are due to chronic AMR, this pattern is also seen
multilamination by EM, peritubular capillary basement in allografts with chronic thrombotic microangiopathy and
membranes cut tangentially should be avoided. A Banff in patients with hepatitis C infection.225
Working Group is currently engaged in efforts to refine the
assessment of peritubular capillaries and other features Peritubular Capillary and Tubulointerstitial Lesions
by EM.157 Duplication of the GBM has many other causes, PTCs may be dilated and prominent, with thick basement
such as TMA and MPGN; however, these do not have C4d membranes, or may altogether disappear, leaving only
in PTC unless there is more than one concurrent pathologic occasional traces of the original basement membrane
process.225 A sequence of four stages of development of behind.240,241 In a subset of patients, PTCs have prominent
chronic AMR has been demonstrated in protocol biopsies C4d deposition (see Fig. 25.4C), which is associated with cir-
of nonhuman primate renal allografts. The process begins culating antidonor HLA class I or II reactive antibodies.242
with antibody production, followed by C4d deposition, and Other allografts with chronic AMR features may show focal
later, morphologic and functional changes.226 Validation of or multifocal C4d staining of PTCs by immunofluorescence
these processes has recently been provided by gene expres- or IHC or dim C4d staining by immunofluorescence. In
sion profiling.227 Proof that antibody is sufficient to initiate studies of protocol biopsies in graft recipients with DSA, rec-
allograft arterial intimal fibrosis has been shown by passive ognition of peritubular capillaritis has come to light as a fea-
transfer of anti-MHC antibody into immunologically defi- ture of early chronic humoral rejection.156,243 Peritubular
cient mice (RAG-1 knockout) bearing cardiac allografts.228 capillaritis, with or without C4d deposition, is commonly
seen as a subclinical rejection feature in patients with DSA
Transplant Glomerulopathy in otherwise stable grafts. Peritubular capillaritis is associ-
TG (chronic allograft glomerulopathy, given a cg score in ated with later development of TG, with a greater risk of TG
the Banff system) increases in frequency from 1 to 5 years in patients with C4d deposition,156 likely reflecting a more
posttransplant (5%–14% of protocol biopsies) and affects active chronic humoral rejection process in those grafts.
graft survival more adversely than IF and inflammation.229 EM reveals splitting and multilayering of the PTC basement
TG has been associated with prior episodes of acute rejec- membrane (see Fig. 25.4D), first described by Monga.244,245
tion, pretransplant hepatitis C antibody positivity, and Each ring probably represents the residue of one previous
anti-HLA antibodies (especially anti-class II), with the episode of endothelial injury going from oldest (outer) to
risk increasing if the antibodies were donor specific.204,220 most recent (inner). Quantitation is necessary to establish
Patients with preformed DSA (+XM grafts) have a particu- diagnostic specificity. Only in chronic rejection were three
larly high risk of TG long term, present in 55% of all surviv- or more PTCs found with five to six circumferential layers or
ing grafts at 5 years, and in 85% of surviving grafts with one PTC with seven or more circumferential layers.223 PTC
anti-HLA class II DSA.204 lamination correlates with TG,223,245 C4d deposition,218
TG is defined as duplication of the GBM with modest and loss of PTCs.241 Marked multilamination (five to six lay-
mesangial expansion, in the absence of specific de novo or ers in three capillaries or more than six in one) was found
recurrent glomerular disease. TG is best revealed in PAS or in 50% of cases with IF that lacked arterial or glomerular
silver stains (see Fig. 25.4A). The glomeruli may show an changes, and may point to past episodes of rejection as the
increase in mesangial cells and matrix with various degrees cause of the fibrosis.245
of scarring and adhesions. In some cases, mesangiolysis Interstitial fibrosis and tubular atrophy (IFTA) is a regu-
or webbing of the mesangium may be prominent as well lar, but nonspecific feature of chronic AMR and does not
as segmental or global sclerosis. EM reveals duplication or serve to distinguish rejection from other causes, such as cal-
multilamination of the GBM (see Fig. 25.4B), often accom- cineurin inhibitor (CNI) toxicity or previous BK polyoma-
panied by cellular (mononuclear or mesangial cell) inter- virus infection. Atrophic tubules typically have thickened,
position, widening or lucency of the subendothelial space, duplicated TBMs and intratubular mononuclear cells and
and a moderate increase in mesangial matrix and cells.230 mast cells.246 This should not be confused with the tubulitis
Glomeruli may show focal and segmental scarring (FSGS), of acute rejection. The TBM not uncommonly has deposi-
especially in more advanced TG, and some cases with col- tion of C3 in a broad segmental pattern. This is an exag-
lapsing FSGS lesions have been observed. EM detects 40% geration of similar changes found in normal kidneys and
more cases of TG than light microscopy.223 The GBM typi- probably represents a residue from prior episodes of tubular
cally has rarefactions, microfibrils, cellular debris but few injury, or possibly a persistent chronic injury. The inter-
or no deposits.231–233 Endothelial cells may appear reac- stitium typically has a sparse mononuclear infiltrate, with
tive with loss of fenestrae, probably undergoing “dediffer- small lymphocytes, plasma cells, and mast cells.247 Nodu-
entiation.”232–234 Podocyte foot process effacement ranges lar collections of quiescent-appearing lymphoid cells are
from minimal to quite extensive,232 corresponding to the sometimes found around small arcuate arteries. Abundant
degree of proteinuria. The nonduplicated GBM may become plasma cells may be present.248
394 Kidney Transplantation: Principles and Practice

Transplant Arteriopathy
Arterial lesions may be a manifestation of chronic AMR.249
Alloantibodies to graft class I antigens are a specific risk fac-
tor for chronic transplant arteriopathy (TA) in human renal
allografts.250,251 Typically, TA is recognized by thickening
of the arterial intima with mononuclear inflammatory cells
(CD3+ T cells or CD68+ monocytes/macrophages) within
the thickened intima. In a recent study, patients with pre-
formed DSA showed accelerated arteriosclerosis on serial
biopsies.249,252 Although the TA lesions were attributable
to DSA on serial biopsies from the same allografts, TA such
as that due to chronic AMR may not be distinguishable from
the arterial intimal thickening seen in hypertension,249 and
other biopsy and serologic features are needed to attribute
the lesion to chronic AMR.249,252 Experiments in animals
show that TA can be initiated by passive transfer of donor-
reactive MHC antibodies in recipients with no functional Fig. 25.6 Chronic allograft arteriopathy: an interlobular artery with
T cells, a complement-independent process mediated by prominent intimal fibroplasia. The presence of scattered mononuclear
NK cells.218,242 cells in the intima and the lack of duplication of the internal elastica
are characteristic of chronic rejection. This biopsy was positive for C4d.

CHRONIC T-CELL-MEDIATED REJECTION


This category is not well developed and subject to refine- in elastin stains. Fibroelastosis, typical of hypertensive,
ment. Using the chronic AMR model, the current Banff clas- atrophic and aging arterial changes, provides a useful dif-
sification defines “chronic active T-cell-mediated rejection” ferential diagnostic feature from rejection. Foamy macro-
as showing morphologic features of chronicity (arterial phages containing lipid droplets are sometimes seen along
intimal fibrosis without elastosis) combined with features the internal elastica and can be found as early as 4 weeks
indicative of ongoing T cell activity (mononuclear cells in after transplantation. Fibrin is sometimes deposited in a
the intima). IF with a mononuclear infiltrate and tubulitis band-like subendothelial location or mural thrombus. Focal
is, in some instances, part of this condition, as surveillance myocyte loss from the media occurs, as shown in mouse
follow-up biopsies after an episode of acute cellular rejection and rat studies.260 Immunofluorescence often shows IgM,
not uncommonly show continued inflammation.253 How- C3, and fibrin (and sometimes IgG) along the endothelium,
ever, at present the arterial lesions are the most definitive. in the intima, or in the media, as a diffuse blush or focal
It is anticipated that molecular gene expression studies will granular deposits.231,250,261–263
help in the future to document the activity of the infiltrate. The endothelium expresses increased adhesion mol-
Other nonspecific features that are commonly present in ecules, notably ICAM-1 and VCAM-1. Antagonism of
association with transplant arteriopathy are loss of PTCs ICAM-1 binding/expression inhibits chronic rejection264
and IFTA.241 and in humans certain ICAM-1 genetic polymorphisms
Small and large arteries, as early as 1 month after trans- (e.g., exon 4, the Mac-1 binding site) appear to confer higher
plantation, can begin to develop severe intimal proliferation risk for chronic rejection.265 The endothelium remains of
and luminal narrowing.249,254,255 The intimal change is donor origin,266,267 however, some of the spindle-shaped
most prominent in the larger arteries, but can be seen at all cells that contribute to the intimal thickening are of recipi-
levels, from interlobular arteries to the main renal artery. ent origin.257,268 The myointimal cells stain prominently
The intima shows pronounced, concentric fibrous thicken- for smooth muscle actin, sometimes so strikingly that a
ing with invasion and proliferation of spindle-shaped myofi- “double media” seems to be formed.269 This phenomenon
broblasts (Fig. 25.6). This vascular change has been termed has also been described as the development of a new artery
“chronic transplant arteriopathy” and when combined with inside and concentric with the old,270 with elastic laminae
an infiltrate of mononuclear cells in the intima, is character- and a muscular media, separated from the old internal elas-
istic of chronic T-cell-mediated rejection (Fig. 25.7). Suben- tic lamina poorly by cellular tissue. By EM, the thickened
dothelial mononuclear cells are one of the most distinctive intima consists of myofibroblasts, collagen fibrils, basement
features, and this suggests that the endothelium itself is a membrane material, and a loose amorphous electron-lucent
target. T cells (CD4+, CD8+, CD45RO+), macrophages, and ground substance.271 The matrix consists of collagen, fibro-
dendritic cells infiltrate the intima.256–258 T cells express nectin, tenascin, proteoglycans (biglycan and decorin), and
cytotoxic markers, including perforin259 and GMP-1767 acid mucopolysaccarides.272–274 Fibronectin has the extra
and markers of proliferation (proliferating cell nuclear domain of cellular fibronectin extra domain A (EDA), typi-
antigen [PCNA]).258 No B cells (CD20) are detected.258 It is cal of embryonic or wound healing fibronectin.272 Several
imagined that this is a dampened version of the endarteritis growth factors/cytokines have been detected. Platelet-
of acute rejection. As noted previously, recent studies have derived growth factor (PDGF) A chain protein is primarily
also indicated that chronic vascular lesions can be acceler- in endothelial cells, whereas the B chain is in macrophages
ated by the presence of alloantibody.249,252 and smooth muscle cells.275 Enhanced PDGF B-type recep-
The second distinctive feature is the lack of multilamina- tor protein was found on intimal cells and on smooth
tion of the elastica interna (fibroelastosis), best appreciated muscle cells of the proliferating vessels.276 FGF-1 and its
25 • Pathology of Kidney Transplantation 395

A B
Fig. 25.7 CNI arteriolopathy. (A) Several arterioles with peripheral nodular hyalinosis, where hyalin deposits replace necrotic/apoptotic smooth muscle
cells in the outermost media. (B) EM, an artery that has “beads” of hyalin (*) along the outer media. L, arteriolar lumen; T, tubule. (PAS 800×; EM 2700×.)

receptor are present in the thickened intima.277 TNFα is in TABLE 25.5 Differentiation Between Acute Humoral
the smooth muscle of vessels with chronic rejection, in con- Rejection and Acute Cellular Rejection
trast to normal kidneys.278
Acute Humoral Acute Cellular
The T-cell-mediated arterial lesions can be divided into Rejection Rejection
three stages, which probably differ in mechanism and
reversibility.234 The stage I lesion is endarteritis, charac- INTERSTITIUM
Infiltrate Variable Moderate–severe
teristic of type II TCMR. This lesion lacks matrix formation. Edema Present Present
This acute stage is believed to be T-cell-mediated endothe- Peritubular capillaries Neutrophils Mononuclear cells
lial injury. Stage II lesions have intimal matrix production C4da Positive Negative
and accumulation of myofibroblasts forming a “neointima.” TUBULES
This stage also contains mononuclear cells (T cells and mac- Acute tubular necrosis Can be present Usually absent
rophages), believed to be active in the intimal proliferation Tubulitis Can be neutro- Mononuclear cell
and accumulation of matrix. Intermediate stages between philic
stage I and II lesions are sometimes found, with lympho- VESSELS
cytes admixed with fibrin and fibromuscular proliferation, Endarteritis Can be present Present in type II
and are well documented in a nonhuman primate model Fibrinoid necrosis Can be present Present in type III
of chronic rejection.279 Secondary factors probably become GLOMERULI
increasingly important as the lesion progresses to stage III, Inflammatory cells Neutrophils Mononuclear cells
where the intima is fibrous and inflammatory cells are scant. Fibrinoid material/necrosis Can be present Typically absent
A fourth category resembling natural atherosclerosis with
cholesterol clefts and calcification has also been proposed.258 aC4d staining in peritubular capillaries indicates activation of the classical
A large body of experimental evidence supports the concept complement pathway by humoral antibody (monoclonal antibody,
immunofluorescence microscopy).
that the arterial lesions are immunologically mediated234:
(1) the lesions do not routinely arise in isografts; (2) the tar-
get antigens can be either major or minor histocompatibility antibodies likely conspire to accelerate the process of allograft
antigens;260,280,281 (3) the specific initiator is probably T cells arteriopathy/arteriosclerosis.249,252
followed by antibody (antibody is necessary and sufficient for Recent data indicate that allograft deterioration is accel-
the fibrous lesion in mice); (4) the target cell is probably the erated by inflammation in scarred areas as well as unscarred
endothelium, but the smooth muscle may also be affected; (5) areas287,288 in contrast to some of the past thought, which
secondary nonimmunologic mechanisms analogous to those tended to disregard inflammation in scarred areas. For
in atherosclerosis are important in the progression of the this reason, recent Banff classification added a new score,
lesion; and ultimately (6) the process may be independent of i-IFTA, which takes into account inflammation in areas
specific antidonor immunologic activity. T cells are sufficient of interstitial fibrosis and tubular atrophy. Furthermore,
to initiate cellular vascular lesions in B cell deficient mice, but a Banff Working Group on T-cell-mediated rejection was
these lesions do not readily progress to fibrosis in the absence formed to consider incorporation of i-IFTA into rejection
of antibody.282 Fibrous lesions are also markedly reduced classification and possible elimination of the “borderline”
in strain combinations that fail to elicit a humoral antibody category; this working group is reevaluating thresholds for
response. The best evidence for T cell mechanisms of chronic inflammation and tubulitis (t) and considering the addi-
allograft injury in humans is that subclinical or late clinical tion of other findings (e.g., edema) in the diagnosis of rejec-
cellular rejection is associated with progressive graft fibrosis tion.157 Along these lines, as shown in Table 25.5, the most
and dysfunction,283–285 and endarteritis is associated with recent Banff meeting has now specified criteria for a diagno-
later transplant arteriopathy.286 As mentioned previously, sis of chronic active TCMR.135
396 Kidney Transplantation: Principles and Practice

present (notably chronic glomerular or arterial lesions).


OTHER SPECIFIC DIAGNOSES
However, the unintended consequence was that “CAN”
The other conditions that can be diagnosed by a renal itself became a diagnosis that inhibited search for specific
biopsy that cause slowly progressive graft dysfunction and and perhaps treatable causes. CAN was replaced in Banff
loss are: calcineurin inhibitor toxicity (CNIT), hypertensive 2005 with category 5: “IF and TA, no evidence of any spe-
vascular disease, PTN, recurrent disease, de novo glomeru- cific etiology.” This now includes only those cases for which
lar disease, obstruction, and renal artery stenosis.16 no specific etiologic features can be defined, and excludes
Chronic CNIT is most specifically diagnosed by the pres- those with pathologic features of chronic AMR, chronic
ence of nodular hyaline replacement of individual smooth CNIT, hypertensive renal disease, PTN, obstruction, or
muscle cells, which may form distinctive deposits on the other de novo or recurrent renal disease.209
outer side of the arteriole, as described by Mihatsch as cyclo-
sporin arteriolopathy. Ordinary hyalinosis due to diabetes,
hypertension, or aging typically is subendothelial.289–291 Protocol Biopsies
To distinguish intimal fibrosis due to hypertension from
that due to chronic rejection, an elastin stain is valuable, “Protocol” or “surveillance” biopsies taken at predetermined
because in hypertension, but not necessarily in rejection, times for evaluation of the status of the renal allograft, inde-
the elastica interna is multilayered (“elastosis”), and in pendent of renal function, are currently the standard of care
chronic rejection the elastica is not duplicated, but may be at several leading transplant centers229,283,284,292–295 and
fractured. A recent study, however, suggested that some widely used in clinical trials to evaluate efficacy.296 Proto-
lesions of vascular intimal thickening due to alloantibody col biopsies have the potential ability to reveal mechanisms
are indistinguishable from those due to hypertension.249 of late graft loss and to identify active processes that might
Foam cells and mononuclear cells in the intima also favor be interrupted therapeutically before irreversible injury has
rejection. The features that point to a component of chronic occurred.297 The risk of protocol biopsy is low. There were
AMR were discussed earlier and include, most specifically, no deaths or graft losses in the Hannover series of more
the presence of C4d in PTC and/or glomeruli. Multilamina- than 1000 biopsies298 and graft loss was 0.04% in another
tion of the GBM or PTC basement membranes is also typical. protocol biopsy series.299
In the absence of C4d in PTC other causes of lamination of The current interest in protocol biopsies started with David
the GBM must be excluded. Demonstration of polyomavi- Rush and colleagues, who made the surprising observation
rus by IHC in previous biopsies can point to a causal role that 30% of biopsies from stable patients 1 to 3 months post-
in the late graft damage, even when the virus is no longer transplant showed histologic rejection300 and those with
detectable.131 these lesions show later loss of renal function.284,301 Many
Obstruction, usually difficult to diagnose by histology, other studies have confirmed this result.229,283,284,292–294
archetypically shows dilated collecting ducts, especially in Mononuclear inflammation that meet the Banff criteria
the outer cortex, lymphatics filled with Tamm-Horsfall pro- for TCMR or borderline acute rejection are found in 5% to
tein, occasionally ruptured tubules with granulomas, and 50% of protocol biopsies in the first 12 months, depending
sometimes acute tubular injury.131 Patients with obstruc- on therapy and patient populations.302 Those with inflam-
tion may show a completely normal histologic appearance mation have a higher risk of graft dysfunction or fibrosis at
on allograft biopsy, however. later time points.229,283,293,294 Grafts with both inflamma-
Renal artery stenosis causes TA (or even acute injury) tion and fibrosis do the worst,229,287,294,303 In one study, the
accompanied by relatively little fibrosis or intraparenchy- best predictor of allograft function 1 year after transplan-
mal arteriolar/arterial lesions.131 tation was persistent inflammation, of any type, including
Recurrent and de novo glomerular diseases are generally those patterns considered in Banff to be irrelevant to the
identified by their light, immunofluorescence, and electron diagnosis of acute rejection (in areas of IF, around large
microscopic criteria in native kidneys.131 blood vessels, in nodules, or in subcapsular areas).304 Infil-
trates in areas of atrophy correlated with IFTA at 6 months
INTERSTITIAL FIBROSIS AND TUBULAR and graft dysfunction at 2 years. In another study, protocol
biopsies at 1 year posttransplant that showed fibrosis and
ATROPHY
inflammation predicted a worse GFR at 5 years compared
There remain cases with IFTA in which no specific diagno- with biopsies with fibrosis and no inflammation and com-
sis can be made. Some of these cases may be the end stage pared with normal biopsies.305 These results and the results
of active processes in which the etiologic agent is no lon- of other studies suggest that these infiltrates are part of the
ger appreciable (e.g., late effects of polyomavirus or TMA). pathogenesis of slow, progressive renal injury.287,288,296
Others may represent burned out or inactive rejection. This What differentiates infiltrates in patients with stable and
might be the case for TG or arteriopathy without C4d depo- unstable graft function? In stable grafts endarteritis is found
sition. Animal studies have shown that limited exposure to rarely (0.3% in one series)306 and can herald an impeding
anti-MHC antibody can cause longstanding arteriopathy, acute episode.300 Among the interstitial infiltrates, only the
despite only transient C4d deposition.228 diffuse pattern (rich in macrophages and granzyme B cyto-
The term “CAN” was created in Banff in 1993 to draw toxic T lymphocytes) was more common in biopsies taken
attention to the fact that not all late graft injury was due for acute dysfunction.304 In contrast, nodular infiltrates
to rejection, and that, to make the diagnosis of rejection, (rich in B cells and activated T cells) were more common
certain more specific features than IF and TA needed to be in protocol biopsies. Similarly, infiltrates rich in activated
25 • Pathology of Kidney Transplantation 397

macrophages distinguished biopsies with clinical versus


subclinical acute rejection.307 Molecular studies have
shown that increased levels of transcripts for T-bet (a Th1
master transcription factor), FasL (cytotoxic mediator), and
CD152 (CTLA-4, an inhibitory costimulatory molecule) are
associated with graft dysfunction.101
Grafts in recipients that are developing tolerance also typ-
ically have graft infiltrates, sometimes termed the “accep-
tance reaction,”308 which spontaneously disappears and is
followed by indefinite graft survival.309,310 The acceptance
reaction had less infiltration by CD3+ T cells and macro-
phages, less T cell activation, long lasting apoptosis of graft
infiltrating T cells, less IFNγ, and more IL-10 than reject-
ing grafts.310,311 Recent evidence shows that regulatory
T cells (Treg) that express the Foxp3 transcription factor
infiltrate tolerated grafts in mice treated with costimulatory
blockade.312 Foxp3 cells can also be found in grafts with
infiltrates interpreted as acute rejection.92 Although the Fig. 25.8 Acute tubular necrosis. Dilated “rigid”-appearing tubular
lumens with loss of brush borders, occasional loss of nuclei, and cyto-
significance of Foxp3+ cells has yet to be determined, high plasmic thinning. Mild edema is present but little inflammation. Glom-
numbers of such Treg cells are likely beneficial,313 in view eruli are normal. PAS stain.
of the known suppressor functions of these cells. The hope
of much ongoing research is the discovery of markers that
predict graft acceptance in a clinical setting.313,314 Acute Tubular Necrosis
Subclinical interaction of antibody with graft endothe-
lium (accommodation) has been revealed by the demon- The morphologic basis of DGF is usually acute ischemic
stration of diffuse C4d in PTCs, found in 2.0% of routine injury (ATN). The most common feature histologically
protocol biopsies,306 and a higher frequency among presen- is loss of the brush borders of proximal tubular cells, best
sitized patients (17%) or patients with ABO-incompatible shown on a PAS stain with focal interstitial edema and
grafts (51%).138,172 The stability of such accommodation, mononuclear cell accumulation (Fig. 25.8). The tubular
referring to the presence of PTC C4d deposition in the lumen appears larger than normal and lacks the usual
absence of other evidence of antibody-mediated injury has artifactual sloughing of the apical cytoplasm in human
not been established. Accommodation is thought to repre- renal biopsies (this sloughing has occurred in vivo and has
sent a process of endothelial cell adaptation to antibody and washed downstream; Fig. 25.9). The other features of ATN
complement over time. In accommodation, donor-specific include flattening of the cytoplasm and loss of cell nuclei
antibodies may be detectable; however, morphologic signs due to apoptosis/death of individual tubular epithelial
of tissue injury are absent. There are no signs of acute or cells and covering of the TBM by the remaining cells. The
chronic TCMR or AMR; more specifically, there is no ATN- lumina contain individual apoptotic detached cells (“anoi-
like minimal inflammation, no glomerulitis [g0], no chronic kis”) and inflammatory cells. Reactive changes in the
TG [cg0], no peritubular capillaritis [ptc0], and no PTC tubular epithelium are seen after 24 to 48 hours, including
basement membrane multilamination. Current Banff crite- large basophilic nuclei with prominent nucleoli, increased
ria refers to this situation as “C4d staining without evidence cytoplasmic basophilia and occasionally mitoses. Focal
of active rejection.”157 If there are simultaneous borderline interstitial, PTC, and glomerular capillary neutrophils may
changes, the cases can be considered indeterminate.200 The be seen but are not as prominent as in acute AMR, and C4d
long-term significance of these relatively uncommon cases is negative. Mechanical flushing of cadaveric kidneys with
is still under investigation.201–203 In nonhuman primates organ preservation fluid immediately before transplanta-
with MHC-incompatible grafts and no immunosuppression, tion (as advocated by some) was associated with abnor-
C4d deposition predicts chronic rejection with glomerulop- mal cellular debris within the tubules and eosinophilic
athy and arteriopathy and ultimate graft loss with a high proteinaceous material within Bowman’s capsule and an
degree of certainty.226 increased frequency of DGF.317 DGF has other causes, and
The most important question is whether treatment of if function has not recovered in 1 to 2 weeks, a diagnostic
subclinical rejection is beneficial (and, if so, what therapy biopsy is recommended to ascertain the presence of occult
is optimal). No study has dared to randomize treatment in acute rejection, found in 18% of patients with DGF at
patients with acute rejection on protocol biopsy. The closest 7 days.318
to a controlled trial was that of Rush and colleagues, who
found that patients with protocol biopsies, treated with
steroid boluses if they had subclinical rejection, had a bet-
Calcineurin Inhibitor
ter outcome than a group of patients who declined a renal Nephrotoxicity
biopsy (and were presumed to have a similar frequency of
subclinical rejection).284 Other diseases revealed by the The CNI class of drugs, including cyclosporine and tacro-
“eye of the needle” clearly benefit from altered therapy, limus, cause both acute and chronic nephrotoxicity that
including CNIT283,315 and polyomavirus infection.316 includes ischemic injury without morphologic features,
398 Kidney Transplantation: Principles and Practice

A B

C D
Fig. 25.9 Polyoma (BK) virus infection. (A) Low power view showing patchy mononuclear inflammation in the medulla with groups of atypical nuclei
in tubular epithelium (arrows) (B) Higher power shows polyomavirus inclusion (arrow), marked tubulitis, and tubular cell apoptosis. (C) Immunohisto-
chemistry, monoclonal antibody to SV40 large T antigen (homologous to BK, JC, and other polyoma viruses), many tubular epithelial cell nuclei appear
dark brown due to immunoreactivity for polyoma virus. (D) EM, high magnification of a tubular cell nucleus (N) containing polyoma virions (arrow), that
are rounded, 30 to 35 nm in diameter and organized in arrays. (From cynomolgus monkey; van Gorder MA, Della Pelle P, Henson JW, Sachs DH, Cosimi AB,
Colvin RB. Cynomolgus polyoma virus infection: a new member of the polyoma virus family causes interstitial nephritis, ureteritis, and enteritis in immunosup-
pressed cynomolgus monkeys. Am J Path 1999;154(4):1273–84.)

vacuolar tubulopathy, acute endothelial injury (TMA), and


arteriolar hyalinosis.290,319 These cause secondary patho-
logic effects, such as TA, IF, and global or segmental glo-
merulosclerosis. As judged by protocol biopsies, chronic
CNIT is universal in renal transplants after about 5 years
according to some studies.283 However, changes attributed
to CNIT have been less in later studies.295 Chronic CNIT can
also damage native kidneys in patients with other organ
transplants and contributes to the 7% to 21% prevalence
of end-stage renal disease in nonrenal transplant recipients
after 5 years.320

ACUTE CALCINEURIN INHIBITOR TOXICITY


Toxic Tubulopathy
The biopsy features of acute toxicity are quite variable. A
normal biopsy is found in “functional CNIT,” which is due
to reversible vasospasm.321 In toxic tubulopathy, proximal Fig. 25.10 Acute calcineurin inhibitor nephrotoxicity with isometric
vacuolization of tubular epithelium. This change can also be seen in
tubules show the most conspicuous morphologic changes other causes of tubular injury, including ischemia, osmotic diuretics,
with loss of brush borders and isometric (uniformly sized), and intravenous immunoglobulin.
clear, fine vacuolization (or microvacuoles) in the epithelial
cells (Fig. 25.10). The microvacuoles contain clear aque-
ous fluid rather than lipid and are indistinguishable from portion. The degree of vacuolization does not correlate
those caused by osmotic diuretics or ischemia. EM shows with drug levels; some patients with CNIT lack the vacu-
that the vacuoles in CsA toxicity are due to dilation of the olar change,323 and isometric vacuoles can be found in a
endoplasmic reticulum and appear empty.322 Isometric minority of patients with stable renal function.324 However,
vacuolization may begin in the straight portion of the prox- reduction of the CNI dosage causes disappearance of tubu-
imal tubule,322 although it can extend to the convoluted lar vacuolization.102
25 • Pathology of Kidney Transplantation 399

A B
Fig. 25.11 TMA associated with calcineurin inhibitors. (A) glomerulus with widespread endothelial swelling, segmental GBM duplication, and focal
collapse resembling a crescent. Arterioles show endothelial swelling and occasional peripheral hyaline nodules. PAS stain. (B) No glomerular or PTC C4d
deposition is detected in this case. Immunohistochemistry for C4d in paraffin, using rabbit polyclonal anti-C4d.

Acute Arteriolar Toxicity and Thrombotic particularly in the hilum,327 the so-called pouch lesion.331
Microangiopathy The endothelial cells are swollen and may completely
Arterioles are a significant target of CNIT. The most char- obliterate the capillary lumina. The GBM is segmentally
acteristic acute changes include individual medial smooth duplicated with cellular (mononuclear or mesangial cell)
muscle cell degeneration, necrosis/apoptosis, and loss.322 interposition best seen by EM, which also shows the loss of
The apoptotic smooth muscle cells are later replaced by fenestrae and swelling of the endothelial cytoplasm. Vari-
rounded, “lumpy” protein deposits or hyalinosis, which is able mesangial expansion, sclerosis, and mesangiolysis331
the beginning of a more chronic arteriolopathy.322 Accu- may be seen. Marked congestion and focal, global, or seg-
mulation of glycogen (PAS positive, diastase sensitive) in mental necrosis can be present.332
smooth muscle cells has been described on high doses.325
Endothelial cells can have prominent vacuolization and Differential Diagnosis
some swelling. Immunofluorescence microscopy of the ves- Acute tubular toxicity of CsA may be indistinguishable
sels often shows deposits of IgM, C3, and sometimes fibrin/ from ischemia or tubulopathy from intravenous immuno-
fibrinogen, but these changes are nonspecific.326 globulin (IVIG) or mannitol, which all have vacuoles by
TMA due to CNI was first reported in bone marrow trans- light microscopy.333 By EM a more coarse and varied vacu-
plant recipients treated with cyclosporine327 and occurs olization is typical of ATN and the periphery of infarcts334
in about 1% to 4% of renal allograft recipients, even with compared with the isometric (uniform) vacuoles of CsA tox-
careful attention to drug levels, suggesting that it is dose icity. The vacuoles of osmotic diuretic injury do not involve
independent and probably idiosyncratic.328,329 Most cases the endoplasmic reticulum, as do those of CsA toxicity.331
present with a delayed onset and a slow loss of function 1 to Necrosis of tubular cells is more common in ATN (0.5% of
5 months posttransplant.330 tubules), characteristically involving whole tubular cross
The pathologic changes are believed to be an exaggera- sections.324 Acute medial apoptosis/degeneration in arteri-
tion of CNI induced endothelial and smooth muscle damage. oles is the only definitive finding favoring CsA toxicity.
The small arteries and arterioles have mucoid intimal thick- Morphology alone cannot distinguish the various eti-
ening with acid mucopolysaccharides and extravasated red ologies of TMA,335,336 which in renal transplants are most
cells and fragments; fibrinoid necrosis and thrombi may commonly CNI, acute AMR, hepatitis C virus (HCV), and
be prominent (Fig. 25.11). Apoptosis of endothelial and recurrent TMA. Recurrence should be the first choice when
smooth muscle cells is seen. The medial smooth muscle can the recipient’s original disease was TMA, unless associated
develop a mucoid appearance with loss of a clear definition with a diarrheal illness. C4d deposition in PTCs is present
of the cells.126 The arterioles may show hypertrophy of the in acute AMR but absent in CNI-associated TMA (see sec-
endothelial cells and have a “constricted” appearance.126 tion on acute AMR). Serum should also be tested for anti-
The vascular lumina may be partially or completely obliter- HLA class I, class II, and antiendothelial antibodies. HCV(+)
ated by the intimal proliferation and endothelial swelling. renal allograft recipients may develop TMA with associated
The vascular lesions are most severe in the interlobular and elevation of circulating anticardiolipin antibody,337 thus
arcuate-sized arteries, and can lead to cortical infarction.330 hepatitis serology and anticardiolipin antibody determina-
By immunofluorescence microscopy, the vessels stain with tion could help distinguish between HCV versus CNI in the
IgM, C3, and fibrin.131 etiology of TMA. The healing phase of TMA may leave inti-
The glomeruli typically have swollen bloodless capillar- mal fibrosis that resembles chronic rejection, even with a
ies with scattered fibrin-platelet thrombi (see Fig. 25.11), few intimal mononuclear cells.131
400 Kidney Transplantation: Principles and Practice

CHRONIC CALCINEURIN INHIBITOR TOXICITY cyclosporine, 15% of protocol biopsies at 6 months showed
CNI-arteriolopathy which increased to 45% in 18-month
Irreversible chronic renal failure due to CNIT was first dem- protocol biopsies349; “nonspecific” hyalinosis showed no
onstrated in native kidneys of heart transplant patients progressive increase. The arteriolar lesions also develop
who received cyclosporine for more than a year.338 Similar in native kidneys of patients who receive even low doses
lesions arise in patients on tacrolimus.339 Biopsies showed of cyclosporine for 2 years.350,351 Mihatsch has suggested
IFTA, arteriolar hyalinosis, and sometimes focal glomeru- a scoring system of CNI-arteriolopathy with improved
lar scarring. These findings have been confirmed and reproducibility.352
extended in numerous other studies.326 More recent stud-
ies of patients mostly on maintenance immunosuppression Glomerular Lesions
with tacrolimus, mycophenolate mofetil, and prednisone After 1 year on cyclosporine, glomeruli show increased
showed less prevalent chronic changes of moderate or numbers with global or segmental sclerosis.341,342 Focal,
severe arteriolar hyalinosis and IFTA than was noted on segmental sclerosis was more common in CNI-treated bone
previous studies of patients on immunosuppressive regi- marrow (13%) and heart transplant (27%) recipients at
mens including cyclosporine.340 Because many features autopsy than their respective CNI-free controls (0% and
resemble chronic rejection in the kidney, the most convinc- 14%).341 Heart transplant recipients have an increase in
ing pathology data come from nonrenal transplant patients the heterogeneity of glomerular volume and size, with more
on cyclosporine.341,342 small and large glomeruli (compensatory hypertrophy),
compared with controls (living kidney donors).343 The shift
CNI-Arteriolopathy to smaller glomeruli becomes more extreme with chronic
The chronic phase of CNI-arteriolopathy is characterized renal failure and the hypertrophied glomeruli disappear.353
by replacement of the degenerated medial smooth muscle Thus hyperfiltration injury probably causes the progressive
cells with hyaline-like deposits, in a beaded pattern along glomerular proteinuria and sclerosis. Bone marrow and
the peripheral outer media (see Fig. 25.7). This has been heart transplant patients at autopsy show glomerular col-
referred to as “nodular protein (hyaline) deposits”289 in lapse in 59% of the patients on CNI versus 8% of those not
a “pearl-like pattern”326 and “peripheral medial nodular on CNI.341 This can develop into florid collapsing glomeru-
hyalinosis,” and is now called “CNI-arteriolopathy.” The lopathy, attributed to the severe CNI-arteriolopathy.354
current evidence supports the view that this type of arte- Immunofluorescence findings are nonspecific (IgM and C3
riolopathy is relatively specific for CNI. In heart and bone in scarred areas). EM in cardiac and liver transplant recipi-
marrow transplant recipient autopsy studies, 55% of cases ents showed diffuse expansion of the mesangial matrix,
on cyclosporine had this type of arteriolopathy in the native with little hypercellularity, GBM, or podocyte lesions.342,343
kidneys compared with 0% in those not on CNIs.341 Evi- Those with frank collapsing glomerulopathy have podo-
dence of apoptosis is sometimes found in the form of kary- cyte foot process effacement and detachment of podocytes
orrhexic debris in the media, but fibrinoid necrosis is not from the GBM.354 The endothelium shows loss of its normal
observed.343 In severe cases the media is nearly devoid of fenestrae, perhaps reflecting a component of TMA.131
smooth muscle cells.343
EM reveals a distinctive replacement of individual smooth Tubules and Interstitium
muscle cells of afferent arterioles with amorphous electron- IFTA was recognized as a feature of CNIT in the early stud-
dense material, which contains cell debris and protrudes ies.355 The interstitium had prominent patchy fibrosis,
into the adventitia (see Fig. 25.7B).326,344,345 This gives with a scanty infiltrate. Band-like narrow zones of IFTA
rise to the beaded hyalinosis distribution in the outer media (“striped fibrosis”) were once regarded as characteristic of
noted by light microscopy. The myocyte nuclei are some- CNIT127,356,357; however, indistinguishable “stripes” occur
times condensed (apoptotic) or have two nuclei or mitotic in patients not maintained on CNI,358 casting doubt on the
figures.344 The cytoplasm is vacuolated, with dilated endo- specificity of that pattern. IF also develops in native kid-
plasmic reticulum, and has degenerated mitochondria, neys in patients on CNI350,359–361 and remains for at least
lipofuscin granules, multivesicular bodies, and a disarray a month after discontinuing the drug.362 Thus even low
of microfibrils and reduced intercellular junctions. The doses of CsA can cause significant and presumably perma-
endothelium sometimes appears “swollen,” protruding into nent loss of renal function by inducing chronic tubulointer-
and narrowing the lumen, and having reduced cell junc- stitial nephritis.131
tions; aggregates of platelets are rare.344,346 These findings
support the view that the smooth muscle myocyte of the Differential Diagnosis
afferent arteriole is a primary target of CNI injury. Immuno- Distinction between chronic rejection and chronic CNIT
fluorescence microscopy shows IgM and C3 in a relatively is a challenge (Table 25.6). The finding that favors CNIT
nonspecific, but conspicuous sheathing of the arterioles.326 most decisively is the arteriolopathy, provided it is distinc-
CNI-arteriolopathy begins and predominates in the affer- tive (isolated smooth muscle cell degeneration and string
ent arterioles but may progress to the small arteries and of pearls replacement by hyalinosis in the outer media).319
efferent arterioles.326,344 Decreased renin immunostaining The arterioles are relatively spared in chronic rejection,
in the juxtaglomerular apparatus suggests that the prime compared with chronic CNIT, and the arteries are more
target of CNI is the renin-producing smooth muscle cell in affected, with proliferative intimal fibrosis without elasto-
the afferent arteriole.347 The frequency of arterioles affected sis.319 PTC C4d deposits or mononuclear cells in the arte-
with hyalinosis is typically small (<15%), and the lesions rial intima are the most useful signs of an active rejection
can easily be overlooked.348 In renal transplant patients on process. An inflammatory infiltrate, including plasma cells,
25 • Pathology of Kidney Transplantation 401

TABLE 25.6 Differentiation Between Chronic Rejection reabsorption. One notable case report described collapsing
and Chronic CNI Toxicity glomerulopathy in a patient with Kaposi’s sarcoma con-
verted to TORi from azathioprine.371 We have seen two
Chronic Rejection CNI Toxicity cases of focal, segmental glomerulosclerosis, in patients
Interstitium started on TORi; one had collapsing glomerulopathy (Cor-
Infiltrate Plasma cells Mild nell et al., unpublished). More pathology studies are clearly
Fibrosis Patchy Patchy, “striped” needed, particularly on those patients started on TORi.131
PERITUBULAR CAPILLARIES
C4d Often positive Negative
BM multilamination Usual Absent Drug-Induced Acute
TUBULES Tubulointerstitial Nephritis
Tubular atrophy Usual Usual
Vacuoles Occasional Occasional Drug-induced interstitial nephritis in the allograft is similar
ARTERIOLES to that in the native kidney and resembles tubulointersti-
Smooth muscle degeneration Absent Usual tial rejection. Both are characterized by an intense mono-
External nodular hyalinosis Absent Present nuclear interstitial infiltrate and tubulitis, and have variable
ARTERIES numbers of eosinophils. Acute rejection occasionally has a
Intimal fibrosis Usual Can be present prominent eosinophilic infiltrate.54,372–376 Conversely, drug-
but unrelated induced interstitial nephritis may have no eosinophils, espe-
Intimal mononuclear cells Common Absent cially those due to nonsteroidal antiinflammatory drugs.377
GLOMERULI Endarteritis, if present, is unequivocal evidence for rejection.
Duplication GBM Usual Absent Strong, but not absolute, evidence for a drug etiology is the
Mesangial expansion Can be present Can be present invasion of multiple tubules by eosinophils, and eosinophils
in tubular casts (Colvin, unpublished observation), usually
BM, basement membrane; CNI, calcineurin inhibitor; GBM, glomerular base- attributed to prophylactic trimethoprim-sulfamethoxazole
ment membrane.
(Bactrim). We have also seen one case of severe acute inter-
stitial nephritis and serum sickness-like syndrome second-
is less common in CNIT than rejection.363 Other features ary to horse anti-thymocyte globulin (ATG).131 Interstitial
are not decisive. IFTA and glomerular sclerosis are found nephritis can sometimes have a granulomatous pattern;
in either. GBM duplication and endothelial dedifferentia- and this pattern can indicate a variety of potential etiologies,
tion can also be seen in either, although perhaps more com- including drugs, infections, and recurrent diseases such as
monly in chronic rejection.131 sarcoidosis and granulomatosis with polyangiitis.378

Target of Rapamycin Inhibitor Infections


Toxicity
Many organisms can infect the transplanted kidney, rang-
Inhibitors of the mammalian target of rapamycin (TORi; ing from mycobacteria and Candida,379 to herpes simplex
rapamycin, everolimus, sirolimus) can cause DGF due to virus380 and human herpesvirus 1 (HHV-1, also known
tubular toxicity that resembles myeloma cast nephropathy. as Herpes simplex virus-1 [HSV-1]).380 In addition, viruses
Pathologically, in addition to acute tubular injury, eosin- such as CMV and HCV can have indirect effects on the
ophilic debris and macrophages were present in tubular transplant, promoting rejection or immune-mediated dis-
lumina, which mimicked myeloma casts, but the casts stain ease.91,116,381 CMV can also directly infect the allograft,
for keratin, rather than immunoglobulin light chains.364 particularly in glomerular endothelial cells. Here we will
TORi can also cause TMA, indistinguishable from that due discuss the three most important types of infections, poly-
to CNI.365 omavirus, adenovirus, and bacterial pyelonephritis.
Increased proteinuria is common in patients switched from
CNI to TORi because they had developed severe CNIT. In these POLYOMAVIRUS TUBULOINTERSTITIAL
patients, GFR improves but increased proteinuria develops in
NEPHRITIS
about 30%.366 CNI exposure is not necessary for the protein-
uric response to TORi. Conversion from azathioprine to TORi PTN has emerged since 1996 as a significant cause of early
can also cause increased proteinuria.367 Patients started on and late graft damage.382–387 Among various series of
TORi without CNI had double the risk of proteinuria at 6 to patients on tacrolimus/mycophenolate mofetil, PTN arises
12 months compared with those on CNI.368 in about 5%, similar to the prevalence of acute rejection.
Few pathologic studies have been published. One The virus was originally isolated from B.K., a Sudanese
reported a variety of glomerular diseases typical of native patient who had distal donor ureteral stenosis 3 months
kidneys, suggesting recurrent disease.369 A recipient begun after a living related transplant.388 BK virus is related to JC
on TORi, developed 12 g/day proteinuria in the first week virus (which also inhabits the human urinary tract) and to
after transplantation, which remitted after the drug was simian virus SV40. These viruses are members of the papo-
discontinued.370 Biopsy showed that no obvious glomeru- vavirus group, which includes the papilloma viruses. The
lar disease was evident by light, immunofluorescence, or BK virus commonly infects urothelium but rarely causes
EM, suggesting the proteinuria was due to failure of tubular morbidity in immunocompetent individuals. However, in
402 Kidney Transplantation: Principles and Practice

A B
Fig. 25.12 Recurrent diabetic nephropathy 12 years after transplant. (A) Glomerulus with prominent Kimmelstiel-Wilson mesangial nodules (arrow)
and arteriolar hyalinosis. PAS stain. (B) EM of another case shows homogeneous thickening of the GBM up to 1100 nm. C, capillary lumen; U, urinary space.

renal transplant recipients three lesions have been attrib- demonstrable. The virus is cytopathic for tubular cells and
uted to BK virus: hemorrhagic cystitis, ureteral stenosis, leads to characteristically destructive tubular lesions, with
and interstitial nephritis.389–391 only TBM remaining. The diagnosis is sometimes only pos-
PTN is characterized by a patchy mononuclear infiltrate sible by review of prior biopsies. Suspicion of PTN is height-
associated with tubulitis and tubular cell injury.387 The ened if tubular destruction is severe. The process may be
infiltrate often contains plasma cells, which sometimes clinically silent: protocol biopsies have shown a subclinical
invade the tubules (see Fig. 25.9). Concurrent TCMR may incidence of PTN of 1.2%.316 Furthermore, PTN can affect
be present. Tubular cell apoptosis is prominent as well as native kidneys of recipients of nonrenal allografts; only a
“dedifferentiation” of tubular epithelial cells, with loss of few cases have been reported, but this may be in part due
polarity and a spindly shape. PTN has three recognized to a presumption of CNIT and a lack of renal biopsies in this
stages: stage A has only minimal inflammation; stage B setting.398
shows marked tubular injury, denudation of the tubular
basement membranes, and interstitial edema with a mixed, ADENOVIRUS
mild to marked inflammatory cell infiltrate; and stage C has
marked IFTA.382,392–395 Adenovirus, most frequently serotype 11, causes hemor-
The recognition of viral nuclear inclusions is the key rhagic cystitis and occasionally tubulointerstitial nephritis
step in histologic diagnosis. The affected nuclei are usu- in renal allografts, which may resemble a space-occupying
ally enlarged with a smudgy, amorphous lavender inclu- lesion by imaging studies.399,400 Biopsy shows necrotizing
sion (see Fig. 25.9B). Other nuclear changes found less inflammation with neutrophils and tubular destruction,
commonly are eosinophilic, granular inclusions with interstitial hemorrhage and red cell casts, granulomatous
or without a halo and a vesicular variant with coarsely inflammation,401–404 or a zonal inflammation localized
clumped, irregular basophilic material.78,385,396 These to the outer medulla.405 Tubular cells have intranuclear
nuclear inclusions tend to be grouped in tubules, particu- ground glass inclusions with a distinct halo surrounded
larly collecting ducts in the cortex and outer medulla, and by a ring of marginated chromatin and glassy smudged
can often be spotted at low power. IHC and EM confirm the nuclei. The diagnosis is established by immunoperoxidase
diagnosis. Monoclonal antibodies are commercially avail- stains for viral antigen in tubular cells and EM to reveal
able that react with BK-specific determinants and with the the intranuclear crystalline arrays of 75- to 80-nm viral
large T antigen of several polyoma species (see Fig. 25.9C). particles. Immune complexes may also contribute to the
In situ hybridization for BK polyomavirus is used at some injury. Decreased immunosuppression has been followed
centers as an alternative to IHC. EM will reveal the char- by recovery.131
acteristic intranuclear paracrystalline arrays of viral par-
ticles of about 40 nm diameter (Fig. 25.12D). Other tests ACUTE PYELONEPHRITIS
useful for monitoring patients at risk are urine cytology
(“decoy cells”) and PCR quantitation of virus in the blood, Pyelonephritis is a potentially devastating complication
although these are not specific enough to make a PTN of transplantation. Pyelonephritis can present as acute
diagnosis.131 renal failure406,407 and cause graft loss.408,409 According
Polyomavirus infections may cause an immune complex to one series, pyelonephritis arises most often 1 year or
deposition along the TBM, as described in 43% of cases in more after transplantation (80% of episodes), and E. coli
a series from Seattle, being the most common cause of IgG was the most common organism (80%).410 Acute pyelo-
deposits in the TBM of transplants.397 Granular IgG, C3, nephritis is a not an uncommon finding on renal biopsy,
and C4d are focally present by immunofluorescence and despite the expectation that the process is patchy.406
amorphous electron-dense deposits by EM. The prognostic Renal biopsies are not the usual method of diagnosis,
significance is not known.131 however, if neutrophils are abundant, especially if they
Late graft fibrosis and scarring “CAN” may be caused form destructive abscesses and casts in tubules, the diag-
by polyomavirus, even though the virus is no longer nosis should be at the top of the list. Other variants are
25 • Pathology of Kidney Transplantation 403

emphysematous pyelonephritis, due to gas-producing


organisms,409 xanthogranulomatous pyelonephri-
tis,411,412 and malakoplakia.413

Major Renal Vascular Disease


Most arterial thromboses develop in the early posttransplant
period and produce acute infarction with microthrombi
and scant inflammation.414 Evidence for underlying rejec-
tion should be sought by careful examination of the larger
arteries for endarteritis. Renal artery stenosis (typically at
the anastomosis site), a cause of late graft dysfunction, can
be deceptive clinically and pathologically.415,416 Biopsies
show tubular injury or atrophy with relatively little inflam-
mation or fibrosis.
Renal vein thrombosis causes a swollen and purple kid-
ney, sometimes with graft rupture.417 The cortex shows Fig. 25.13 De novo membranous glomerulonephritis: subepithelial
severe hemorrhagic congestion and extensive infarction electron-dense deposits (arrows) along the GBM with intervening
basement membrane spikes. Podocyte (P) foot processes are effaced.
and necrosis418 sometimes with diffuse microcapillary C, capillary lumen; U, urinary space.
thrombi. Intracapillary leukocytes can be a clue as in native
kidneys. Late renal vein thrombosis is associated with pro-
teinuria due to membranous glomerulonephritis or TG, The pathogenesis of de novo MGN has not been established.
sometimes with graft loss.419 Lupus anticoagulant has been The literature supports the hypothesis that de novo MGN
detected in a few patients.420 may be a form of AMR or directed at minor histocompatibil-
ity antigen(s) in the glomerulus, presumably on the podo-
cyte or a special type of chronic rejection.234,427,429 The
De Novo Glomerular Disease common presence of TG is consistent with this hypothe-
sis.425,427 Phospholipase A2 receptor (PLA2R) staining can
Patients without previous glomerular disease occasionally be useful in the evaluation of allograft MGN. In one study,
develop lesions in the allograft that resemble a primary glo- PLA2R staining had a sensitivity of 83% and specificity of
merular disease, rather than the usual transplant glomeru- 92% (for recurrent MG; PLA2R staining was almost always
lopathy. Although some are no doubt coincidental, at least negative in de novo MGN.430
three are related to an alloimmune response to the allograft:
membranous glomerulonephritis, anti-GBM disease in ANTI-GBM NEPHRITIS
Alport’s syndrome, and recurrent nephrotic syndrome in
congenital nephrosis. A fourth relatively common de novo Patients with Alport’s syndrome or hereditary nephritis
glomerular disease, FSGS, is believed to be related to hyper- commonly develop anti-GBM alloantibodies, because they
filtration injury of the allograft or marked microvascular genetically lack self-tolerance to GBM collagen components.
compromise due to CNIT.131 However, this leads to glomerulonephritis in only a minor-
ity. Overall, de novo crescentic and necrotizing glomerulo-
nephritis due to anti-GBM antibodies after transplantation
MEMBRANOUS GLOMERULONEPHRITIS
is uncommon, seen in only 5% of male adult renal allograft
De novo MGN is typically a late complication, with a prev- recipients with typical Alport’s syndrome431,432 or hereditary
alence of about 1% to 2%.421–423 In contrast, recurrent nephritis with deafness.433 The pathology is similar to that
MGN can present early.424 The risk factors for de novo in native kidney with prominent crescents (not a feature of
MGN include time after transplant, de novo MGN in a first allograft rejection), segmental necrosis, and red cell casts. Sec-
graft,421 and HCV infection.422,423 Light microscopy usu- ond transplantation with and without recurrent anti-GBM
ally shows rather mild GBM changes. Mesangial hyper- nephritis have both been reported.434–436 The 5-year graft
cellularity is found in about 33%. Mononuclear cells can survival may be equal to that of non-Alport’s recipients.437
be abundant in glomerular capillaries, raising the possibil-
ity of transplant glomerulitis or renal vein thrombosis.425 DE NOVO PODOCYTOPATHY IN CONGENITAL
Immunofluorescence shows granular deposits along the NEPHROSIS
GBM that stain for IgG, C3, C4d, and factor H426; about
35% are more irregular and segmental in distribution Congenital nephrotic syndrome of the Finnish type, an
than typical primary (idiopathic) MGN.425,427 By EM, sub- autosomal recessive disease due to mutations in the neph-
epithelial electron-dense deposits are present (Fig. 25.13), rin gene NPHS1, paradoxically leads to posttransplant
which are smaller and more irregular in distribution than nephrotic syndrome.438,439 The podocyte pathology resem-
primary MGN.425,427 Endothelial changes and GBM dupli- bles minimal change disease and usually responds to cyclo-
cation typical of TG are present in half of the cases.425,427 phosphamide.440,441 De novo “minimal change disease” is
Repeat biopsies have shown persistence or progression of thought to be caused by the alloantibodies to nephrin in
the deposits in most cases and occasional resolution.425,428 some cases.442
404 Kidney Transplantation: Principles and Practice

TABLE 25.7 Classification of Recurrent Renal Disease


I. USUALLY RECUR (>50% PATIENTS)
A. Adverse effecta Primary HUS
Dense deposit disease
Collapsing FSGSc
B. Little or no adverse Immunotactoid/fibrillary glomerulopathyc
affect Systemic light chain diseasec
Diabetes mellitusd
II. COMMONLY RECUR (5%–50%)
A. Adverse effect Focal, segmental glomerulosclerosis
Membranoproliferative GN,b type I
Membranous GN
ANCA related diseases
Wegener’s granulomatosis
Pauci-immune GN
Microscopic polyarteritis
Progressive systemic sclerosis
Sickle cell nephropathyc
Fig. 25.14 De novo collapsing glomerulopathy: collapsed glomeru- B. Little or no adverse IgA nephropathy
lar capillaries and prominent podocyte proliferation, hypertrophy, affect Henoch-Schönlein purpura
and abundant reabsorption droplets. Severe arteriolar hyalinosis with Amyloidosis
peripheral nodules typical of CNI arteriolopathy was present. This is a III. RARELY RECUR (<5%)
native kidney in a patient with a heart-lung transplant.354 PAS stain.
A. Adverse effect Anti-GBM disease
B. Little or no adverse Systemic lupus erythematosus
FOCAL SEGMENTAL GLOMERULOSCLEROSIS affect Fabry’s disease
Cystinosis
De novo FSGS has been described in adult recipients of IV. NEVER RECUR (ESSENTIALLY 0%)
pediatric kidneys,443,444 in which the presumed patho- A. Unique complications Hereditary nephritis/Alport’s syndrome
genesis is hyperfiltration injury; in long-standing grafts, (anti-GBM disease)
in which parenchymal loss due to CNIT or chronic rejec- Congenital nephrosis (nephrotic syn-
tion leads to hyperfiltration injury of residual glomeruli; drome; nephrin autoantibody?)
B. No unique complica- Polycystic disease (all genetic types)
and as the collapsing variant of FSGS, probably related to tions Osteo-onychodysplasia (nail-patella)c
CNI-arteriolopathy.445 Acquired cystic disease
De novo collapsing glomerulopathy presents months Secondary HUS (infection)
to years after transplantation with proteinuria (2–12 g/ Secondary FGS
day).445–447 Diffuse or focal, global or segmental collapse Familial FGSc
Postinfectious acute glomerulonephritisc
of glomeruli was evident with prominent hyperreactive V. Unclassified, recurrence Thrombotic thrombocytopenic purpura
podocytes (Fig. 25.14). Arteriolar hyalinosis, arteriosclero- reportede Adenosine phosphoribosyl transferase
sis, and IF were also present. A rapid progression to renal deficiency
failure occurred in 80% of the patients (2–12 months). The Familial fibronectin glomerulopathy
Lipoprotein glomerulopathy
cause is unknown; all patients were HIV negative. Collaps- Malacoplakia
ing glomerulopathy can also develop in native kidneys in
patients on CNI (see Fig. 25.14).354 aAdverse effect defined as graft loss of >5% (when disease recurs)
bANCA, antineutrophil cytoplasmic antibody; FSGS, focal segmental glomeru-
losclerosis; GBM, glomerular basement membrane; GN, glomerulonephri-
Recurrent Renal Disease tis; HUS, hemolytic-uremic syndrome.
cLimited experience: few cases reported (n < 10).
dArteriolar and glomerular lesions will recur to some degree in most if not all

Recurrent disease is a significant cause of allograft fail- cases, but severe form (nodular) delayed until >5 years.
eRecurrence occurs, but too few cases reported to classify frequency or
ure.448–450 The frequency and clinical significance of recur-
consequences.
rence varies with the disease (Table 25.7). In one study,
glomerular diseases, including recurrent and de novo glo-
merulonephritis and transplant glomerulopathy, were the reversibility of preexisting lesions in the donor kidney
responsible for 37% of cases of graft loss, and 14% of death- (e.g., diabetes, IgA nephropathy). For example, in early
censored graft losses were due to recurrent glomerular dis- recurrent MGN, as early as 2 weeks posttransplant the
ease297 (e.g., recurrent dense deposit disease in Fig. 25.15). glomeruli can show staining in a membranous pattern by
Recurrence of immune-mediated disease may become a immunofluorescence for IgG, C4d, and kappa and lambda
greater problem in the future with longer graft survival and light chains, but corresponding electron-dense deposits
development of tolerance protocols that require no immu- may not be present ultrastructurally; these features can be
nosuppression. The reader is referred to a comprehensive seen on biopsy without proteinuria clinically.424 Later biop-
review elsewhere for detailed information regarding specific sies of the allografts show a more typical membranous pat-
diseases.9 tern with subepithelial deposits by EM.451
Transplantation also can uniquely illuminate the early Diabetic nephropathy begins with an increase in allograft
pathologic events that precede clinical signs and determine glomerular volume at 6 months,452 followed by increases
25 • Pathology of Kidney Transplantation 405

A B
Fig. 25.15 Recurrent dense deposit disease. (A) EM, widespread very electron-dense deposits that are continuous, linear, and embedded in the GBM
proper, (i.e., intramembranous [arrows]). Similar deposits are also seen in the mesangium (M). C, capillary lumen; U, urinary space. (B) immunofluores-
cence microscopy, staining for C3 shows broad, linear ribbon-like deposits along the GBM and blob-like deposits in the mesangium (mesangial rings).

in mesangial volume453 and mesangial sclerosis, which is immunosuppression are also at increased risk for malig-
present 10 years after transplantation in the majority of nancy,462 likely in part due to various functional immune
patients who had diabetes at baseline, as was nicely dem- system abnormalities related to uremia and dialysis. Those
onstrated in one study.454 Thickening of the GBM is first cancers increased in kidney transplant recipients compared
evident after 2 to 3 years453,455 and nodular diabetic glo- with patients on the transplant waiting list include Kaposi’s
merulosclerosis at 5 to 15 years posttransplant (see Fig. sarcoma (associated with human herpesvirus-8 [HHV-8]),
25.12).456 PTLD (some associated with Epstein-Barr virus [EBV]), gen-
Tubulointerstitial diseases may also recur, such as with itourinary cancers in women (some associated with human
recurrent oxalate nephropathy in primary hyperoxal- papillomavirus), nonmelanoma skin cancers, melanoma,
uria.457 A chronic tubulointerstitial nephritis mediated by and mouth and esophageal cancers, among others.460,461
antibodies against the proximal tubule brush border can The renal pathologist must be particularly aware of PTLD,
recur after transplantation. Patients present with renal as it can affect the graft and cause graft dysfunction.
failure ranging from slowly progressive to acute with little PTLD refers to a spectrum of lymphocytic or plasmacytic
or no proteinuria and a bland urinary sediment. Tubules proliferations, ranging from EBV-positive polyclonal prolif-
appear injured, and focal tubulitis may be present amid an erations of plasmacytic hyperplasia and infectious mononu-
infiltrate of lymphocytes, plasma cells, and few eosinophils. cleosis-like PTLD to EBV-positive or -negative monoclonal
On PAS stains, there is a diffuse loss in proximal tubule lymphomas.463 Early and late-occurring PTLDs are rela-
brush borders. Immunofluorescence shows widespread tively distinct clinicopathologic entities, with late-occurring
granular deposits along the TBM that stain for IgG and C3, PTLD resembling lymphoproliferative disorders in immuno-
and prominent amorphous electron-dense deposits are seen competent patients.464,465 PTLDs that arise relatively early
in the TBM on EM.458 Recent data indicate that LDL recep- posttransplant (<1 year) tend to be associated with EBV466
tor-related protein 2 (LRP2), also known as megalin, is the and are composed of lymphocytes of donor origin.467 In
target in this anti-brush border antibody disease (ABBA one study, most donor-origin PTLDs developed in the kid-
disease).459 ney allograft; 79% of these were EBV-positive.467 PTLDs
that arise late posttransplant are less frequently associated
with EBV, more likely to be of recipient origin,467 and have
Posttransplant Malignancy and a poorer prognosis.464,466
PTLD occurs in approximately 0.3% of kidney transplant
Posttransplant recipients by 1 year posttransplant and 1.6% by 10 years.468
Lymphoproliferative Disease Risk factors for development of PTLD in renal transplant
patients include recipient EBV seronegativity with an EBV
Immunosuppression leads to an increased risk of malig- seropositive donor,469 recipient age <18 years470 (due to
nancy, particularly those neoplasms caused by viruses and higher rate of EBV seronegativity in children), age over 60
ultraviolet radiation. These malignancies are presumptively years at the time of transplantation in adult patients,471
suppressed by immune responses that recognize the viral or greater degree of immunosuppression, antirejection therapy
mutation-derived neoantigens. Immunosuppressive agents with OKT3 or antithymocyte globulin,468 use of belatacept
may interfere with normal immune surveillance and mech- (T cell costimulation blocker) for immunosuppression,472,473
anisms of DNA repair, and some common posttransplant and viral coinfection with HCV or CMV.
viral infections are associated with malignancy.460 There The morphologic appearance of PTLD can vary (e.g.,
is an approximately twofold increased risk of common can- Fig. 25.16). Most (86%–92%) are B cell predominant; 68%
cers (colon, lung, prostate, and breast) in kidney transplant to 81% are EBV-positive.471,474 PTLD involves the renal
patients in the first 3 years posttransplant compared with allograft in at least 21% of cases (with or without extrare-
the general population,461 so transplant patients should be nal involvement),474 with one study showing graft involve-
part of appropriate cancer screening protocols. End-stage ment in 49% of PTLD cases in renal transplant recipients.467
renal disease patients and hemodialysis patients not on Most PTLDs involving the allograft have a polymorphous
406 Kidney Transplantation: Principles and Practice

A B
Fig. 25.16 Posttransplant lymphoproliferative disease. (A) Dense mononuclear cell infiltrate in the interstitium that permeates between the tubules
without tubulitis (although tubulitis may occur in PTLD). The monomorphic infiltrate and the lack of edema distinguish PTLD from the usual cellular
rejection. (B) In situ hybridization, nuclei of mononuclear cells stain dark, brown-black for EBER (Epstein-Barr virus encoded RNA), which is the definitive
test for the diagnosis of PTLD.

appearance.463 The kidney is infiltrated with cells that typically high-grade urothelial carcinomas481; however,
include small lymphocytes, plasma cells, and varying they may exhibit unique morphologies such as micropapil-
numbers of immunoblasts with large nuclei and promi- lary479 and giant cell components.478 Collecting duct carci-
nent nucleoli. The plasma cells usually (but not always) nomas have also been described.482,483
have a monotypic staining pattern for kappa or lambda
light chain. Rarely, other types of PTLD occur in the kid-
ney, including pure plasmacytic lesions, T cell lymphomas, Future Directions in Biopsy
and NK cell lymphomas.463 If needed, consultation with a Assessment
hematopathologist can help the renal pathologist make the
diagnosis of a PTLD. Biopsy assessment will likely further improve from advances
PTLD involving the kidney can resemble TCMR in having in image analysis techniques and molecular understanding.
a widespread mononuclear infiltrate invading tubules and The contribution from a variety of “-omics” fields and tech-
even vessels.60,475,476 In TCMR, the infiltrating cells are not nologies has led to improvements in allograft biopsy assess-
positive for EBV, a feature that helps distinguish TCMR and ment.484,485 Molecular phenotypes have been characterized
PTLD in the early posttransplant period. EBV is best dem- for a variety of pathologic states in renal allograft biopsies;
onstrated by in situ hybridization for EBER (EBV encoded however, the clinical utility of these molecular phenotypes
RNA; see Fig. 25.16). In some cases, a useful clue that will need additional validation before it is understood the
favors PTLD is when the infiltrate forms a dense sheet of circumstances in which molecular assessment can be supe-
monomorphic, large lymphoid cells with vesicular chroma- rior to histopathology. In addition, before molecular biopsy
tin and prominent nucleoli, without accompanying edema assessment is clinically feasible as an adjunct to histopa-
or granulocytes (see Fig. 25.16). Serpiginous necrosis of the thology, additional improvements are needed in molecular
lymphoid cells (irregular patches) has been described, but is method turnaround time, cost, and the reporting required
not always present in PTLD.476 for high-dimensional “-omics” data.
Carcinoma of the urinary tract has been associated with Digital microscopic techniques (e.g., whole slide scan-
polyomavirus infection.477–481 In the past, the possibility ning) are also emerging that will likely improve biopsy
that polyomavirus could contribute to tumorigenesis was assessment. Whole histology slide images contain highly
not widely considered; however, in transplant recipients detailed image information, allowing data mining through
with a history of polyomavirus-associated nephropathy, computer-based image analysis techniques. For example,
expression of the SV40 polyomavirus large T antigen has interstitial fibrosis assessment can be automated; and auto-
recently been demonstrated in numerous renourinary mation can likely make interstitial fibrosis assessment more
tumors (e.g., transplant kidney, native kidney, renal pel- reproducible.486–488 Algorithms are available for additional
vicalyceal system, graft ureter, bladder, and metastases features such as inflammatory cell infiltration, microvessel
from some of these tumors). In polyomavirus infection, density, and a variety of other parameters.489,490 Multipa-
polyomavirus early and late proteins (large tumor antigen rameter staining techniques can be coupled with digital
[LTag] and viral capsid protein 1 [VP1], respectively) are imaging and analysis algorithms to provide more objective
expressed; and LTag inactivates the p53 tumor suppres- and quantitative assessment of molecular derangements
sor gene, leading to positive P53 staining. As cells become in the renal biopsies.489 Advancements in technology such
dysplastic and frankly malignant, cells become positive for as artificial intelligence/machine learning and pathologic
Ki67, indicating proliferative activity, and both P53 and understanding will likely provide a more complete picture
P16, indicating tumor suppressor gene inactivation and and allow enhanced patient care.
cell cycle regulation loss. Later in the process, VP1 staining
can become negative due to a process referred to as early/ Acknowledgments
late viral gene region (EVGR/LVGR) uncoupling. The reti- Many thanks to a coauthor of a prior version, Shamila
noblastoma tumor suppressor protein (pRB) is also impli- Mauiyyedi, MD, and to Dr. Paul J. Kurtin, for his useful sug-
cated in the pathogenesis of these tumors. The tumors are gestions on the manuscript.
25 • Pathology of Kidney Transplantation 407

References 25. Pokorna E, Vitko S, Chadimova M, Schuck O, Ekberg H. Proportion


of glomerulosclerosis in procurement wedge renal biopsy cannot
1. Kiss D, Landman J, Mihatsch M, Huser B, Brunner F, Thiel G. Risks alone discriminate for acceptance of marginal donors. Transplanta-
and benefits of graft biopsy in renal transplantation under cyclospo- tion 2000;69(1):36–43.
rin-A. Clin Nephrol 1992;38(3):132–4. 26. Edwards EB, Posner MP, Maluf DG, Kauffman HM. Reasons for
2. Kon SP, Templar J, Dodd SM, Rudge CJ, Raftery MJ. Diagnostic con- non-use of recovered kidneys: the effect of donor glomeruloscle-
tribution of renal allograft biopsies at various intervals after trans- rosis and creatinine clearance on graft survival. Transplantation
plantation. Transplantation 1997;63(4):547–50. 2004;77(9):1411–5.
3. Pascual M, Vallhonrat H, Cosimi AB, et al. The clinical usefulness 27. Wang HJ, Kjellstrand CM, Cockfield SM, Solez K. On the influence of
of the renal allograft biopsy in the cyclosporine era: a prospective sample size on the prognostic accuracy and reproducibility of renal
study. Transplantation 1999;67(5):737–41. transplant biopsy. Neph Dial Transplant 1998;13(1):165–72.
4. Williams WW, Taheri D, Tolkoff-Rubin N, Colvin RB. Clinical role of 28. Furness PN, Taub N, Assmann KJ, et al. International variation in
the renal transplant biopsy. Nat Rev Nephrol 2012;8(2):110–21. histologic grading is large, and persistent feedback does not improve
5. Halloran PF, Famulski KS, Reeve J. Molecular assessment of dis- reproducibility. Am J Path Surg Path 2003;27(6):805–10.
ease states in kidney transplant biopsy samples. Nat Rev Nephrol 29. Remuzzi G, Cravedi P, Perna A, et al. Long-term outcome of renal
2016;12(9):534–48. transplantation from older donors. N Engl J Med 2006;354(4):
6. Reeve J, Bohmig GA, Eskandary F, et al. Assessing rejection-related 343–52.
disease in kidney transplant biopsies based on archetypal analysis of 30. Karpinski J, Lajoie G, Cattran D, et al. Outcome of kidney transplan-
molecular phenotypes. JCI Insight 2017;2(12). tation from high-risk donors is determined by both structure and
7. Adam B, Mengel M. Transplant biopsy beyond light microscopy. function. Transplantation 1999;67(8):1162–7.
BMC Nephrol 2015;16:132:1–7. 31. Taub HC, Greenstein SM, Lerner SE, Schechner R, Tellis VA. Reas-
8. Adam B, Mengel M. Molecular nephropathology: ready for prime sessment of the value of post-vascularization biopsy performed
time? Am J Physiol Renal Physiol 2015;309(3):F185–8. at renal transplantation: the effects of arteriosclerosis. J Urol
9. Nickeleit V, Mengel M, Colvin RB. Renal transplant pathology. In: 1994;151(3):575–7.
Jennette JC, Olson JL, Silva FG, D’Agati VD, editors. Heptinstall’s 32. McCall SJ, Tuttle-Newhall JE, Howell DN, Fields TA. Prognostic
Pathology of the Kidney. 7th ed. Philadelphia, PA: Wolters Kluwer; significance of microvascular thrombosis in donor kidney allograft
2015. p. 1321–459. biopsies. Transplantation 2003;75(11):1847–52.
10. Solez K, Axelsen RA, Benediktsson H, et al. International stan- 33. Soares KC, Arend LJ, Lonze BE, et al. Successful renal transplanta-
dardization of criteria for the histologic diagnosis of renal allograft tion of deceased donor kidneys with 100% glomerular fibrin thrombi
rejection: the Banff working classification of kidney transplant and acute renal failure due to disseminated intravascular coagula-
pathology. Kidney Int 1993;44(2):411–22. tion. Transplantation 2017;101(6):1134–8.
11. Colvin RB, Cohen AH, Saiontz C, et al. Evaluation of pathologic crite- 34. Sood P, Randhawa PS, Mehta R, Hariharan S, Tevar AD. Donor
ria for acute renal allograft rejection: reproducibility, sensitivity, and kidney microthrombi and outcomes of kidney transplant: a single-
clinical correlation. J Am Soc Nephrol 1997;8(12):1930–41. center experience. Clin Transplant 2015;29(5):434–8.
12. Wang H, Nanra RS, Carney SL, et al. The renal medulla in acute 35. Batra RK, Heilman RL, Smith ML, et al. Rapid resolution of donor-
renal allograft rejection: comparison with renal cortex. Neph Dial derived glomerular fibrin thrombi after deceased donor kidney trans-
Transplant 1995;10(8):1428–31. plantation. Am J Transplant 2016;16(3):1015–20.
13. Cohen AH, Gonzalez S, Nast CC, Wilkinson A, Danovitch GM. Fro- 36. Abouna GM, Al Adnani MS, Kremer GD, Kumar SA, Daddah SK,
zen-section analysis of allograft renal biopsy specimens. Reliable Kusma G. Reversal of diabetic nephropathy in human cadaveric
histopathologic data for rapid decision making. A Path Lab Med kidneys after transplantation into non-diabetic recipients. Lancet
1991;115(4):386–9. 1983;2(1274):1274–6.
14. Collins AB, Schneeberger EE, Pascual MA, et al. Complement acti- 37. Ji S, Liu M, Chen J, et al. The fate of glomerular mesangial IgA depo-
vation in acute humoral renal allograft rejection: diagnostic signifi- sition in the donated kidney after allograft transplantation. Clin
cance of C4d deposits in peritubular capillaries. J Am Soc Nephrol Transplant 2004;18(5):536–40.
1999;10(10):2208–14. 38. Nakazawa K, Shimojo H, Komiyama Y, et al. Preexisting membra-
15. Ivanyi B, Kemeny E, Szederkenyi E, Marofka F, Szenohradszky P. nous nephropathy in allograft kidney. Nephron 1999;81(1):76–80.
The value of electron microscopy in the diagnosis of chronic renal 39. Lipkowitz GS, Madden RL, Kurbanov A, et al. Transplantation and
allograft rejection. Mod Path 2001;14(12):1200–8. 2-year follow-up of kidneys procured from a cadaver donor with a
16. Collins AB, Chicano SL, Cornell LD, et al. Putative antibody-medi- history of lupus nephritis. Transplantation 2000;69(6):1221–4.
ated rejection with C4d deposition in HLA-identical, ABO-compati- 40. Brunt EM, Kissane JM, Cole BR, Hanto DW. Transmission and resolu-
ble renal allografts. Transplant Proc 2006;38(10):3427–9. tion of type I membranoproliferative glomerulonephritis in recipients
17. Mengel M, Sis B, Haas M, et al. Banff 2011 Meeting report: of cadaveric renal allografts. Transplantation 1988;46(4):595–8.
new concepts in antibody-mediated rejection. Am J Transplant 41. Williams GM, Hume DM, Huson Jr RP, Morris PJ, Kano K, Milgrom
2012;12(3):563–70. F. “Hyperacute” renal-homograft rejection in man. N Eng J Med
18. Nyberg G, Hedman L, Blohme I, Svalander C. Morphologic findings 1968;279(611):611–5.
in baseline kidney biopsies from living related donors. Transplant 42. Kissmeyer-Nielsen F, Olsen S, Petersen VP, Fjeldborg O. Hyperacute
Proc 1992;24(1):355–6. rejection of kidney allografts, associated with pre-existing humoral
19. Cahen R, Dijoud F, Couchoud C, et al. Evaluation of renal grafts by antibodies against donor cells. Lancet 1966;2(7465):662–5.
pretransplant biopsy. Transplant Proc 1995;27:2470. 43. Gaber LW, Gaber AO, Vera SR, Braxton F, Hathaway D. Successful
20. Liapis H, Gaut JP, Klein C, et al. Banff histopathological consen- reversal of hyperacute renal allograft rejection with the anti-CD3
sus criteria for preimplantation kidney biopsies. Am J Transplant monoclonal OKT3. Transplantation 1992;54(5):930–2.
2017;17(1):140–50. 44. Halloran PF, Wadgymar A, Ritchie S, Falk J, Solez K, Srinivasa NS.
21. Mohan S, Campenot E, Chiles MC, et al. Association between reperfu- The significance of the anti-class I antibody response. I. Clinical and
sion renal allograft biopsy findings and transplant outcomes. J Am pathologic features of anti-class I-mediated rejection. Transplanta-
Soc Nephrol 2017;28(10):3109–17. Available online at: https:// tion 1990;49(1):85–91.
doi.org/10.1681/ASN.2016121330. 45. Ahern AT, Artruc SB, DellaPelle P, et al. Hyperacute rejection of
22. Gaber LW, Moore LW, Alloway RR, Amiri MH, Vera SR, Gaber AO. HLA-AB-identical renal allografts associated with B lymphocyte
Glomerulosclerosis as a determinant of posttransplant function of and endothelial reactive antibodies. Transplantation 1982;33(1):
older donor renal allografts. Transplantation 1995;60(4):334–9. 103–6.
23. Randhawa PS, Minervini MI, Lombardero M, et al. Biopsy of mar- 46. Paul L, Class F, van Es L, Kalff M, de Graeff J. Accelerated rejection
ginal donor kidneys: correlation of histologic findings with graft dys- of a renal allograft associated with pretransplantation antibodies
function. Transplantation 2000;69(7):1352–7. directed against donor antigens on endothelium and monocytes. N
24. Escofet X, Osman H, Griffiths DF, Woydag S, Adam Jurewicz W. The Engl J Med 1979;300(22):1258–9.
presence of glomerular sclerosis at time zero has a significant impact 47. Sibley RK, Payne W. Morphologic findings in the renal allograft
on function after cadaveric renal transplantation. Transplantation biopsy. Sem Nephrol 1985;5(4):294–306.
2003;75(3):344–6.
408 Kidney Transplantation: Principles and Practice

48. Lucas ZJ, Coplon N, Kempson R, Cohn RB. Early renal transplant 71. August C, Schmid KW, Dietl KH, Heidenreich S. Prognostic value of
failure associated with subliminal sensitization. Transplantation lymphocyte apoptosis in acute rejection of renal allografts. Trans-
1970;10:522–8. plantation 1999;67(4):581–5.
49. Metzgar RS, Seigler HF, Ward FE, Rowlands DTJ. Immunologi- 72. Bishop GA, Hall BM, Duggin GG, Horvath JS, Sheil AG, Tiller
cal studies on elutes from human renal allografts. Transplantation DJ. Immunopathology of renal allograft rejection analyzed with
1972;13(131):131–7. monoclonal antibodies to mononuclear cell markers. ;29(708):
50. Colovai AI, Vasilescu ER, Foca-Rodi A, et al. Acute and hyperacute 708–17.
humoral rejection in kidney allograft recipients treated with anti- 73. Fuggle SV, McWhinnie DL, Morris PJ. Precise specificity of induced
human thymocyte antibodies. Hum Immunol 2005;66(5):501–12. tubular HLA-class II antigens in renal allografts. Transplantation
51. Gaber LW, Gaber AO, Tolley EA, Hathaway DK. Prediction by post- 1987;44(2):214–20.
revascularization biopsies of cadaveric kidney allografts of rejec- 74. Fuggle SV, McWhinnie DL, Chapman JR, Taylor HM, Morris PJ.
tion, graft loss, and preservation nephropathy. Transplantation Sequential analysis of HLA class II antigen expression in human
1992;53(6):1219–25. renal allografts: Induction of tubular class II antigens and correla-
52. Fusaro F, Murer L, Busolo F, Rigamonti W, Zanon GF, Zacchello tion with clinical parameters. Transplantation 1985;42:144–50.
G. CMV and BKV ureteritis: which prognosis for the renal graft? J 75. Waltzer WC, Miller F, Arnold A, Anaise D, Rapaport FT. Immunohis-
Nephrol 2003;16(4):591–4. tologic analysis of human renal allograft dysfunction. Transplanta-
53. Kirk AD, Mannon RB, Kleiner DE, et al. Results from a human tion 1987;43(1):100–5.
renal allograft tolerance trial evaluating T-cell depletion with 76. Barrett M, Milton AD, Barrett J, et al. Needle biopsy evaluation of
alemtuzumab combined with deoxyspergualin. Transplantation class II major histocompatibility complex antigen expression for the
2005;80(8):1051–9. differential diagnosis of cyclosporine nephrotoxicity from kidney
54. Almirall J, Campistol JM, Sole M, Andreu J, Revert L. Blood and graft graft rejection. Transplantation 1987;44:223–7.
eosinophilia as a rejection index in kidney transplant. Nephron 77. Ozdemir BH, Aksoy PK, Haberal AN, Demirhan B, Haberal M.
1993;65(2):304–9. Relationship of HLA-DR expression to rejection and mononu-
55. Nickeleit V, Vamvakas EC, Pascual M, Poletti BJ, Colvin RB. The clear cell infiltration in renal allograft biopsies. Renal Failure
prognostic significance of specific arterial lesions in acute renal 2004;26(3):247–51.
allograft rejection. J Am Soc Nephrol 1998;9:1301–8. 78. Nickeleit V, Hirsch HH, Zeiler M, et al. BK-virus nephropathy in renal
56. Meleg-Smith S, Gauthier PM. Abundance of interstitial eosinophils transplants-tubular necrosis, MHC-class II expression and rejection
in renal allografts is associated with vascular rejection. Transplanta- in a puzzling game. Neph Dial Transplant 2000;15(3):324–32.
tion 2005;79(4):444–50. 79. Faull RJ, Russ GR. Tubular expression of intercellular adhesion
57. Danilewicz M, Wagrowska-Danilewicz M. Immunohistochemical molecule-1 during renal allograft rejection. Transplantation
analysis of the interstitial mast cells in acute rejection of human 1989;48(2):226–30.
renal allografts. Med Sci Monit 2004;10(5):BR151–6. 80. Fuggle SV, Sanderson JB, Gray DW, Richardson A, Morris PJ. Varia-
58. Charney DA, Nadasdy T, Lo AW, Racusen LC. Plasma cell-rich acute tion in expression of endothelial adhesion molecules in pretrans-
renal allograft rejection. Transplantation 1999;68(6):791–7. plant and transplanted kidneys—correlation with intragraft events.
59. Aiello FB, Calabrese F, Rigotti P, et al. Acute rejection and graft sur- Transplantation 1993;55(1):117–23.
vival in renal transplanted patients with viral diseases. Mod Path 81. Brockmeyer C, Ulbrecht M, Schendel DJ, et al. Distribution of cell
2004;17(2):189–96. adhesion molecules (ICAM-1, VCAM-1, ELAM-1) in renal tissue dur-
60. Meehan SM, Domer P, Josephson M, et al. The clinical and patho- ing allograft rejection. Transplantation 1993;55(3):610–5.
logic implications of plasmacytic infiltrates in percutaneous renal 82. Briscoe DM, Pober JSS, Harmon WE, Cotran RS. Expression of vas-
allograft biopsies. Hum Pathol 2001;32(2):205–15. cular cell adhesion molecule-1 in human renal allografts. J Am Soc
61. Kataoka K, Naomoto Y, Shiozaki S, Matsuno T, Sakagami K, Oku- Nephrol 1992;3(5):1180–5.
mura OK. Infiltration of perforin-positive mononuclear cells into the 83. Niemann-Masanek U, Mueller A, Yard BA, Waldherr R, van der
rejected kidney allograft. Transplantation 1992;53(1):240–2. Woude FJ. B7-1 (CD80) and B7-2 (CD 86) expression in human tubu-
62. Pascoe MD, Marshall SE, Welsh KI, Fulton LM, Hughes DA. Increased lar epithelial cells in vivo and in vitro. Nephron 2002;92(3):542–56.
accuracy of renal allograft rejection diagnosis using combined perfo- 84. Morel D, Normand E, Lemoine C, et al. Tumor necrosis factor alpha
rin, granzyme B, and Fas ligand fine-needle aspiration immunocytol- in human kidney transplant rejection—analysis by in situ hybridiza-
ogy. Transplantation 2000;69(12):2547–53. tion. Transplantation 1993;55(4):773–7.
63. Akasaka Y, Ishikawa Y, Kato S, et al. Induction of Fas-mediated 85. Wong WK, Robertson H, Carroll HP, Ali S, Kirby JA. Tubulitis in
apoptosis in a human renal epithelial cell line by interferon-gamma: renal allograft rejection: role of transforming growth factor-beta
involvement of Fas-mediated apoptosis in acute renal rejection. Mod and interleukin-15 in development and maintenance of CD103+
Path 1998;11(11):1107–14. intraepithelial T cells. Transplantation 2003;75(4):505–14.
64. Kummer J, Wever P, Kamp A, ten Berge I, Hack C, Weening J. Expres- 86. Ozdemir BH, Ozdemir FN, Haberal N, Emiroglu R, Demirhan B,
sion of granzyme A and B proteins by cytotoxic lymphocytes involved Haberal M. Vascular endothelial growth factor expression and
in acute renal allograft rejection. Kidney Int 1995;47:70–7. cyclosporine toxicity in renal allograft rejection. Am J Transplant
65. Rowshani AT, Florquin S, Bemelman F, Kummer JA, Hack CE, Ten 2005;5(4 Pt 1):766–74.
Berge IJ. Hyperexpression of the granzyme B inhibitor PI-9 in human 87. Alchi B, Nishi S, Kondo D, et al. Osteopontin expression in acute
renal allografts: a potential mechanism for stable renal function in renal allograft rejection. Kidney Int 2005;67(3):886–96.
patients with subclinical rejection. Kidney Int 2004;66(4):1417–22. 88. Robertson H, Ali S, McDonnell BJ, Burt AD, Kirby JA. Chronic
66. Mengel M, Mueller I, Behrend M, et al. Prognostic value of cytotoxic renal allograft dysfunction: the role of T cell-mediated tubu-
T-lymphocytes and CD40 in biopsies with early renal allograft rejec- lar epithelial to mesenchymal cell transition. J Am Soc Nephrol
tion. Transpl Int 2004;17(6):293–300. 2004;15(2):390–7.
67. Meehan S, McCluskey R, Pascual M, Anderson P, Schlossman S, Col- 89. Kriz W, Kaissling B, Le Hir M. Epithelial-mesenchymal tran-
vin R. Cytotoxicity and apoptosis in human renal allografts: iden- sition (EMT) in kidney fibrosis: fact or fantasy? J Clin Invest
tification, distribution, and quantitation of cells with a cytotoxic 2011;121(2):468–74.
granule protein GMP-17 (TIA-1) and cells with fragmented nuclear 90. Farris AB, Colvin RB. Renal interstitial fibrosis: mechanisms and eval-
DNA. Lab Invest 1997;76:639–49. uation. Curr Opin Nephrol Hypertens 2012;21(3):289–300. Avail-
68. Noronha IL, Hartley B, Cameron JS, Waldherr R. Detection of IL-1 able online at: https://doi.org/10.1097/MNH.0b013e3283521cfa.
beta and TNF-alpha message and protein in renal allograft biopsies. 91. Tuazon TV, Schneeberger EE, Bhan AK, et al. Mononuclear cells in
Transplantation 1993;56(4):1026–9. acute allograft glomerulopathy. Am J Path 1987;129(1):119–32.
69. Ito H, Kasagi N, Shomori K, Osaki M, Adachi H. Apoptosis in the 92. Veronese F, Rotman S, Smith RN, et al. Pathological and clinical cor-
human allografted kidney. Analysis by terminal deoxynucleotidyl relates of FOXP3(+) cells in renal allografts during acute rejection. Am J
transferase-mediated DUTP-botin nick end labeling. Transplanta- Transplant 2007; 7(4);914–22.
tion 1995;60(8):794–8. 93. Robertson H, Wheeler J, Thompson V, Johnson JS, Kirby JA, Morley
70. Noronha IL, Oliveira SG, Tavares TS, et al. Apoptosis in kidney and AR. In situ lymphoproliferation in renal transplant biopsies. Histo-
pancreas allograft biopsies. Transplantation 2005;79(9):1231–5. chem Cell Biol 1995;104:331–4.
25 • Pathology of Kidney Transplantation 409

94. Kooijmans-Coutinho MF, Bruijn JA, Hermans J, et al. Evaluation by 116. Richardson WP, Colvin RB, Cheeseman SH, et al. Glomerulopathy
histology, immunohistology and PCR of protocollized renal biop- associated with cytomegalovirus viremia in renal allografts. N Engl J
sies 1 week post-transplant in relation to subsequent rejection epi- Med 1981;305(2):57–63.
sodes. Neph Dial Transplant 1995;10(6):847–54. 117. Messias NC, Eustace JA, Zachary AA, Tucker PC, Charney D,
95. Andrews PA, Finn JE, Lloyd CM, Zhou W, Mathieson PW, Sacks Racusen LC. Cohort study of the prognostic significance of acute
SH. Expression and tissue localization of donor-specific comple- transplant glomerulitis in acutely rejecting renal allografts. Trans-
ment C3 synthesized in human renal allografts. Europ J Immunol plantation 2001;72(4):655–60.
1995;25(4):1087–93. 118. Hiki Y, Leong AY, Mathew TH, Seymour AE, Pascoe V, Wood-
96. Pratt JR, Basheer SA, Sacks SH. Local synthesis of complement com- roofe AJ. Typing of intraglomerular mononuclear cells associated
ponent C3 regulates acute renal transplant rejection. Nature Med with transplant glomerular rejection. Clin Nephrol 1986;26(5):
2002;8(6):582–7. 244–9.
97. Strehlau J, Pavlakis M, Lipman M, et al. Quantitative detection of 119. Cosio FG, Sedmak DD, Henry ML, et al. The high prevalence of severe
immune activation transcripts as a diagnostic tool in kidney trans- early posttransplant renal allograft pathology in hepatitis C positive
plantation. Proc Nat Acad Sci 1997;94:695–700. recipients. Transplantation 1996;62(8):1054–9.
98. Sharma VK, Bologa RM, Li B, et al. Molecular executors of cell 120. Girlanda R, Kleiner DE, Duan Z, et al. Monocyte infiltration and kid-
death—differential intrarenal expression of Fas ligand, Fas, gran- ney allograft dysfunction during acute rejection. Am J Transplant
zyme B, and perforin during acute and/or chronic rejection of 2008;8(3):600–7.
human renal allografts. Transplantation 1996;62(12):1860–6. 121. Kirk AD, Hale DA, Mannon RB, et al. Results from a human renal
99. Lipman ML, Stevens AC, Strom TB. Heightened intragraft CTL allograft tolerance trial evaluating the humanized CD52-specific
gene expression in acutely rejecting renal allografts. J Immunol monoclonal antibody alemtuzumab (CAMPATH-1H). Transplanta-
1994;152(10):5120–7. tion 2003;76(1):120–9.
100. Strehlau J, Pavlakis M, Lipman M, Maslinski W, Shapiro M, Strom 122. Kawai T, Cosimi AB, Spitzer TR, et al. HLA-mismatched renal trans-
TB. The intragraft gene activation of markers reflecting T-cell-acti- plantation without maintenance immunosuppression. N Engl J Med
vation and -cytotoxicity analyzed by quantitative RT-PCR in renal 2008;358(4):353–61.
transplantation. Clin Nephrol 1996;46(1):30–3. 123. Farris AB, Taheri D, Kawai T, et al. Acute renal endothelial injury
101. Hoffmann SC, Hale DA, Kleiner DE, et al. Functionally significant during marrow recovery in a cohort of combined kidney and bone
renal allograft rejection is defined by transcriptional criteria. Am J marrow allografts. Am J Transplant 2011;11(7):1464–77.
Transplant 2005;5(3):573–81. 124. Leventhal J, Abecassis M, Miller J, et al. Chimerism and tolerance
102. Suthanthiran M. Molecular analyses of human renal allografts: without GVHD or engraftment syndrome in HLA-mismatched com-
differential intragraft gene expression during rejection. Kidney Int bined kidney and hematopoietic stem cell transplantation. Sci Trans-
1997;58:S15–21. lational Med 2012;4(124):124–8.
103. Desvaux D, Schwarzinger M, Pastural M, et al. Molecular diagno- 125. Kawai T, Sachs DH, Sprangers B, et al. Long-term results in recipients
sis of renal-allograft rejection: correlation with histopathologic of combined HLA-mismatched kidney and bone marrow transplan-
evaluation and antirejection-therapy resistance. Transplantation tation without maintenance immunosuppression. Am J Transplant
2004;78(5):647–53. 2014;14(7):1599–611.
104. Einecke G, Melk A, Ramassar V, et al. Expression of CTL associated 126. Neild GH, Taube DH, Hartley RB, et al. Morphological differentiation
transcripts precedes the development of tubulitis in T-cell mediated between rejection and cyclosporin nephrotoxicity in renal allografts.
kidney graft rejection. Am J Transplant 2005;5(8):1827–36. J Clin Path 1986;39(2):152–9.
105. Einecke G, Fairhead T, Hidalgo LG, et al. Tubulitis and epithelial 127. Sibley RK, Rynasiewicz J, Ferguson RM, et al. Morphology of
cell alterations in mouse kidney transplant rejection are inde- cyclosporine nephrotoxicity and acute rejection in patients
pendent of CD103, perforin or granzymes A/B. Am J Transplant immunosuppressed with cyclosporine and prednisone. Surgery
2006;6(9):2109–20. 1983;94(2):225–34.
106. Nast CC, Zuo XJ, Prehn J, Danovitch GM, Wilkinson A, Jordan SC. 128. Taube DH, Neild GH, Williams DG, et al. Differentiation between
Gamma-interferon gene expression in human renal allograft fine- allograft rejection and cyclosporin nephrotoxicity in renal trans-
needle aspirates. Transplantation 1994;57(4):498–502. plant recipients. Lancet 1985;2(171):171–4.
107. Schroeder TJ, Weiss MA, Smith RD, Stephens GW, First MR. 129. Marcussen N, Lai R, Olsen TS, Solez K. Morphometric and immu-
The efficacy of OKT3 in vascular rejection. Transplantation nohistochemical investigation of renal biopsies from patients with
1991;51(2):312–5. transplant ATN, native ATN, or acute graft rejection. Transplant
108. Kooijmans-Coutinho MF, Hermans J, Schrama E, et al. Intersti- Proc 1996;28(1):470–6.
tial rejection, vascular rejection, and diffuse thrombosis of renal 130. Curtis JJ, Julian BA, Sanders CE, Herrera GA, Gaston RS. Dilemmas
allografts: predisposing factors, histology, immunohistochemistry, in renal transplantation: when the clinical course and histological
and relation to outcome. Transplantation 1996;61(9):1338–44. findings differ. Am J Kidney Dis 1996;27(3):435–40.
109. Bates WD, Davies DR, Welsh K, Gray DW, Fuggle SV, Morris PJ. 131. Colvin RB, Nickeleit V. Renal transplant pathology. In: Jennette JC,
An evaluation of the Banff classification of early renal allograft Olson JL, Schwartz MM, Silva FG, editors. Heptinstall’s Pathology
biopsies and correlation with outcome. Neph Dial Transplant of the Kidney. 6th ed. Philadelphia, PA: Lippincott Williams and
1999;14(10):2364–9. Wilkins; 2006. p. 1347–490.
110. Bellamy CO, Randhawa PS. Arteriolitis in renal transplant 132. Khanduja S, Ghoshal U, Ghoshal UC. Phylogenetic analysis of genet-
biopsies is associated with poor graft outcome. Histopathology ically distinct Enterocytozoon bieneusi infecting renal transplant
2000;36(6):488–92. recipients. Acta Parasitol 2017;62(1):63–8.
111. Sis B, Bagnasco SM, Cornell LD, et al. Isolated endarteritis and kid- 133. Kicia M, Wesolowska M, Kopacz Z, et al. Prevalence and molecular
ney transplant survival: a multicenter collaborative study. J Am Soc characteristics of urinary and intestinal microsporidia infections in
Nephrol 2015;26(5):1216–27. renal transplant recipients. Clin Microbiol Infect 2016;22(5):462.
112. Alpers CE, Gordon D, Gown AM. Immunophenotype of vascular e465–e469.
rejection in renal transplants. Mod Path 1990;3(2):198–203. 134. Hocevar SN, Paddock CD, Spak CW, et al. Microsporidiosis acquired
113. Woywodt A, Schroeder M, Gwinner W, et al. Elevated numbers of through solid organ transplantation: a public health investigation.
circulating endothelial cells in renal transplant recipients. Trans- Ann Intern Med 2014;160(4):213–20.
plantation 2003;76(1):1–4. 135. Haas M, Loupy A, Lefaucheur C, et al. The Banff.2017 kidney
114. Watschinger B, Vychytil A, Attar M, et al. Pattern of endothelin meeting report: revised diagnostic criteria for chronic active T cell-
immunostaining during rejection episodes after kidney transplanta- mediated rejection, antibody-mediated rejection, and prospects
tion. Clin Nephrol 1994;41(2):86–93. for integrative endpoints for next-generation clinical trials. Am J
115. Trpkov K, Campbell P, Pazderka F, Cockfield S, Solez K, Halloran PF. Transplant 2018;18(2):293–307. Available online at: https://doi.
Pathologic features of acute renal allograft rejection associated with org/10.1111/ajt.14625.
donor-specific antibody. Analysis using the Banff grading schema. 136. Halloran PF, Schlaut J, Solez K, Srinivasa NS. The significance of the
Transplantation 1996;61(11):1586–92. anti-class I antibody response. II. Clinical and pathologic features of
410 Kidney Transplantation: Principles and Practice

renal transplants with anti-class I-like antibody. Transplantation 160. Magil AB, Tinckam K. Monocytes and peritubular capillary
1992;53(3):550–5. C4d deposition in acute renal allograft rejection. Kidney Int
137. Scornik JC, LeFor WM, Cicciarelli JC, et al. Hyperacute and acute kid- 2003;63(5):1888–93.
ney graft rejection due to antibodies against B cells. Transplantation 161. Nickeleit V, Zeiler M, Gudat F, Thiel G, Mihatsch MJ. Detection of
1992;54(1):61–4. the complement degradation product C4d in renal allografts: diag-
138. Fidler ME, Gloor JM, Lager DJ, et al. Histologic findings of antibody- nostic and therapeutic implications. J Am Soc Nephrol 2002;13(1):
mediated rejection in ABO blood-group-incompatible living-donor 242–51.
kidney transplantation. Am J Transplant 2004;4(1):101–7. 162. Lobo PI, Spencer CE, Stevenson WC, Pruett TL. Evidence demon-
139. Yard B, Spruyt-Gerritse M, Claas F, et al. The clinical significance strating poor kidney graft survival when acute rejections are asso-
of allospecific antibodies against endothelial cells detected with an ciated with IgG donor-specific lymphocytotoxin. Transplantation
antibody-dependent cellular cytotoxicity assay for vascular rejec- 1995;59:357–60.
tion and graft loss after renal transplantation. Transplantation 163. Poduval RD, Kadambi PV, Josephson MA, et al. Implications of
1993;55(6):1287–93. immunohistochemical detection of C4d along peritubular cap-
140. Collins AB, Chicano S, Cornell LD, et al. Putative antibody-mediated illaries in late acute renal allograft rejection. Transplantation
rejection with C4d deposition in HLA-identical, ABO compatible 2005;79(2):228–35.
renal allografts. Transplantation Proc 2006;38(10):3427–9. 164. Desvaux D, Le Gouvello S, Pastural M, et al. Acute renal allograft
141. Lorenz M, Regele H, Schillinger M, et al. Risk factors for capillary C4d rejections with major interstitial oedema and plasma cell-rich infil-
deposition in kidney allografts: evaluation of a large study cohort. trates: high {gamma}-interferon expression and poor clinical out-
Transplantation 2004;78(3):447–52. come. Nephrol Dial Transplant 2004;19(4):933–9.
142. Mauiyyedi S, Colvin RB. Humoral rejection in kidney transplanta- 165. Dragun D, Muller DN, Brasen JH, et al. Angiotensin II type 1-recep-
tion: new concepts in diagnosis and treatment. Curr Op Nephrol tor activating antibodies in renal-allograft rejection. N Engl J Med
Hypertens 2002;11(6):609–18. 2005;352(6):558–69.
143. Nickeleit V, Mihatsch MJ. Kidney transplants, antibodies and 166. Magil AB, Tinckam KJ. Focal peritubular capillary C4d deposition in
rejection: is C4d a magic marker? Nephrol Dial Transplant acute rejection. Nephrol Dial Transplant 2006;21(5):1382–8.
2003;18(11):2232–9. 167. Haas M, Ratner LE, Montgomery RA. C4d staining of perioperative
144. Rotman S, Collins AB, Colvin RB. C4d deposition in allografts: cur- renal transplant biopsies. Transplantation 2002;74(5):711–7.
rent concepts and interpretation. Transplant Rev 2005;19:65–77. 168. Sund S, Hovig T, Reisaeter AV, Scott H, Bentdal O, Mollnes TE. Com-
145. Feucht HE, Felber E, Gokel MJ, et al. Vascular deposition of comple- plement activation in early protocol kidney graft biopsies after living-
ment-split products in kidney allografts with cell-mediated rejection. donor transplantation. Transplantation 2003;75(8):1204–13.
Clin Experiment Immunol 1991;86(3):464–70. 169. Koo DD, Roberts IS, Quiroga I, et al. C4d deposition in early
146. Crespo M, Pascual M, Tolkoff-Rubin N, et al. Acute humoral rejec- renal allograft protocol biopsies. Transplantation 2004;78(3):
tion in renal allograft recipients: I. Incidence, serology and clinical 398–403.
characteristics. Transplantation 2001;71(5):652–8. 170. Mauiyyedi S, Crespo M, Collins AB, et al. Acute humoral rejection
147. Mauiyyedi S, Crespo M, Collins AB, et al. Acute humoral rejection in kidney transplantation: II. Morphology, immunopathology, and
in kidney transplantation: II. Morphology, immunopathology, and pathologic classification. J Am Soc Nephrol 2002;13(3):779–87.
pathologic classification. J Am Soc Nephrol 2002;13(3):779–87. 171. Bohmig GA, Exner M, Habicht A, et al. Capillary C4d deposition in
148. Racusen LC, Colvin RB, Solez K, et al. Antibody-mediated rejection kidney allografts: a specific marker of alloantibody-dependent graft
criteria - an addition to the Banff 97 classification of renal allograft injury. J Am Soc Nephrol 2002;13(4):1091–9.
rejection. Am J Transplant 2003;3(6):708–14. 172. Haas M, Rahman MH, Racusen LC, et al. C4d and C3d staining in
149. Burns JM, Cornell LD, Perry DK, et al. Alloantibody levels and acute biopsies of ABO- and HLA-incompatible renal allografts: correlation
humoral rejection early after positive crossmatch kidney transplan- with histologic findings. Am J Transplant 2006;6(8):1829–40.
tation. Am J Transplant 2008;8(12):2684–94. 173. Martin L, Guignier F, Mousson C, Rageot D, Justrabo E, Rifle G.
150. Sellares J, de Freitas DG, Mengel M, et al. Understanding the Detection of donor-specific anti-HLA antibodies with flow cytometry
causes of kidney transplant failure: the dominant role of anti- in eluates and sera from renal transplant recipients with chronic
body-mediated rejection and nonadherence. Am J Transplant allograft nephropathy. Transplantation 2003;76(2):395–400.
2012;12(2):388–99. 174. Grafft CA, Cornell LD, Gloor JM, et al. Antibody-mediated rejection
151. Lerut E, Kuypers DR, Verbeken E, et al. Acute rejection in non-com- following transplantation from an HLA-identical sibling. Nephrol
pliant renal allograft recipients: a distinct morphology. Clin Trans- Dial Transplant 2010;25(1):307–10.
plant 2007;21(3):344–51. 175. Konvalinka A, Tinckam K. Utility of HLA antibody testing in kidney
152. Takemoto SK, Zeevi A, Feng S, et al. National conference to assess transplantation. J Am Soc Nephrol 2015;26(7):1489–502.
antibody-mediated rejection in solid organ transplantation. Am J 176. Jackson AM, Sigdel TK, Delville M, et al. Endothelial cell antibod-
Transplant 2004;4(7):1033–41. ies associated with novel targets and increased rejection. J Am Soc
153. Cecka JM. Current methodologies for detecting sensitization to HLA Nephrol 2015;26(5):1161–71.
antigens. Curr Opin Organ Transplant 2011;16(4):398–403. 177. Duquesnoy RJ, Kamoun M, Baxter-Lowe LA, et al. Should HLA mis-
154. Gebel HM, Bray RA. The evolution and clinical impact of human match acceptability for sensitized transplant candidates be deter-
leukocyte antigen technology. Curr Opin Nephrol Hypertens mined at the high-resolution rather than the antigen level? Am J
2010;19(6):598–602. Transplant 2015;15(4):923–30.
155. Cosio FG, Lager DJ, Lorenz EC, Amer H, Gloor JM, Stegall MD. Signifi- 178. Duquesnoy RJ, Gebel HM, Woodle ES, et al. High-resolution HLA
cance and implications of capillaritis during acute rejection of kidney typing for sensitized patients: advances in medicine and science
allografts. Transplantation 2010;89(9):1088–94. require us to challenge existing paradigms. Am J Transplant
156. Loupy A, Suberbielle-Boissel C, Hill GS, et al. Outcome of sub- 2015;15(10):2780–1.
clinical antibody-mediated rejection in kidney transplant recipi- 179. Cecka JM, Reed EF, Zachary AA. HLA high-resolution typing for sen-
ents with preformed donor-specific antibodies. Am J Transplant sitized patients: a solution in search of a problem? Am J Transplant
2009;9(11):2561–70. 2015;15(4):855–6.
157. Loupy A, Haas M, Solez K, et al. The Banff 2015 kidney meeting 180. Wahrmann M, Exner M, Schillinger M, et al. Pivotal role of com-
report: current challenges in rejection classification and prospects plement-fixing HLA alloantibodies in presensitized kidney allograft
for adopting molecular pathology. Am J Transplant 2017;17(1): recipients. Am J Transplant 2006;6(5 Pt 1):1033–41.
28–41. 181. Nadasdy GM, Bott C, Cowden D, Pelletier R, Ferguson R, Nadasdy
158. Herman J, Lerut E, Van Damme-Lombaerts R, Emonds MP, Van T. Comparative study for the detection of peritubular capillary C4d
Damme B. Capillary deposition of complement C4d and C3d in deposition in human renal allografts using different methodologies.
pediatric renal allograft biopsies. Transplantation 2005;79(10): Hum Pathol 2005;36(11):1178–85.
1435–40. 182. Kuypers DR, Lerut E, Evenepoel P, Maes B, Vanrenterghem Y, Van
159. Regele H, Exner M, Watschinger B, et al. Endothelial C4d deposition Damme B. C3d deposition in peritubular capillaries indicates a vari-
is associated with inferior kidney allograft outcome independently of ant of acute renal allograft rejection characterized by a worse clinical
cellular rejection. Nephrol Dial Transplant 2001;16(10):2058–66. outcome. Transplantation 2003;76(1):102–8.
25 • Pathology of Kidney Transplantation 411

183. Nishi S, Imai N, Ito Y, et al. Pathological study on the relationship 206. Solez K. History of the Banff classification of allograft pathol-
between C4d, CD59 and C5b-9 in acute renal allograft rejection. Clin ogy as it approaches its. 20th year. Curr Opin Organ Transplant
Transplant 2004;18(Suppl. 11):18–23. 2010;15(1):49–51.
184. Jabs WJ, Logering BA, Gerke P, et al. The kidney as a second site 207. Racusen LC, Solez K, Colvin RB, et al. The Banff 97 working classifi-
of human C-reactive protein formation in vivo. Eur J Immunol cation of renal allograft pathology. Kidney Int 1999;55(2):713–23.
2003;33(1):152–61. 208. Solez K, Colvin RB, Racusen LC, et al. Banff ‘05 Meeting Report:
185. Imai N, Nishi S, Alchi B, et al. Immunohistochemical evidence of differential diagnosis of chronic allograft injury and elimina-
activated lectin pathway in kidney allografts with peritubular capil- tion of chronic allograft nephropathy (‘CAN’). Am J Transplant
lary C4d deposition. Nephrol Dial Transplant 2006;21(9):2589–95. 2007;7(3):518–26.
186. Hirohashi T, Chase CM, Della Pelle P, et al. A novel pathway of 209. Solez K, Colvin RB, Racusen LC, et al. Banff ‘05 meeting report:
chronic allograft rejection mediated by NK cells and alloantibody. differential diagnosis of chronic allograft injury and elimination of
Am J Transplant 2012;12(2):313–21. chronic allograft nephropathy (‘CAN’). Am J Transplant 2007;7(3):
187. Akiyoshi T, Hirohashi T, Alessandrini A, et al. Role of comple- 518–26.
ment and NK cells in antibody mediated rejection. Hum Immunol 210. Herzenberg AM, Gill JS, Djurdjev O, Magil AB. C4d deposition in
2012;73(12):1226–32. acute rejection: an independent long-term prognostic factor. J Am
188. Hidalgo LG, Sis B, Sellares J, et al. NK cell transcripts and NK cells Soc Nephrol 2002;13(1):234–41.
in kidney biopsies from patients with donor-specific antibodies: evi- 211. Saad R, Gritsch HA, Shapiro R, et al. Clinical significance of renal
dence for NK cell involvement in antibody-mediated rejection. Am J allograft biopsies with “borderline changes,” as defined in the Banff
Transplant 2010;10(8):1812–22. Schema. Transplantation 1997;64(7):992–5.
189. Hirohashi T, Chase CM, DellaPelle P, et al. Depletion of T regulatory 212. Schweitzer EJ, Drachenberg CB, Anderson L. Significance of the
cells promotes natural killer cell-mediated cardiac allograft vascu- Banff borderline biopsy. Am J Kid Dis 1996;28:585–91.
lopathy. Transplantation 2014;98(8):828–34. 213. Meehan SM, Siegel CT, Aronson AJ, et al. The relationship of
190. Haas M, Sis B, Racusen LC, et al. Banff 2013 meeting report: inclu- untreated borderline infiltrates by the Banff criteria to acute rejec-
sion of c4d-negative antibody-mediated rejection and antibody-asso- tion in renal allograft biopsies. J Am Soc Nephrol 1999;10(8):
ciated arterial lesions. Am J Transplant 2014;14(2):272–83. 1806–14.
191. Haas M, Mirocha J. Early ultrastructural changes in renal allografts: 214. Gough J, Rush D, Jeffery J, et al. Reproducibility of the Banff schema
correlation with antibody-mediated rejection and transplant glo- in reporting protocol biopsies of stable renal allografts. Nephrol Dial
merulopathy. Am J Transplant 2011;11(10):2123–31. Transplant 2002;17(6):1081–4.
192. Haas M. C4d-negative antibody-mediated rejection in renal 215. Veronese FV, Manfro RC, Roman FR, et al. Reproducibility of the
allografts: evidence for its existence and effect on graft survival. Clin Banff classification in subclinical kidney transplant rejection. Clin
Nephrol 2011;75(4):271–8. Transplant 2005;19(4):518–21.
193. Haas M. Pathologic features of antibody-mediated rejection in renal 216. Terasaki PI, Ozawa M. Predictive value of HLA antibodies and serum
allografts: an expanding spectrum. Curr Opin Nephrol Hypertens creatinine in chronic rejection: results of a 2-year prospective trial.
2012;21(3):264–71. Transplantation 2005;80(9):1194–7.
194. Loupy A, Suberbielle-Boissel C, Hill GS, et al. Outcome of sub- 217. Hourmant M, Cesbron-Gautier A, Terasaki PI, et al. Frequency
clinical antibody-mediated rejection in kidney transplant recipi- and clinical implications of development of donor-specific and non-
ents with preformed donor-specific antibodies. Am J Transplant donor-specific HLA antibodies after kidney transplantation. J Am
2009;9(11):2561–70. Soc Nephrol 2005;16(9):2804–12.
195. Sis B, Jhangri GS, Bunnag S, Allanach K, Kaplan B, Halloran PF. 218. Regele H, Bohmig GA, Habicht A, et al. Capillary deposition of com-
Endothelial gene expression in kidney transplants with alloantibody plement split product C4d in renal allografts is associated with base-
indicates antibody-mediated damage despite lack of C4d staining. ment membrane injury in peritubular and glomerular capillaries: a
Am J Transplant 2009;9(10):2312–23. contribution of humoral immunity to chronic allograft rejection. J
196. Stegall MD, Diwan T, Raghavaiah S, et al. Terminal complement Am Soc Nephrol 2002;13(9):2371–80.
inhibition decreases antibody-mediated rejection in sensitized renal 219. Gloor JM, Winters JL, Cornell LD, et al. Baseline donor-specific anti-
transplant recipients. Am J Transplant 2011;11(11):2405–13. body levels and outcomes in positive crossmatch kidney transplanta-
197. Thurman JM, Lucia MS, Ljubanovic D, Holers VM. Acute tubular tion. Am J Transplant 2010;10(3):582–9.
necrosis is characterized by activation of the alternative pathway of 220. Gloor JM, Sethi S, Stegall MD, et al. Transplant glomerulopathy:
complement. Kidney Int 2005;67(2):524–30. subclinical incidence and association with alloantibody. Am J Trans-
198. Artz MA, Steenbergen EJ, Hoitsma AJ, Monnens LA, Wetzels JF. plant 2007;7(9):2124–32.
Renal transplantation in patients with hemolytic uremic syndrome: 221. Haas M, Montgomery RA, Segev DL, et al. Subclinical acute anti-
high rate of recurrence and increased incidence of acute rejections. body-mediated rejection in positive crossmatch renal allografts. Am
Transplantation 2003;76(5):821–6. J Transplant 2007;7(3):576–85.
199. Lerut E, Kuypers D, Van Damme B. C4d deposition in the peritubular 222. Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Evolution and clinical
capillaries of native renal biopsies. Histopathology 2005;47(4):430–2. pathologic correlations of de novo donor-specific HLA antibody post
200. Sis B, Mengel M, Haas M, et al. Banff ‘09 meeting report: antibody kidney transplant. Am J Transplant 2012;12(5):1157–67.
mediated graft deterioration and implementation of Banff Working 223. Ivanyi B, Fahmy H, Brown H, Szenohradszky P, Halloran PF, Solez K.
Groups. Am J Transplant 2010;10(3):464–71. Available online at: Peritubular capillaries in chronic renal allograft rejection: a quanti-
https://doi.org/10.1111/j.1600-6143.2009.02987.x. tative ultrastructural study. Hum Pathol 2000;31(9):1129–38.
201. Colvin RB, Chang A, Farris AB, et al. Diagnostic Pathology: Kidney 224. Liapis G, Singh HK, Derebail VK, Gasim AM, Kozlowski T, Nickeleit
Diseases. Salt Lake City, UT: Amirsys; 2011. V. Diagnostic significance of peritubular capillary basement mem-
202. Takahashi K. Recent findings in ABO-incompatible kidney trans- brane multilaminations in kidney allografts: old concepts revisited.
plantation: classification and therapeutic strategy for acute anti- Transplantation 2012;94(6):620–9.
body-mediated rejection due to ABO-blood-group-related antigens 225. Baid-Agrawal S, Farris 3rd AB, Pascual M, et al. Overlapping path-
during the critical period preceding the establishment of accommo- ways to transplant glomerulopathy: chronic humoral rejection,
dation. Clin Experiment Nephrol 2007;11(2):128–41. hepatitis C infection, and thrombotic microangiopathy. Kidney Int
203. Velankar MM, Kini AR. Ocular involvement by T-cell post-trans- 2011;80(8):879–85.
plant lymphoproliferative disorder of cutaneous origin. Pathology 226. Smith RN, Kawai T, Boskovic S, et al. Chronic antibody medi-
2007;39(3):369–71. ated rejection of renal allografts: pathological, serological and
204. Bentall A, Cornell LD, Gloor JM, et al. Five-year outcomes in liv- immunologic features in nonhuman primates. Am J Transplant
ing donor kidney transplants with a positive crossmatch. Am J 2006;6(8):1790–8.
Transplant 2013;13(1):76–85. Available online at: https://doi. 227. Adam BA, Smith RN, Rosales IA, et al. Chronic antibody-medi-
org/10.1111/j.1600-6143.2012.04291.x. ated rejection in nonhuman primate renal allografts: validation
205. Bentall A, Tyan DB, Sequeira F, et al. Antibody-mediated rejec- of human histological and molecular phenotypes. Am J Trans-
tion despite inhibition of terminal complement. Transpl Int plant 2017;17(11):2841–50. Available online at: https://doi.
2014;27(12):1235–43. org/10.1111/ajt.14327.
412 Kidney Transplantation: Principles and Practice

228. Uehara S, Chase CM, Cornell LD, Madsen JC, Russell PS, Colvin RB. 251. Davenport A, Younie ME, Parsons JE, Klouda PT. Development of
Chronic cardiac transplant arteriopathy in mice: relationship of allo- cytotoxic antibodies following renal allograft transplantation is asso-
antibody, C4d deposition and neointimal fibrosis. Am J Transplant ciated with reduced graft survival due to chronic vascular rejection.
2007;7(1):57–65. Neph Dial Transplant 1994;9(9):1315–9.
229. Cosio FG, Grande JP, Wadei H, Larson TS, Griffin MD, Stegall MD. 252. Hill GS, Nochy D, Loupy A. Accelerated arteriosclerosis: a
Predicting subsequent decline in kidney allograft function from early form of transplant arteriopathy. Curr Opin Organ Transplant
surveillance biopsies. Am J Transplant 2005;5(10):2464–72. 2010;15(1):11–5.
230. Habib R, Broyer M. Clinical significance of allograft glomerulopathy. 253. Gago M, Cornell LD, Kremers WK, Stegall MD, Cosio FG. Kidney
Kidney Int 1993;43:S95–8. allograft inflammation and fibrosis, causes and consequences. Am J
231. Busch GJ, Galvanek EG, Reynolds ES. Human renal allografts: Transplant 2012;12(5):1199–207.
analysis of lesions in long-term survivors. Human Pathol 254. Burke BA, Chavers BM, Gillingham KJ, et al. Chronic renal allograft
1971;2(253):253–98. rejection in the first 6 months posttransplant. Transplantation
232. Hsu HC, Suzuki Y, Churg J, Grishman E. Ultrastructure of transplant 1995;60(12):1413–7.
glomerulopathy. Histopathology 1980;4(4):351–67. 255. Colvin R, Chase C, Winn H, Russell P. Chronic allograft arteriopathy:
233. Porter KA, Dossetor JB, Marchioro TL, Peart WS, Rendall JST, Tera- insights from experimental models. In: Orosz C, editor. Transplant
saki PI. Human renal transplants. I. Glomerular changes. Lab Invest Vascular Sclerosis. Austin, TX: R.G. Landes Biomedical Publishers;
1967;16(153):153–81. 1995. p. 7–34.
234. Colvin RB. Renal transplant pathology. In: Jennette JC, Olson JL, 256. Salomon RN, Hughes CC, Schoen FJ, Payne DD, Pober JS, Libby P.
Schwartz MM, Silva FG, editors. Heptinstall’s Pathology of the Kid- Human coronary transplantation-associated arteriosclerosis: evi-
ney. 5th ed. Philadelphia: Lippincott-Raven; 1998. p. 1409–540. dence for a chronic immune reaction to activated graft endothelial
235. Sijpkens YW, Joosten SA, Wong MC, et al. Immunologic risk factors cells. Am J Path 1991;138(4):791–8.
and glomerular C4d deposits in chronic transplant glomerulopathy. 257. Oguma S, Banner B, Zerbe T, Starzl T, Demetris AJ. Participation
Kidney Int 2004;65(6):2409–18. of dendritic cells in vascular lesions of chronic rejection of human
236. Glassock RJ, Feldman D, Reynolds ES, Dammin GJ, Merrill JP. allografts. Lancet 1988;2:933–6.
Human renal isografts: a clinical and pathologic analysis. Medicine 258. Gouldesbrough DR, Axelsen RA. Arterial endothelialitis in chronic
1968;47:411–24. renal allograft rejection: a histopathological and immunocytochem-
237. Olsen S, Bohman SO, Petersen VP. Ultrastructure of the glomerular ical study. Neph Dial Transplant 1994;9(1):35–40.
basement membrane in long term renal allografts with transplant 259. Fox WM, Hameed A, Hutchins GM, et al. Perforin expression local-
glomerular disease. Laboratory Investigation 1974;30(176):176– izing cytotoxic lymphocytes in the intimas of coronary arteries
89. with transplant-related accelerated arteriosclerosis. Hum Pathol
238. Porter KA, Andres GA, Calder MW, Dossetor JB, Hsu KC, Rendall JM, 1993;24(5):477–82.
et al. Human renal transplants. II. Immunofluorescence and immu- 260. Russell PS, Chase CM, Winn HJ, Colvin RB. Coronary atheroscle-
noferritin studies. Lab Invest 1968;18(159):159–75. rosis in transplanted mouse hearts. I. Time course and immu-
239. Kieran N, Wang X, Perkins J, et al. Combination of peritubular C4d nogenetic and immunopathological considerations. Am J Path
and transplant glomerulopathy predicts late renal allograft failure. J 1994;144(2):260–74.
Am Soc Nephrol 2009;20(10):2260–8. 261. McKenzie I, Whittingham S. Deposits of immunoglobulin and fibrin
240. Bishop GA, Waugh JA, Landers DV, Krensky AM, Hall BM. Micro- in human renal allografted kidneys. Lancet 1968;2(1313):1313–5.
vascular destruction in renal transplant rejection. Transplantation 262. Petersen VP, Olsen TS, Kissmeyer NF, et al. Late failure of human
1989;48(3):408–14. renal transplants: an analysis of transplant disease and graft failure
241. Ishii Y, Sawada T, Kubota K, Fuchinoue S, Teraoka S, Shimizu A. among 125 recipients surviving for one to eight years. Medicine
Injury and progressive loss of peritubular capillaries in the develop- 1975;54(45):45–71.
ment of chronic allograft nephropathy. Kidney Int 2005;67(1):321– 263. Andres GA, Accinni L, Hsu KC, et al. Human renal transplants: III.
32. Immunopathologic studies. Lab Invest 1970;22:588–95.
242. Mauiyyedi S, Pelle PD, Saidman S, et al. Chronic humoral rejection: 264. Russell PS, Chase CM, Colvin RB. Coronary atherosclerosis in trans-
identification of antibody-mediated chronic renal allograft rejec- planted mouse hearts. IV. Effects of treatment with monoclonal
tion by C4d deposits in peritubular capillaries. J Am Soc Nephrol antibodies to intercellular adhesion molecule-1 and leukocyte func-
2001;12(3):574–82. tion-associated antigen-1. Transplantation 1995;60(7):724–9.
243. Lerut E, Naesens M, Kuypers DR, Vanrenterghem Y, Van Damme 265. McLaren AJ, Marshall SE, Haldar NA, et al. Adhesion molecule
B. Subclinical peritubular capillaritis at 3 months is associated with polymorphisms in chronic renal allograft failure. Kidney Int
chronic rejection at 1 year. Transplantation 2007;83(11):1416– 1999;55(5):1977–82.
22. 266. Sedmak D, Sharma H, Czajka C, Ferguson R. Recipient endothelial-
244. Monga G, Mazzucco G, Messina M, Motta M, Quaranta S, Novara R. ization of renal allografts. An immunohistochemical study utilitizing
Intertubular capillary changes in kidney allografts: a morphologic blood group antigens. Transplantation 1988;46:907–10.
investigation on 61 renal specimens. Mod Path 1992;5(2):125–30. 267. Hruban RH, Long PP, Perlman EJ, et al. Fluorescence in situ hybrid-
245. Mazzucco G, Motta M, Segoloni G, Monga G. Intertubular capil- ization for the Y-chromosome can be used to detect cells of recipient
lary changes in the cortex and medulla of transplanted kidneys origin in allografted hearts following cardiac transplantation. Am J
and their relationship with transplant glomerulopathy: an ultra- Path 1993;142(4):975–80.
structural study of 12 transplantectomies. Ultrastructural Pathol 268. Kennedy LJ, Weissman IL. Dual origin of intimal cells in cardiac
1994;18(6):533–7. allograft arteriosclerosis. N Engl J Med 1971;285:884–8.
246. Colvin RB, Dvorak AM, Dvorak HF. Mast cells in the cortical tubu- 269. Sacchi G, Bertalot G, Cancarini C, Cristinelli L, Tonini G, Cannella
lar epithelium and interstitium in human renal disease. Hum Pathol G. Atheromatosis and double media: uncommon vascular lesions of
1974;5(3):315–26. renal allografts. Pathologica 1993;85(1096):183–94.
247. Colvin RB, Dvorak HF. Basophils and mast cells in renal allograft 270. Howie AJ, Bryan RL, Gunson BK. Arteries and veins formed within
rejection. Lancet 1974;1:212–4. renal vessels: a previously neglected observation. Virchows Arch A,
248. Desvaux D, Le Gouvello S, Pastural M, et al. Acute renal allograft Path Anat Histopathol. 1992;420(4):301–4.
rejections with major interstitial oedema and plasma cell-rich infil- 271. Porter KA. Renal transplantation. In: Heptinstall RH, editor. The
trates: high gamma-interferon expression and poor clinical out- pathology of the kidney. 4th ed. Boston: Little, Brown and Company;
come. Nephrol Dial Transplant 2004;19(4):933–9. 1990. p. 1799–933.
249. Hill GS, Nochy D, Bruneval P, et al. Donor-specific antibodies accel- 272. Gould VE, Martinez LV, Virtanen I, Sahlin KM, Schwartz MM. Differ-
erate arteriosclerosis after kidney transplantation. J Am Soc Nephrol ential distribution of tenascin and cellular fibronectins in acute and
2011;22(5):975–83. chronic renal allograft rejection. Lab Invest 1992;67(1):71–9.
250. Jeannet M, Pinn VW, Flax MH, Winn HJ, Russell PS. Humoral 273. Colvin RB. Pathology of renal allografts. In: Colvin RB, Bhan AK,
antibodies in renal allotransplantation in man. N Engl J Med McCluskey RT, editors. Diagnostic immunopathology. 2nd ed. New
1970;282(111):111–7. York: Raven Press; 1995. p. 329–66.
25 • Pathology of Kidney Transplantation 413

274. McManus BM, Malcom G, Kendall TJ, et al. Prominence of coro- 298. Schwarz A, Gwinner W, Hiss M, Radermacher J, Mengel M, Haller
nary arterial wall lipids in human heart allografts: implications for H. Safety and adequacy of renal transplant protocol biopsies. Am J
pathogenesis of allograft arteriopathy. Am J Path 1995;147(2): Transplant 2005;5(8):1992–6.
293–308. 299. Furness PN, Philpott CM, Chorbadjian MT, et al. Protocol biopsy of
275. Alpers CE, Davis CL, Barr D, Marsh CL, Hudkins KL. Identification of the stable renal transplant: a multicenter study of methods and com-
platelet-derived growth factor A and B chains in human renal vascu- plication rates. Transplantation 2003;76(6):969–73.
lar rejection. Am J Path 1996;148(2):439–51. 300. Rush DN, Henry SF, Jeffery JR, Schroeder TJ, Gough J. Histological
276. Fellström B, Klareskog L, Heldin CH, et al. Platelet-derived growth findings in early routine biopsies of stable renal allograft recipients.
factor receptors in the kidney—upregulated expression in inflamma- Transplantation 1994;57(2):208–11.
tion. Kidney Int 1989;36(6):1099–102. 301. Rush DN, Jeffery JR, Gough J. Sequential protocol biopsies in renal
277. Kerby JD, Verran DJ, Luo KL, et al. Immunolocalization of FGF-1 and transplant patients. Clinico-pathological correlations using the Banff
receptors in glomerular lesions associated with chronic human renal schema. Transplantation 1995;59(4):511–4.
allograft rejection. Transplantation 1996;62(2):190–200. 302. Nankivell BJ, Chapman JR. The significance of subclinical rejection
278. Noronha IL, Eberlein-Gonska M, Hartley B, Stephens S, Cameron JS, and the value of protocol biopsies. Am J Transplant 2006;6(9):2006–
Waldherr R. In situ expression of tumor necrosis factor-alpha, inter- 12.
feron-gamma, and interleukin-2 receptors in renal allograft biopsies. 303. Shishido S, Asanuma H, Nakai H, et al. The impact of repeated
Transplantation 1992;54:1017–24. subclinical acute rejection on the progression of chronic allograft
279. Wieczorek G, Bigaud M, Menninger K, et al. Acute and chronic vas- nephropathy. J Am Soc Nephrol 2003;14(4):1046–52.
cular rejection in non-human primate kidney tranplantation. Am J 304. Mengel M, Gwinner W, Schwarz A, et al. Infiltrates in protocol biop-
Transplant 2006;6(6):1285–96. sies from renal allografts. Am J Transplant 2007;7(2):356–65.
280. Cramer DV, Qian SQ, Harnaha J, et al. Cardiac transplantation in the 305. Park WD, Griffin MD, Cornell LD, Cosio FG, Stegall MD. Fibrosis with
rat. I. The effect of histocompatibility differences on graft arterioscle- inflammation at one year predicts transplant functional decline. J
rosis. Transplantation 1989;47:414–9. Am Soc Nephrol 2010;21(11):1987–97.
281. Adams DH, Tilney NL, Collins JJJ, Karnovsky MJ. Experimental graft 306. Mengel M, Bogers J, Bosmans JL, et al. Incidence of C4d stain in pro-
arteriosclerosis. I. The Lewis-to-F-344 allograft model. Transplanta- tocol biopsies from renal allografts: results from a multicenter trial.
tion 1992;53(5):1115–9. Am J Transplant 2005;5(5):1050–6.
282. Russell PS, Chase CM, Colvin RB. Alloantibody- and T cell-mediated 307. Grimm PC, McKenna R, Nickerson P, et al. Clinical rejection is
immunity in the pathogenesis of transplant arteriosclerosis: lack of distinguished from subclinical rejection by increased infiltra-
progression to sclerotic lesions in B cell-deficient mice. Transplanta- tion by a population of activated macrophages. J Am Soc Nephrol
tion 1997;64(11):1531–6. 1999;10(7):1582–9.
283. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Allen RD, Chap- 308. Shimizu A, Yamada K, Meehan SM, Sachs DH, Colvin RB. Intragraft
man JR. The natural history of chronic allograft nephropathy. N cellular events associated with tolerance in pig allografts: the “accep-
Engl J Med 2003;349(24):2326–33. tance reaction.” Transplant Proc. 1997;29:1155.
284. Rush D, Nickerson P, Gough J, et al. Beneficial effects of treatment 309. Russell PS, Chase CM, Colvin RB, Plate JM. Kidney transplants in
of early subclinical rejection: a randomized study. J Am Soc Nephrol mice: an analysis of the immune status of mice bearing long-term,
1998;9(11):2129–34. H-2 incompatible transplants. J Exp Med 1978;147(5):1449–68.
285. Cornell LD, Colvin RB. Chronic allograft nephropathy. Curr Op 310. Blancho G, Gianello PR, Lorf T, et al. Molecular and cellular events
Nephrol Hyperten 2005;14(3):229–34. implicated in local tolerance to kidney allografts in miniature swine.
286. Kuypers DR, Chapman JR, O’Connell PJ, Allen RD, Nankivell BJ. Pre- Transplantation 1997;63:26–33.
dictors of renal transplant histology at three months. Transplanta- 311. Shimizu A, Yamada K, Meehan SM, Sachs DH, Colvin RB. Acceptance
tion 1999;67(9):1222–30. reaction: intragraft events associated with tolerance to renal allografts
287. Mannon RB, Matas AJ, Grande J, et al. Inflammation in areas of in miniature swine. J Am Soc Nephrol 2000;11(12):2371–80.
tubular atrophy in kidney allograft biopsies: a potent predictor of 312. Lee I, Wang L, Wells AD, Dorf ME, Ozkaynak E, Hancock WW.
allograft failure. Am J Transplant 2010;10(9):2066–73. Recruitment of Foxp3+ T regulatory cells mediating allograft toler-
288. Mengel M, Reeve J, Bunnag S, et al. Scoring total inflammation is ance depends on the CCR4 chemokine receptor. J Experiment Med
superior to the current Banff inflammation score in predicting out- 2005;201(7):1037–44.
come and the degree of molecular disturbance in renal allografts. 313. Miyajima M, Chase CM, Alessandrini A, et al. Early acceptance of
Am J Transplant 2009;9(8):1859–67. renal allografts in mice is dependent on foxp3(+) cells. Am J Pathol
289. Mihatsch MJ, Morozumi K, Strom EH, Ryffel B, Gudat F, Thiel G. 2011;178(4):1635–45.
Renal transplant morphology after long-term therapy with cyclo- 314. Fudaba Y, Spitzer TR, Shaffer J, et al. Myeloma responses and tol-
sporine. Transplant Proc 1995;27(1):39–42. erance following combined kidney and nonmyeloablative marrow
290. Mihatsch MJ, Theil G, Spichtin HP, et al. Morphological findings in transplantation: in vivo and in vitro analyses. Am J Transplant
kidney transplants after treatment with cyclosporine. Transplant 2006;6(9):2121–33.
Proc 1983;15(Suppl. 1):2821–35. 315. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Chapman JR, Allen
291. Morozumi K, Takeda A, Uchida K, Mihatsch MJ. Cyclosporine neph- RD. Calcineurin inhibitor nephrotoxicity: longitudinal assessment
rotoxicity: how does it affect renal allograft function and transplant by protocol histology. Transplantation 2004;78(4):557–65.
morphology?. Transplant Proc 2004;36(Suppl. 2). 251S-6S. 316. Buehrig CK, Lager DJ, Stegall MD, et al. Influence of surveillance
292. Schwarz A, Mengel M, Gwinner W, et al. Risk factors for chronic renal allograft biopsy on diagnosis and prognosis of polyomavirus-
allograft nephropathy after renal transplantation: a protocol biopsy associated nephropathy. Kidney Int 2003;64(2):665–73.
study. Kidney Int 2005;67(1):341–8. 317. Roake JA, Fawcett J, Koo DD, Fuggle SV, Gray DW, Morris PJ. Late
293. Kee TY, Chapman JR, O’Connell PJ, et al. Treatment of subclinical reflush in clinical renal transplantation: protection against delayed
rejection diagnosed by protocol biopsy of kidney transplants. Trans- graft function not observed. Transplantation 1996;62(1):114–6.
plantation 2006;82(1):36–42. 318. Jain S, Curwood V, White SA, Furness PN, Nicholson ML. Sub-clin-
294. Moreso F, Ibernon M, Goma M, et al. Subclinical rejection associated ical acute rejection detected using protocol biopsies in patients with
with chronic allograft nephropathy in protocol biopsies as a risk fac- delayed graft function. Transplant Int 2000;13(Suppl. 1):S52–5.
tor for late graft loss. Am J Transplant 2006;6(4):747–52. 319. Mihatsch MJ, Ryffel B, Gudat F. The differential diagnosis between
295. Stegall MD, Park WD, Larson TS, et al. The histology of solitary renal rejection and cyclosporine toxicity. Kidney Int 1995;52:S63–9.
allografts at 1 and 5 years after transplantation. Am J Transplant 320. Ojo AO, Held PJ, Port FK, et al. Chronic renal failure after transplan-
2011;11(4):698–707. tation of a nonrenal organ. N Engl J Med 2003;349(10):931–40.
296. Colvin RB. Eye of the needle. Am J Transplant 2007;7(2):267–8. 321. Remuzzi G, Perico N. Cyclosporine-induced renal dysfunction in
297. El-Zoghby ZM, Stegall MD, Lager DJ, et al. Identifying specific causes experimental animals and humans. Kidney Int 1995;52:S70–4.
of kidney allograft loss. Am J Transplant 2009;9(3):527–35. 322. Mihatsch MJ, Thiel G, Ryffel B. Cyclosporine nephrotoxicity.
Advance Nephrol Necker Hosp 1988;17:303–20.
414 Kidney Transplantation: Principles and Practice

323. Marucci G, Morandi L, Macchia S, et al. Fibrinogen storage disease history and relationship to cyclosporine therapy. Virchows Arch A
without hypofibrinogenaemia associated with acute infection. His- Pathol Anat Histopathol 1991;418(2):129–41.
topathology 2003;42(1):22–5. 346. Antonovych TT, Sabnis SG, Austin HA, et al. Cyclosporine A-induced
324. Solez K, Racusen LC, Marcussen N, et al. Morphology of isch- arteriolopathy. Transplant Proc 1988;20(3 Suppl. 3):951–8.
emic acute renal failure, normal function, and cyclosporine toxic- 347. Strom EH, Epper R, Mihatsch MJ. Ciclosporin-associated arteriolopa-
ity in cyclosporine-treated renal allograft recipients. Kidney Int thy: the renin producing vascular smooth muscle cells are more sen-
1993;43(5):1058–67. sitive to ciclosporin toxicity. Clin Nephrol 1995;43(4):226–31.
325. Larsen S, Brun C, Duun S, Lokkegaard H, Thomsen HS. Early arte- 348. Strom EH, Thiel G, Mihatsch MJ. Prevalence of cyclosporine-associ-
riolopathy following “high-dose” cyclosporine in kidney transplan- ated arteriolopathy in renal transplant biopsies from 1981 to 1992.
tation. Apmis 1988;(Suppl.)4:66–73. Transplant Proc 1994;26(5):2585–7.
326. Bergstrand A, Bohmann SO, Farnsworth A, et al. Renal histopa- 349. Savoldi S, Scolari F, Sandrini S, Scaini P, Sacchi R, Tardanico R, et al.
thology in kidney transplant recipients immunosuppressed with Cyclosporine chronic nephrotoxicity: histologic follow up at 6 and 18
cyclosporin A: results of an international workshop. Clin Nephrol months after renal transplant. Transplant Proc 1988;20(3 Suppl. 3):
1985;24(3):107–19. 777–84.
327. Shulman H, Striker G, Deeg HJ, Kennedy M, Storb R, Thomas ED. 350. Young EW, Ellis CN, Messana JM, et al. A prospective study of renal
Nephrotoxicity of cyclosporin A after allogeneic marrow transplan- structure and function in psoriasis patients treated with cyclosporin.
tation: glomerular thromboses and tubular injury. N Eng J Med Kidney Int 1994;46(4):1216–22.
1981;305:1392–5. 351. Pei Y, Scholey JW, Katz A, Schachter R, Murphy GF, Cattran D.
328. Hochstetler LA, Flanigan MJ, Lager DJ. Transplant-associated Chronic nephrotoxicity in psoriatic patients treated with low-dose
thrombotic microangiopathy: the role of IgG administration as ini- cyclosporine. Am J Kidney Dis 1994;23(4):528–36.
tial therapy. Am J Kidney Dis 1994;23(3):444–50. 352. Sis B, Dadras F, Khoshjou F, Cockfield S, Mihatsch MJ, Solez K.
329. Candinas D, Keusch G, Schlumpf R, Burger HR, Gmur J, Largiader Reproducibility studies on arteriolar hyaline thickening scoring in
F. Hemolytic-uremic syndrome following kidney transplantation: calcineurin inhibitor-treated renal allograft recipients. Am J Trans-
prognostic factors. Schweizerische Med Wochenschrift J Suisse Med plant 2006;6(6):1444–50.
1994;124(41):1789–99. 353. Myers BD, Newton L, Boshkos C, et al. Chronic injury of human renal
330. Sommer BG, Innes JT, Whitehurst RM, Sharma HM, Ferguson microvessels with low-dose cyclosporine therapy. Transplantation
RM. Cyclosporine-associated renal arteriopathy resulting in loss of 1988;46(5):694–703.
allograft function. Am J Surg 1985;149(6):756–64. 354. Goes NB, Colvin RB. Case records of the Massachusetts General Hospi-
331. Mihatsch MJ, Gudat F, Ryffel B, Thiel G. Cyclosporine nephropathy. tal. Case 12-2007. A 56-year-old woman with renal failure after heart-
In: Tisher CC, Brenner BM, editors. Renal pathology: with clinical lung transplantation. N Engl J Med 2007; 19;356(16):1657–65.
and functional correlations. 2nd ed. Philadelphia: J.B. Lippincott; 355. Thiru S, Maher ER, Hamilton DV, Evans DB, Calne RY. Tubular
1994. p. 1641–81. changes in renal transplant recipients on cyclosporine. Transplant
332. Van den Berg-Wolf MG, Kootte AM, Weening JJ, Paul LC. Recur- Proc 1983;15:2846–51.
rent hemolytic uremic syndrome in a renal transplant recipient 356. Farnsworth A, Hall BM, Ng A, et al. Renal biopsy morphology in
and review of the Leiden experience. Transplantation 1988;45(1): renal transplantation. Am J Path Surg Path 1984;8:243–52.
248–51. 357. Rosen S, Greenfeld Z, Brezis M. Chronic cyclosporine-induced
333. Haas M, Sonnenday CJ, Cicone JS, Rabb H, Montgomery RA. Isomet- nephropathy in the rat. Transplantation 1990;49:445–52.
ric tubular epithelial vacuolization in renal allograft biopsy speci- 358. Dell’Antonio G, Randhawa PS. “Striped” pattern of medullary ray
mens of patients receiving low-dose intravenous immunoglobulin fibrosis in allograft biopsies from kidney transplant recipients main-
for a positive crossmatch. Transplantation 2004;78(4):549–56. tained on tacrolimus. Transplantation 1999;67(3):484–6.
334. Mihatsch M, Thiel G, Ryffel B. Cyclosporine nephrotoxicity. Advance 359. Zachariae H, Hansen HE, Kragballe K, Olsen S. Morphologic renal
Nephrol 1988;17:303–20. changes during cyclosporine treatment of psoriasis. Studies on pre-
335. Loomis LJ, Aronson AJ, Rudinsky R, Spargo BH. Hemolytic uremic treatment and posttreatment kidney biopsy specimens. J Am Acad
syndrome following bone marrow transplantation: a case report and Dermatol 1992;26:415–9.
review of the literature. Am J Kidney Dis 1989;14(4):324–8. 360. Palestine AG, Austin III HA, Balow JE, et al. Renal histopathologic
336. Noris M, Remuzzi G. Thrombotic microangiopathy after kidney alterations in patients treated with cyclosporine for uveitis. N Engl J
transplantation. Am J Transplant 2010;10(7):1517–23. Med 1986;314:1293–8.
337. Baid S, Pascual M, Williams Jr WW, et al. Renal thrombotic 361. Mihatsch MJ, Helmchen U, Casanova P, et al. Kidney biopsy findings
microangiopathy associated with anticardiolipin antibodies in in cyclosporine-treated patients with insulin-dependent diabetes
hepatitis C-positive renal allograft recipients. J Am Soc Nephrol mellitus. Klinische Wochenschrift 1991;69(8):354–9.
1999;10(1):146–53. 362. Messana JM, Johnson KJ, Mihatsch MJ. Renal structure and function
338. Myers BD, Ross J, Newton L, Luetscher J, Perlroth M. Cyclosporine- effects after low dose cyclosporine in psoriasis patients: a preliminary
associated chronic nephropathy. N Eng J Med 1984;311(699): report. Clin Nephrol 1995;43(3):150–3.
699–705. 363. Nadasdy T, Krenacs T, Kalmar KN, Csajbok E, Boda K, Ormos J. Impor-
339. Randhawa PS, Shapiro R, Jordan ML, Starzl TE, Demetris AJ. The tance of plasma cells in the infiltrate of renal allografts: an immuno-
histopathological changes associated with allograft rejection and histochemical study. Pathol Res Prac 1991;187(2-3):178–83.
drug toxicity in renal transplant recipients maintained on FK506. 364. Smith KD, Wrenshall LE, Nicosia RF, et al. Delayed graft function
Clinical significance and comparison with cyclosporine. Am J Path and cast nephropathy associated with tacrolimus plus rapamycin
Surg Path 1993;17(1):60–8. use. J Am Soc Nephrol 2003;14(4):1037–45.
340. Stegall MD, Park WD, Larson TS, et al. The histology of solitary renal 365. Reynolds JC, Agodoa LY, Yuan CM, Abbott KC. Thrombotic micro-
allografts at 1 and 5 years after transplantation. Am J Transplant angiopathy after renal transplantation in the United States. Am J
2011;11(4):698–707. Kidney Dis 2003;42(5):1058–68.
341. Nizze H, Mihatsch MJ, Zollinger HU, et al. Cyclosporine-associated 366. Letavernier E, Pe’raldi MN, Pariente A, Morelon E, Legendre C. Pro-
nephropathy in patients with heart and bone marrow transplants. teinuria following a switch from calcineurin inhibitors to sirolimus.
Clin Nephrol 1988;30:248–60. Transplantation 2005;80(9):1198–203.
342. Dische FE, Neuberger J, Keating J, Parsons V, Calne RY, Williams R. 367. van den Akker JM, Wetzels JF, Hoitsma AJ. Proteinuria following
Kidney pathology in liver allograft recipients after long-term treat- conversion from azathioprine to sirolimus in renal transplant recipi-
ment with cyclosporin A. Lab Invest 1988;58(4):395–402. ents. Kidney Int 2006;70(7):1355–7.
343. Myers BD, Sibley R, Newton L, et al. The long-term course of cyclo- 368. Stephany BR, Augustine JJ, Krishnamurthi V, et al. Differences in
sporine-associated chronic nephropathy. Kidney Int 1988;33(2): proteinuria and graft function in de novo sirolimus-based vs. cal-
590–600. cineurin inhibitor-based immunosuppression in live donor kidney
344. Yamaguchi Y, Teraoka S, Yagisawa T, Takahashi K, Toma H, Ota transplantation. Transplantation 2006;82(3):368–74.
K. Ultrastructural study of cyclosporine-associated arteriolopathy in 369. Dittrich E, Schmaldienst S, Soleiman A, Horl WH, Pohanka E.
renal allografts. Transplant Proc 1989;21(1 Pt 2):1517–22. Rapamycin-associated post-transplantation glomerulonephritis and
345. Rossmann P, Jirka J, Chadimova M, Reneltova I, Saudek F. Arte- its remission after reintroduction of calcineurin-inhibitor therapy.
riolosclerosis of the human renal allograft: morphology, origin, life Transplant Int 2004;17(4):215–20.
25 • Pathology of Kidney Transplantation 415

370. Straathof-Galema L, Wetzels JF, Dijkman HB, Steenbergen EJ, Hil- 395. Nickeleit V, Mihatsch MJ. Polyomavirus nephropathy: pathogenesis,
brands LB. Sirolimus-associated heavy proteinuria in a renal trans- morphological and clinical aspects. In: Kreipe HH, editor. Verh dtsch ges
plant recipient: evidence for a tubular mechanism. Am J Transplant pathol, 88. Tagung. Muenchen. Jena: Urban & Fischer; 2004. p. 69–84.
2006;6(2):429–33. 396. Nickeleit V, Steiger J, Mihatsch MJ. BK virus infection after kidney
371. Izzedine H, Brocheriou I, Frances C. Post-transplantation protein- transplantation. Graft 2002;5(Dec suppl.):S46–57.
uria and sirolimus. N Engl J Med 2005;353(19):2088–9. 397. Bracamonte ER, Furmanczyk PS, Smith KD, Nicosia RF, Alpers CE,
372. Kormendi F, Amend W. The importance of eosinophil cells in kidney Kowelewska J. Tubular basement membrane immune deposits asso-
allograft rejection. Transplantation 1988;45(3):537–9. ciated with polyoma virus nephropathy in renal allografts. Mod Path
373. Weir MR, Hall-Craggs M, Shen SY, et al. The prognostic value of 2006;19:259A.
the eosinophil in acute renal allograft rejection. Transplantation 398. Limaye AP, Smith KD, Cook L, et al. Polyomavirus nephropathy in
1986;41(6):709–12. native kidneys of non-renal transplant recipients. Am J Transplant
374. Hongwei W, Nanra RS, Stein A, Avis L, Price A, Hibberd AD. 2005;5(3):614–20.
Eosinophils in acute renal allograft rejection. Transplant Immunol 399. Lim AK, Parsons S, Ierino F. Adenovirus tubulointerstitial nephritis
1994;2(1):41–6. presenting as a renal allograft space occupying lesion. Am J Trans-
375. Hallgren R, Bohman SO, Fredens K. Activated eosinophil infiltration plant 2005;5(8):2062–6.
and deposits of eosinophil cationic protein in renal allograft rejec- 400. Yagisawa T, Nakada T, Takahashi K, Toma H, Ota K, Yaguchi H.
tion. Nephron 1991;59(2):266–70. Acute hemorrhagic cystitis caused by adenovirus after kidney trans-
376. Ten RM, Gleich GJ, Holley KE, Perkins JD, Torres VE. Eosinophil plantation. Urol Int 1995;54(3):142–6.
granule major basic protein in acute renal allograft rejection. Trans- 401. Nickeleit V. Critical commentary to: Acute adenoviral infection of
plantation 1989;47(6):959–63. a graft by serotype 35 following renal transplantation pathology,
377. Colvin RB, Fang LS-T. Interstitial nephritis. In: Tisher CC, Brenner research and practice. Jena 2003;199:701–2.
BM, editors. Renal pathology. 2nd ed. Philadelphia, PA: JB Lippin- 402. Singh HK, Nickeleit V. Kidney disease caused by viral infections.
cott; 1994. p. 723–68. Curr Diag Pathol 2004;10:11–21.
378. Farris AB, Ellis CL, Rogers TE, Chon WJ, Chang A, Meehan SM. Renal 403. Ito M, Hirabayashi N, Uno Y, Nakayama A, Asai J. Necrotizing tubu-
allograft granulomatous interstitial nephritis: observations of an lointerstitial nephritis associated with adenovirus infection. Hum
uncommon injury pattern in 22 transplant recipients. Clin Kidney J Pathol 1991;22(12):1225–31.
2017;10(2):240–8. 404. Bruno B, Zager RA, Boeckh MJ, et al. Adenovirus nephritis
379. Meehan SM, Josephson MA, Haas M. Granulomatous tubulointersti- in hematopoietic stem-cell transplantation. Transplantation
tial nephritis in the renal allograft. Am J Kidney Dis 2000;36(4):E27. 2004;77(7):1049–57.
380. Silbert PL, Matz LR, Christiansen K, Saker BM, Richardson M. Herpes 405. Mathur SC, Squiers EC, Tatum AH, et al. Adenovirus infection of the
simplex virus interstitial nephritis in a renal allograft. Clin Nephrol renal allograft with sparing of pancreas graft function in the recipi-
1990;33:264–8. ent of a combined kidney-pancreas transplant. Transplantation
381. Cosio FG, Roche Z, Agarwal A, Falkenhain ME, Sedmak DD, Fergu- 1998;65(1):138–41.
son RM. Prevalence of hepatitis C in patients with idiopathic glo- 406. Yang CW, Kim YS, Yang KH, Chang YS, Yoon YS, Bang BK.
merulonephritis in native and transplant kidneys. Am J Kidney Dis Acute focal bacterial nephritis presented as acute renal failure and
1996;28:752–8. hepatic dysfunction in a renal transplant recipient. Am J Nephrol
382. Nickeleit V, Mihatsch MJ. Polyomavirus nephropathy in native kid- 1994;14(1):72–5.
neys and renal allografts: an update on an escalating threat. Transpl 407. Gillum DM, Kelleher SP. Acute pyelonephritis as a cause of late
Int 2006;19(12):960–73. transplant dysfunction. Am J Path Med 1985;78(1):156–8.
383. Drachenberg CB, Hirsch HH, Ramos E, Papadimitriou JC. Polyoma- 408. Hansen BL, Rohr N, Svendsen V, Olsen H, Birkeland SA. Bacterial
virus disease in renal transplantation: review of pathological find- urinary tract infection in cyclosporine-A immunosuppressed renal
ings and diagnostic methods. Hum Pathol 2005;36(12):1245–55. transplant recipients. Scand J Infect Dis 1988;20(4):425–7.
384. Drachenberg CB, Beskow CO, Cangro CB, et al. Human polyoma 409. Kalra OP, Malik N, Minz M, Gupta KL, Sakhuja V, Chugh KS.
virus in renal allograft biopsies: morphological findings and correla- Emphysematous pyelonephritis and cystitis in a renal transplant
tion with urine cytology. Hum Pathol 1999;30(8):970–7. recipient—computed tomographic appearance. Int J Artificial Org
385. Nickeleit V, Hirsch HH, Binet IF, et al. Polyomavirus infection of 1993;16(1):41–4.
renal allograft recipients: from latent infection to manifest disease. J 410. Pearson JC, Amend Jr WJ, Vincenti FG, Feduska NJ, Salvatierra Jr O.
Am Soc Nephrol 1999;10(5):1080–9. Post-transplantation pyelonephritis: factors producing low patient
386. Mathur VS, Olson JL, Darragh TM, Yen TS. Polyomavirus-induced and transplant morbidity. J Urol 1980;123(2):153–6.
interstitial nephritis in two renal transplant recipients: case reports 411. Elkhammas EA, Mutabagani KH, Sedmak DD, Tesi RJ, Henry ML,
and review of the literature. Am J Kidney Dis 1997;29(5):754–8. Ferguson RM. Xanthogranulomatous pyelonephritis in renal
387. Pappo O, Demetris AJ, Raikow RB, Randhawa PS. Human polyoma allografts: report of 2 cases. J Urol 1994;151(1):127–8.
virus infection of renal allografts: histopathologic diagnosis, clinical 412. Jones BF, Nanra RS, Grant AB, Ferguson NW, White KH. Xantho-
significance, and literature review. Mod Path 1996;9(2):105–9. granulomatous pyelonephritis in a renal allograft: a case report.
388. Gardner SD, Field AM, Coleman DV, Hulme B. New human papova- J Urol 1989;141(4):926–7.
virus (B.K.) isolated from urine after renal transplantation. Lancet 413. Stern SC, Lakhani S, Morgan SH. Renal allograft dysfunction due
1971;1:1253–7. to vesicoureteric obstruction by nodular malakoplakia. Neph Dial
389. Coleman DV, MacKenzie EFD, Gardner SD, Poulding JM, Amer B, Transplant 1994;9(8):1188–90.
Russell WJI. Human polyoma virus (BK) infection and ureteric ste- 414. Bakir N, Sluiter WJ, Ploeg RJ, van Son WJ, Tegzess AM. Primary
nosis in renal allograft recipients. J Clin Path 1978;31:338–47. renal graft thrombosis. Neph Dial Transplant 1996;11(1):140–7.
390. Gardner SD, MacKenzie EF, Smith C, Porter AA. Prospective study of 415. Simmons RL, Tallent MB, Kjellstrand CM, Najarian JS. Renal allo­
the human polyomaviruses BK and JC and cytomegalovirus in renal graft rejection simulated by arterial stenosis. Surgery 1970;68(5):
transplant recipients. J Clin Path 1984;37(5):578–86. 800–4.
391. Hogan TF, Borden EC, McBain JA, Padgett BL, Walker DL. Human 416. Bruno S, Remuzzi G, Ruggenenti P. Transplant renal artery stenosis.
polyomavirus infections with JC virus and BK virus in renal trans- J Am Soc Nephrol 2004;15(1):134–41.
plant patients. Ann Intern Med 1980;92(3):373–8. 417. Said R, Duarte R, Chaballout A, el Boghdadly S, Nezamuddin N.
392. Drachenberg CB, Hirsch HH, Ramos E, Papadimitriou JC. Polyoma- Spontaneous rupture of renal allograft. Urology 1994;43(4):554–8.
virus disease in renal transplantation: review of pathological find- 418. Merion RM, Calne RY. Allograft renal vein thrombosis. Transplant
ings and diagnostic methods. Hum Pathol 2005;36(12):1245–55. Proc 1985;17:1746–50.
393. Drachenberg CB, Papadimitriou JC, Hirsch HH, et al. Histological 419. Schwarz A, Krause PH, Offermann G, Keller F. Impact of de novo
patterns of polyomavirus nephropathy: correlation with graft out- membranous glomerulonephritis on the clinical course after kidney
come and viral load. Am J Transplant 2004;4(12):2082–92. transplantation. Transplantation 1994;58(6):650–4.
394. Hirsch HH, Brennan DC, Drachenberg CB, et al. Polyomavirus-asso- 420. Marcen R, Pascual J, Quereda C, et al. Lupus anticoagulant
ciated nephropathy in renal transplantation: interdisciplinary analy- and thrombosis of kidney allograft vessels. Transplant Proc
ses and recommendations. Transplantation 2005;79(10):1277–86. 1990;22(4):1396–8.
416 Kidney Transplantation: Principles and Practice

421. Heidet L, Gagnadoux ME, Beziau A, Niaudet P, Broyer M, Habib R. 444. Neumayer HH, Huls S, Schreiber M, Riess R, Luft FC. Kidneys from
Recurrence of de novo membranous glomerulonephritis on renal pediatric donors: risk versus benefit. Clin Nephrol 1994;41(2):
grafts. Clin Nephrol 1994;41(5):314–8. 94–100.
422. Morales JM, Pascual-Capdevila J, Campistol JM, et al. Membranous 445. Meehan SM, Pascual M, Williams WW, et al. De novo collapsing glo-
glomerulonephritis associated with hepatitis C virus infection in merulopathy in renal allografts. Transplantation 1998;65(9):1192–7.
renal transplant patients. Transplantation 1997;63(11):1634–9. 446. Nadasdy T, Allen C, Zand MS. Zonal distribution of glomerular col-
423. Cruzado JM, Carrera M, Torras J, Grinyo JM. Hepatitis C virus infec- lapse in renal allografts: possible role of vascular changes. Hum
tion and de novo glomerular lesions in renal allografts. Am J Trans- Pathol 2002;33(4):437–41.
plant 2001;1(2):171–8. 447. Stokes MB, Davis CL, Alpers CE. Collapsing glomerulopathy in renal
424. Rodriguez EF, Cosio FG, Nasr SH, et al. The pathology and clinical allografts: a morphological pattern with diverse clinicopathologic
features of early recurrent membranous glomerulonephritis. Am J associations. Am J Kidney Dis 1999;33(4):658–66.
Transplant 2012;12(4):1029–38. 448. Cameron JS. Recurrent primary disease and de novo nephritis fol-
425. Monga G, Mazzucco G, Basolo B, et al. Membranous glomerulone- lowing renal transplantation. Pediatr Nephrol 1991;5(4):412–21.
phritis (MGN) in transplanted kidneys: investigation on 256 renal 449. Ramos EL, Tisher CC. Recurrent diseases in the kidney transplant.
allografts. Mod Path 1993;6(3):249–58. Am J Kidney Dis 1994;24(1):142–54.
426. Cosyns JP, Kazatchkine MD, Bhakdi S, et al. Immunohistochemical 450. Floege J. Recurrent glomerulonephritis following renal transplanta-
analysis of C3 cleavage fragments, factor H, and the C5b-9 termi- tion: an update. Nephrol Dial Transplant 2003;18(7):1260–5.
nal complex of complement in de novo membranous glomerulo- 451. Rodriguez EF, Cosio FG, Nasr SH, et al. The pathology and clinical
nephritis occurring in patients with renal transplant. Clin Nephrol features of early recurrent membranous glomerulonephritis. Am J
1986;26(4):203–8. Transplant 2012;12(4):1029–38.
427. Truong L, Gelfand J, D’Agati V, et al. De novo membranous glomeru- 452. Østerby R, Nyberg G, Karlberg I, Svalander C. Glomerular volume in
lonephropathy in renal allografts: a report of ten cases and review of kidneys transplanted into diabetic and non-diabetic patients. Diabet
the literature. Am J Kidney Dis 1989;14(2):131–44. Med 1992;9(2):144–9.
428. Antignac C, Hinglais N, Gubler MC, Gagnadoux MF, Broyer M, Habib 453. Wilczek HE, Jaremko G, Tyden G, Groth CG. Evolution of diabetic
R. De novo membranous glomerulonephritis in renal allografts in nephropathy in kidney grafts: evidence that a simulatneously
children. Clin Nephrol 1988;30(1):1–7. transplanted kidney exerts a protective effect. Transplantation
429. Thoenes GH, Pielsticker K, Schubert G. Transplantation-induced 1995;59:51–7.
immune complex kidney disease in rats with unilateral manifesta- 454. Stegall MD, Cornell LD, Park WD, Smith BH, Cosio FG. Renal
tions in the allografted kidney. Lab Invest 1979;41(321):321–9. allograft histology at 10 years after transplantation in the tacro-
430. Larsen CP, Walker PD. Phospholipase A2 receptor (PLA2R) staining limus era: evidence of pervasive chronic injury. Am J Transplant
is useful in the determination of de novo versus recurrent membra- 2018;18(1):180–8. Available online at: https://doi.org/10.1111/
nous glomerulopathy. Transplantation 2013;95(10):1259–62. ajt.14431.
431. Kashtan CE. Alport syndrome and thin glomerular basement mem- 455. Bohman SO, Tyden G, Wilczek H, et al. Prevention of kidney graft
brane disease. J Am Soc Nephrol 1998;9(9):1736–50. diabetic nephropathy by pancreas transplantation in man. Diabetes
432. Kashtan CE. Alport syndrome: renal transplantation and donor 1985;34:306–8.
selection. Renal Fail 2000;22(6):765–8. 456. Hariharan S, Smith RD, Viero R, First MR. Diabetic nephropathy
433. Bach D, Peters A, Rowemeier H, Degenhardt S, Helmchen U, Gra- after renal transplantation. Clinical and pathologic features. Trans-
bensee B. [Anti-basal membrane glomerulonephritis after homolo- plantation 1996;62(5):632–5.
gous kidney transplantation in hereditary Alport’s nephropathy]. 457. Bergstralh EJ, Monico CG, Lieske JC, et al. Transplantation outcomes
Deutsche Med Wochenschrift (1946) 1991;116(46):1752–6. in primary hyperoxaluria. Am J Transplant 2010;10(11):2493–501.
434. Vangelista A, Frasca GM, Martella D, Bonomini V. Glomeru- 458. Rosales IA, Collins AB, do Carmo PA, Tolkoff-Rubin N, Smith RN,
lonephritis in renal transplantation. Nephrol Dial Transplant Colvin RB. Immune complex tubulointerstitial nephritis due to auto-
1990;1:42–6. antibodies to the proximal tubule brush border. J Am Soc Nephrol
435. Diaz JI, Valenzuela R, Gephardt G, Novick A, Tubbs RR. Anti-glo- 2016;27(2):380–4.
merular and anti-tubular basement membrane nephritis in a renal 459. Larsen CP, Trivin-Avillach C, Coles P, et al. LDL receptor-related
allograft recipient with Alport’s syndrome. Ann Path Lab Med protein 2 (megalin) as a target antigen in human kidney anti-brush
1994;118(7):728–31. border antibody disease. J Am Soc Nephrol 2018;29(2):644–53.
436. Goldman M, Depierreux M, De Pauw L, et al. Failure of two sub- Available online at: https://doi.org/10.1681/ASN.2017060664.
sequent renal grafts by anti-GBM glomerulonephritis in Alport’s 460. Morath C, Mueller M, Goldschmidt H, Schwenger V, Opelz G,
syndrome: case report and review of the literature. Transplant Int Zeier M. Malignancy in renal transplantation. J Am Soc Nephrol
1990;3(2):82–5. 2004;15(6):1582–8.
437. Gobel J, Olbricht CJ, Offner G, et al. Kidney transplantation in Alport’s 461. Kasiske BL, Snyder JJ, Gilbertson DT, Wang C. Cancer after kid-
syndrome: long-term outcome and allograft anti-GBM nephritis. ney transplantation in the United States. Am J Transplant
Clin Nephrol 1992;38(6):299–304. 2004;4(6):905–13.
438. Mahan JD, Maver SM, Sibley RK, Vernier RL. Congenital nephrotic 462. Maisonneuve P, Agodoa L, Gellert R, et al. Cancer in patients on dial-
syndrome: evolution of medical management and results of trans- ysis for end-stage renal disease: an international collaborative study.
plantation. J Pediatr 1984;105. 549-7. Lancet 1999;354(9173):93–9.
439. Nyberg G, Friman S, Svalander C, Norden G. Spectrum of hereditary 463. Swerdlow SH, Campo E, Harris NL, et al., editors. WHO Classification
renal disease in a kidney transplant population. Neph Dial Trans- of Tumours of Haematopoietic and Lymphoid Tissues. Albany, NY:
plant 1995;10(6):859–65. WHO Publications Center; 2008.
440. Lane PH, Schnaper HW, Vernier RL, Bunchman TE. Steroid-depen- 464. Dotti G, Fiocchi R, Motta T, et al. Epstein-Barr virus-negative lym-
dent nephrotic syndrome following renal transplantation for con- phoproliferate disorders in long-term survivors after heart, kidney,
genital nephrotic syndrome. Pediatr Nephrol 1991;5(3):300–3. and liver transplant. Transplantation 2000;69(5):827–33.
441. Flynn JT, Schulman SL, deChadarevian JP, et al. Treatment of ste- 465. Nelson BP, Nalesnik MA, Bahler DW, Locker J, Fung JJ, Swerdlow
roid-resistant post-transplant nephrotic syndrome with cyclophos- SH. Epstein-Barr virus-negative post-transplant lymphoproliferative
phamide in a child with congenital nephrotic syndrome. Pediatr disorders: a distinct entity? Am J Surg Pathol 2000;24(3):375–85.
Nephrol 1992;6(6):553–5. 466. Quinlan SC, Pfeiffer RM, Morton LM, Engels EA. Risk factors for
442. Patrakka J, Ruotsalainen V, Reponen P, et al. Recurrence of early-onset and late-onset post-transplant lymphoproliferative dis-
nephrotic syndrome in kidney grafts of patients with congenital order in kidney recipients in the United States. Am J hematology
nephrotic syndrome of the Finnish type: role of nephrin. Transplan- 2011;86(2):206–9.
tation 2002;73(3):394–403. 467. Olagne J, Caillard S, Gaub MP, Chenard MP, Moulin B. Post-trans-
443. Woolley AC, Rosenberg ME, Burke BA, Nath KA. De novo focal plant lymphoproliferative disorders: determination of donor/recipi-
glomerulosclerosis after kidney transplantation. Am J Path Med ent origin in a large cohort of kidney recipients. Am J Transplant
1988;84(2):310–4. 2011;11(6):1260–9.
25 • Pathology of Kidney Transplantation 417

468. Opelz G, Dohler B. Lymphomas after solid organ transplanta- 479. Lavien G, Alger J, Preece J, Alexiev BA, Alexander RB. BK virus-asso-
tion: a collaborative transplant study report. Am J Transplant ciated invasive urothelial carcinoma with prominent micropapillary
2004;4(2):222–30. carcinoma component in a cardiac transplant patient: case report
469. Sampaio MS, Cho YW, Shah T, Bunnapradist S, Hutchinson IV. and review of literature. Clin Genitourin Cancer 2015;13(6):e397–9.
Impact of Epstein-Barr virus donor and recipient serostatus on the 480. Alexiev BA, Randhawa P, Vazquez Martul E, et al. BK virus-associ-
incidence of post-transplant lymphoproliferative disorder in kidney ated urinary bladder carcinoma in transplant recipients: report of
transplant recipients. Nephrol Dial Transplant 2012;27(7):2971–9. 2 cases, review of the literature, and proposed pathogenetic model.
470. Cherikh WS, Kauffman HM, McBride MA, Maghirang J, Swinnen LJ, Hum Pathol 2013;44(5):908–17.
Hanto DW. Association of the type of induction immunosuppression 481. Papadimitriou JC, Randhawa P, Rinaldo CH, Drachenberg CB,
with posttransplant lymphoproliferative disorder, graft survival, and Alexiev B, Hirsch HH. BK Polyomavirus infection and renourinary
patient survival after primary kidney transplantation. Transplanta- tumorigenesis. Am J Transplant 2016;16(2):398–406.
tion 2003;76(9):1289–93. 482. Emerson LL, Carney HM, Layfield LJ, Sherbotie JR. Collecting duct
471. Caillard S, Lelong C, Pessione F, Moulin B. Post-transplant lymphop- carcinoma arising in association with BK nephropathy post-trans-
roliferative disorders occurring after renal transplantation in adults: plantation in a pediatric patient: a case report with immunohis-
report of 230 cases from the French Registry. Am J Transplant tochemical and in situ hybridization study. Pediatr Transplant
2006;6(11):2735–42. 2008;12(5):600–5.
472. Arora S, Tangirala B, Osadchuk L, Sureshkumar KK. Belatacept: a 483. Dufek S, Haitel A, Muller-Sacherer T, Aufricht C. Duct Bellini car-
new biological agent for maintenance immunosuppression in kidney cinoma in association with BK virus nephropathy after lung trans-
transplantation. Expert Opin Biol Therapy 2012;12(7):965–79. plantation. J Heart Lung Transplant 2013;32(3):378–9.
473. Snanoudj R, Frangie C, Deroure B, et al. The blockade of T-cell co- 484. Mengel M, Gwinner W, Schwarz A, et al. Infiltrates in protocol biop-
stimulation as a therapeutic stratagem for immunosuppression: sies from renal allografts. Am J Transplant 2007;7(2):356–65.
focus on belatacept. Biol Targets Therapy 2007;1(3):203–13. 485. Schwimmer JA, Markowitz GS, Valeri AM, Imbriano LJ, Alvis
474. Trofe J, Buell JF, Beebe TM, et al. Analysis of factors that influence R, D’Agati VD. Secondary focal segmental glomerulosclerosis in
survival with post-transplant lymphoproliferative disorder in renal non-obese patients with increased muscle mass. Clin Nephrol
transplant recipients: the Israel Penn International Transplant Tumor 2003;60(4):233–41.
Registry experience. Am J Transplant 2005;5(4 Pt 1):775–80. 486. Farris AB, Colvin RB. Renal interstitial fibrosis: mechanisms and
475. Schmidtko J, Wang R, Wu CL, et al. Posttransplant lymphoprolifera- evaluation. Curr Opin Nephrol Hypertens 2012;21(3):289–300.
tive disorder associated with an Epstein-Barr-related virus in cyno- 487. Farris AB, Alpers CE. What is the best way to measure renal fibrosis?:
molgus monkeys. Transplantation 2002;73(9):1431–9. A pathologist’s perspective. Kidney Int 2014;4(1):9–15.
476. Randhawa PS, Magnone M, Jordan M, Shapiro R, Demetris AJ, Nale- 488. Farris AB, Chan S, Climenhaga J, et al. Banff fibrosis study: multi-
snik M. Renal allograft involvement by Epstein-Barr virus associated center visual assessment and computerized analysis of interstitial
post-transplant lymphoproliferative disease. Am J Path Surg Path fibrosis in kidney biopsies. Am J Transplant 2014;14(4):897–907.
1996;20(5):563–71. 489. Farris AB, Cohen C, Rogers TE, Smith GH. Whole slide imag-
477. Dalianis T, Hirsch HH. Human polyomaviruses in disease and can- ing for analytical anatomic pathology and telepathology: practi-
cer. Virology 2013;437(2):63–72. cal applications today, promises, and perils. Arch Pathol Lab Med
478. Alexiev BA, Papadimitriou JC, Chai TC, Ramos E, Staats PN, 2017;141(4):542–50.
Drachenberg CB. Polyomavirus (BK)-associated pleomorphic giant 490. Farris AB, Ellis CL, Rogers TE, Lawson D, Cohen C, Rosen S. Renal
cell carcinoma of the urinary bladder: a case report. Path Res Pract medullary and cortical correlates in fibrosis, epithelial mass, micro-
2013;209(4):255–9. vascularity, and microanatomy using whole slide image analysis
morphometry. PLoS One 2016;11(8):e0161019.
26 Biomarkers of Kidney
Injury and Rejection
PHILIP JOHN O’CONNELL, KAREN L. KEUNG, MADHAV C. MENON,
and BARBARA MURPHY

CHAPTER OUTLINE Definition of Biomarker and Surrogate Biomarkers of Chronic Allograft Injury
Endpoint Anti-HLA Antibodies as a Biomarker of
New Technologies for Biomarker Assays Graft Loss
Biomarkers of Acute Kidney Injury Development of Biomarkers for Studies of
Biomarkers of Acute Rejection in Renal Tolerance
Transplantation The Use of Biomarkers as an Enrichment
Potential Urinary Biomarkers Strategy for Randomized Trials of
Potential Peripheral Blood Biomarkers of Immunosuppressive Therapy in
Acute Rejection Transplantation
Conclusion

As is the case for other complex therapies and diseases, Definition of Biomarker and
transplantation is at the cusp of transitioning from empiri-
cal therapy where “one size fits all” to an era of personal- Surrogate Endpoint
ized medicine, where therapy is tailored to meet the specific
needs of an individual patient. Although the transplant Biomarker is one of the most misused terms in transplanta-
community has realized the inadequacy of our current tion. A PubMed search of “biomarker and kidney transplan-
treatments, we currently lack the tools to stratify patients tation” revealed 5857 articles. Yet most of these potential
according to predictive risk.1 We have limited options when biomarkers have not undergone the rigorous validation
it comes to induction and maintenance immunosuppres- required for clinical use. The definition of biomarker and
sion and no approved treatments for antibody-mediated SEPs are outlined in Table 26.1. According to the Biomark-
rejection (AMR; acute and chronic), delayed graft function ers Definitions Working Group a biomarker is: “a charac-
(DGF), or desensitization. Furthermore, apart from a trans- teristic that is objectively measured and evaluated as an
plant biopsy, we lack the diagnostic tools to differentiate indicator of normal biological processes, pathogenic pro-
acute kidney injury (AKI) from acute rejection and we lack cesses, or pharmacologic responses to a therapeutic inter-
the prognostic biomarkers that can reliably predict long- vention.”4 Predictive biomarkers (i.e., those that forecast
term outcomes at a time when a change in therapy may be the likely response to a specific treatment) require extensive
advantageous. external validation, ideally in external randomized clinical
To be able to evaluate new therapies for these condi- trials and meta-analysis.2 Biomarkers can take on many
tions there has been an increasing need for biomarkers forms, from cellular assays, to imaging or serum markers.
and surrogate endpoints (SEPs). In other specialties bio- However, as transplant immunosuppression becomes more
markers have been shown to reliably assess prognosis target specific, such as with newer agents like belatacept
and predict how individual patients will respond to treat- and Janus kinase (JAK) inhibitors, molecular and genetic
ment.2 Related to this, SEPs have the potential to more biomarkers will become increasingly important. If trans-
rapidly evaluate the clinical effectiveness of therapeutic plant immunosuppression is to become more personalized
interventions in clinical trials and reliably predict clinical then biomarker discovery is an essential prerequisite. With-
endpoints such as patient and graft survival. Although out validated biomarkers it will not be possible to rationally
there have been countless papers describing “biomark- stratify patients to different treatments based on risk and
ers” for specific conditions in transplantation, very few, if expected response to therapy. Therefore the first step is a
any, have undergone the rigorous validation required to sound understanding of the underlying pathophysiology of
fulfill the definition of a biomarker. The first steps to iden- what we are trying to prevent (e.g., acute cellular rejection
tifying a biomarker for conditions such as AKI or rejec- [ACR]). The more biologically plausible the biomarker is,
tion is to identify clinical indicators of each condition, to the more likely it is to be adopted for clinical use.
understand their effect on long-term outcomes, and most Prognostic biomarkers do not necessarily have to predict
importantly, to better understand the pathogenesis of the the effect of treatment on a particular outcome but they still
underlying condition.1,3 require robust statistical validation to be clinically useful

418
26 • Biomarkers of Kidney Injury and Rejection 419

(Fig. 26.1). First, there must be a demonstration that a cor- patients at high risk of graft loss would require a completely
relation exists between the biomarker and the outcome of different strategy. Recent trials have shown that we are not
interest (e.g., graft loss). Second, there must be an indepen- good at predicting this early after transplant,5–8 and a prog-
dent statistical validation of the relationship. Depending on nostic biomarker would be useful if it could reliably stratify
the robustness of the clinical data used to show an associa- patients to different treatment options.
tion between the biomarker and the endpoint further stud- Just as biomarkers are an essential component for stratify-
ies will be required. If the association was demonstrated on ing patients for clinical trials, SEPs will be an essential com-
retrospectively collected samples or tissues, then it needs to ponent of trial design. SEPs are measurements that provide
be validated prospectively and then by multicenter valida- early and accurate prediction of both the clinical endpoint
tion.2 Ultimately its clinical usefulness will require evalua- (e.g., graft loss) and the effects of treatment on this endpoint.
tion in randomized clinical trials where it is not clear what For surrogates to be fully accepted they need to be validated
the most appropriate treatment is for an individual patient in clinical trials. Whereas observational studies and meta-
based on the best clinical indicators available. For instance, analysis can demonstrate that a SEP is prognostic for disease
patients at low risk of rejection or graft loss may benefit outcome, it requires randomized clinical trials to show that
from minimizing immunosuppression over time, whereas the effect of the intervention on the SEP correlates sufficiently
with the effect on the true endpoint. This concept is more
advanced in oncology than it is in transplantation. Yet even
in oncology there are few SEP for solid tumors, whereas the
TABLE 26.1 Definitions of Biomarkers and Surrogate most accepted SEP is in nonsolid hematologic tumors where
Endpoints
clinical remission has long been regarded as a marker of time
Term Definition to disease progression and overall survival.2
Biomarker A characteristic that is objectively measured
There are several candidates for SEP in transplanta-
and evaluated as an indicator of normal tion. The most important SEP is one that predicts patient
biologic processes, pathogenic pro- death and/or graft loss. In transplantation, death and graft
cesses, or pharmacologic responses to a loss are relatively late events and therefore are impracti-
therapeutic intervention cal endpoints for clinical trials in transplantation. There-
Prognostic biomarker Biomarker that forecasts the likely course
of disease irrespective of treatment fore trials focused on events in the first 1 to 3 years tend
Predictive biomarker Biomarker that forecasts the likely to give little insight into important outcomes over the long
response to a specific treatment term.1,9 People have proposed several SEPs that are predic-
Clinical endpoint Measurement providing information tive of death and graft failure including: 1-year serum cre-
on how a patient feels, functions, or
survives
atinine,10,11 doubling of serum creatinine, acute rejection,
Surrogate endpoint Measurement providing early and accurate spot albumin:creatinine ratios, and delta estimated glomer-
prediction of both a clinical endpoint ular filtration rate (eGFR).12 Although none of these is per-
and the effects of treatment on this fect, delta eGFR seems the most promising at this point. A
endpoint good SEP needs to be both prevalent and predictive. When
Validation Confirmation by robust statistical methods
that a candidate prognostic or predictive evaluated in patients enrolled in the ANZDATA registry a
biomarker or surrogate endpoint fulfils a decline in eGFR of ≥30% from year 1 to 3 posttransplant
set of conditions that are necessary and not only was found to be reasonably common, occurring in
sufficient for its use in the clinic 10% of patients but also was associated with a 2.2-fold risk
From Buyse M, Sargent DJ, Grothey A, Matheson A, de Gramont A. Biomarkers
of death and a 5-fold increase in death-censored graft fail-
and surrogate end points—the challenge of statistical validation. Nat Rev ure.12 Other SEPs such as 1-year serum creatinine were less
Clin Oncol 2010;7(6):309–17. predictive than a delta measurement, whereas a doubling

Discovery Validation Randomized


Phase Phase Multicenter Validation
Trial Health Evaluation
Results Marker validated in separate Implementation
Biomarker Technology in Clinical
compared across platforms Multicenter of Biomarker
converted to Assessment Setting
to current gold & in different Trials
standard populations assay suitable for
clinical application

Fig. 26.1 Steps required to translate research findings predictive of a clinical outcome into a validated biomarker suitable for clinical use. (Adapted from
Willis JC, Lord GM. Immune biomarkers: the promises and pitfalls of personalized medicine. Nat Rev Immunol 2015;15(5):323–9.)
420 Kidney Transplantation: Principles and Practice

TABLE 26.2 Prentice Criteria of Use of Surrogate associated with graft injury and acute rejection. As with all
Endpoint in a Clinical Trial new technologies they need robust validation in the clini-
cal setting, and initial exploratory findings must be devel-
1. The treatment or intervention must affect the surrogate endpoint. oped into easy-to-use, reproducible, and accessible clinical
2. The treatment or intervention must affect the true endpoint.
3. The association of the surrogate endpoint and the true endpoint assays. Older genomic techniques confined the study of dis-
must be consistent between the treatment or intervention. ease phenomena to one or a few candidate genes at a given
4. There is an association between the surrogate and the true end- time. These genes were selected based on biologic plausibil-
point. ity18,19 or animal experimentation, and the studies were
hypothesis-driven.20 High-throughput technologies, such
as microarray and RNA-sequencing, are able to identify
the differential expression patterns of thousands of genes
of serum creatinine was more predictive than a ≥30% associated with a particular outcome or disease state. By
change in GFR but also less prevalent, occurring in only combining these with computational analysis it is possible
1% of recipients. Spot urine albumin:creatinine ratio might to reveal the cumulative signaling pathways operational
be predictive of hard endpoints but is difficult to standard- in a pathologic state. With this information, it is possible to
ize and lacks specificity.13 Other more recent predictors of develop gene-based biomarkers that can be used clinically
graft loss include inflammation in areas of interstitial fibrosis, to stratify patients according to risk.3
inflammation within areas of interstitial fibrosis and tubular Important characteristics of commonly employed strategies
atrophy (iIFTA), and de novo donor-specific antibody (DSA). to examine genomic information in research settings in trans-
More information is required to determine how predictive plantation are summarized in Table 26.3. Microarrays work
iIFTA is of graft loss using modern immunosuppression, but on the principle of DNA complementarity (see review).21
it does require a biopsy to derive the SEP.14 De novo DSA will The NanoString nCounter system is a new technology for
be discussed in detail in the later part of this chapter. molecular profiling that uses a novel color-coded barcode
The problem with all these measurements is that most technology, with each barcode representing a single target
have been validated from registry studies, which are retro- molecule.22 It enables digital counting of individual mole-
spective populations recruited over a long timeframe and cules without the need for amplification. Up to 800 transcripts
covering many different eras of immunosuppression. For can be profiled in a single reaction, and it has been applied to
instance, patients on cyclosporine and azathioprine may samples of blood, raw cells, and urine. Its role in transcrip-
have more episodes of rejections than those on other immu- tome profiling in other biomedical fields, such as oncology,
nosuppressive regimens, often with greater severity, and this has been prominent in recent years, but only recently has it
may skew the assessments of prediction. What is required been used in the setting of renal transplantation.23,24
now is to validate these tests in prospectively recruited Proteomics is the large-scale study of proteins. Previously,
patient cohorts on current standard of care immunosup- profiling of the proteome was limited by its complexity,
pression to confirm these original observations. If these but has gained significant momentum with technologic
observations are confirmed, the next step is to use the SEP in advancements, particularly in relation to mass spectrom-
a randomized clinical trial to test its clinical utility. For a SEP etry (MS). Urine has been frequently used in both nontar-
to be useful as an endpoint in a clinical trial the effect of any geted and targeted proteomic studies of the renal allograft,
trial therapy on the SEP must be equivalent to the effect on as has peripheral blood.25–33 Earlier studies using MS-based
the hard endpoint (e.g., death-censored graft loss). technology in urine proteomic profiling identified protein
In essence, the greater the association of the SEP with the signals that were present in subjects with acute rejection,
clinical endpoint and the greater the statistical validation, the but did not further characterize the protein associated with
more likely it is to be accepted for interventional clinical trials. these signals.34,35 In a proof of principle study, surface-
Ideally SEP should adhere to the Prentice criteria (Table 26.2). enhanced laser desorption/ionization—time of flight—mass
The surrogate should predict the clinical endpoint, treatment spectrometry (SELDI-TOF-MS) was used to discover urine
or intervention should have a significant effect on both the protein peaks associated with ACR. A follow-up study
surrogate and the clinical endpoint, and the treatment effect revealed that these were protein derivatives of nontryptic
on the surrogate should mirror the treatment effect on the cleaved forms of β-2 microglobulin.36 However, a multi-
clinical endpoint.15 This latter point is ideal but has been center study did not identify a statistically significant cor-
proven to be unrealistic. An alternative definition is that the relation of intact or cleaved β-2 microglobulin in urine in
surrogate must be prognostic of transplant outcome and the the presence of acute rejection.33 This prospective targeted
effect of treatment on the SEP must correlate sufficiently with observational study conducted through the Clinical Trials
the effect on the true outcome.2,16,17 Either way the correla- in Organ Transplantation-01 (CTOT-1) protocol is described
tion must be validated in clinical trials or meta-analysis.2 In in more detail later. Other studies have applied a variety of pro-
addition, the SEP should be biologically plausible. teomic techniques to identify protein expression in an unbi-
ased high-throughput manner in allograft rejection, before
using targeted enzyme-linked immunosorbent assay (ELISA)
New Technologies for Biomarker for validation of biomarker panels.27,31,37 However, as with
Assays the majority of promising proteomic studies to date, multi-
center prospective trials are required to validate these candi-
The advent of the new “omic” technologies has trans- date markers before meaningful conclusions can be made.
formed the field of biomarker development. They provide Transcriptomics is the study of the RNA transcripts encoded
new tools to accurately identify the underlying pathology by the genome. Although much of the work in the past decade
26 • Biomarkers of Kidney Injury and Rejection 421

TABLE 26.3 Summary of High-Throughput Technologies Used in Modern Biomarker Discovery


Advantages Disadvantages
Quantitative RT-PCR □ Established gold standard of quantitative expression □ Low throughput—limited number of genes/transcripts can
profiling be evaluated in a single experiment—often used for valida-
□ High sensitivity tion.
□ High dynamic range □ Larger amounts of sample required compared with other
□ High reproducibility methods
□ Depends on fidelity of “housekeeping genes”
Microarray □ Well established method □ Reference genome required (i.e., determines relative and not
□ Widely available absolute expression)
□ Medium to high throughput—thousands of genes/ □ Only detects sequences complementary to those on the array
transcripts can be profiled in a single experiment □ Lower dynamic range
□ High reproducibility (at mid to high expression range) □ Lower specificity and sensitivity compared with other tech-
□ Publicly available databases on thousands of niques
microarray studies □ Analysis can be difficult comparing data sets from different
□ Relatively low cost microarray platforms
Next Generation □ High throughput—entire transcriptome can be □ Comparatively more expensive per sample
Sequencing (NGS) profiled in a single experiment □ Sequence-specific biases
□ Can identify novel transcripts □ Complex/time-consuming computational data analysis
□ Direct digital counting
□ High dynamic range
□ High sensitivity
□ High reproducibility
NanoString nCounter □ Medium throughput—up to 800 genes/transcripts in □ High upfront costs of code sets and consumables
analysis system a single experiment □ Prior knowledge of genes required
□ Direct digital counting □ Not ideal for nontargeted gene discovery studies—fewer
□ No reverse transcription or RNA amplification step, genes/transcripts per experiment compared with NGS and/or
therefore reducing technical biases microarray
□ High sensitivity and specificity
□ High dynamic range
□ High reproducibility
□ Paraffin-embedded tissues can be used
□ Rapid turnaround time (from sample processing to
data acquisition)

From Menon MC, Keung KL, Murphy B, O’Connell PJ. The use of genomics and pathway analysis in our understanding and prediction of clinical renal transplant
injury. Transplantation 2016;100(7):1405–14.

has been in mRNA evaluation, studies in microRNA (miRNA) metabolism have been well documented,46–48 studies dem-
expression profiling in the setting of renal transplantation has onstrating its potential role in monitoring kidney transplants
increased exponentially in recent years. Small, noncoding are limited. It has been used to identify borderline tubulitis
RNAs, miRNAs regulate gene expression of target mRNAs. and acute T cell mediated rejection in pediatric renal trans-
Similar to genome and mRNA profiling, large-scale sequenc- plant recipients.49 Smaller studies have also been published
ing of miRNAs has been dominated by microarray applica- evaluating the differential urinary and serum metabolomic
tions. One of the advantages of this technology is that there profile in adult allograft recipients with acute rejection.50–52
are far fewer miRNA than genes and hence the computational
analysis is simpler, and there remains the possibility that a
more robust marker could be identified as a predictor of either Biomarkers of Acute Kidney Injury
acute rejection or IFTA. Scian and Mas38,39 have elegantly
summarized miRNA profiling studies of the renal allograft Being able to differentiate AKI from acute rejection would
in their reviews. To date no conclusive patterns of miRNA be a useful tool in the management of transplant recipients
expression have been associated with acute rejection40–42 or especially in the first few weeks after transplantation. In
IFTA.43–45 addition, a test undertaken in the donor or within the donor
Metabolomics relates to the global profiling of small mol- allograft that predicted DGF or ultimate graft performance
ecule (<1500 Da) metabolites. Because the genome, tran- would help in donor graft allocation and the manage-
scriptome, and proteome can undergo further changes ment of patients postoperatively. In nephrology, neutro-
after translation, the metabolome reflects the downstream phil gelatinase associated lipocalin (NGAL), kidney injury
products of these processes. There have been advances in molecule-1 (Kim-1), and N-acetyl-b-D-glucosaminidase
separation techniques, such as capillary electrophoresis and (NAG) have been identified as biomarkers of AKI.53 NGAL
ultrahigh-pressure liquid chromatography (UPLC) systems, is released from tubules, tends to rise before serum creati-
and improvements in analytic approaches, being MS-based nine, and is not affected by volume depletion.54–57 Kim-1
methods and high-resolution nuclear magnetic resonance was overexpressed in kidney biopsy specimens in patients
(NMR) spectroscopy. This has enabled the simultaneous with acute tubular injury, and urine excretion of Kim-1
study of dozens of small molecule metabolites in tissue and has been shown to distinguish acute tubular injury from
biologic fluids. Whereas the use of nontargeted metabo- other forms of AKI.58 In kidney transplant recipients, high
lomic approaches in the study of renal diseases and drug urinary excretion of Kim-1 was found to be an independent
422 Kidney Transplantation: Principles and Practice

predictor of graft loss within a 4-year follow-up with hazard significantly associated with DGF, deceased donor status
ratios between 3.6 and 5.1.59 However, its sensitivity and and interstitial inflammation, but not histology. IRRAT
specificity as a test were not evaluated. scores have since been observed to be elevated in both ACR
The advantage of these biomarkers is that they are and AMR, suggesting the nonspecific nature of this injury
measured in the urine and do not require renal tissue for response signature.69,70 Of note, AKI-related gene expres-
analysis. However, evidence of the utility of these bio- sion patterns identified by different groups in transplan-
markers in transplantation has been sparse. When mea- tation have shown significant overlap with both mouse
sured in deceased donor urine, NGAL, in combination orthologs identified in ischemia-reperfusion models,66,71
with liver fatty acid-binding protein (LFABP), were pre- and with each other, suggesting molecular homogeneity in
dictors of kidney injury and were associated with worse this injury response.67,68,72 Currently, genetic markers of
eGFR at 6 months,60 whereas expression of YKL-40, a AKI have relied on analysis of biopsy material. From a clini-
marker of kidney repair, was associated with improved cal utility perspective, this makes it no more beneficial than
function after DGF.61 Other studies have shown an asso- histologic evaluation, which is the current gold standard.
ciation between elevated urinary levels of NGAL and For biomarker development IRRAT scores or other genetic
interleukin (IL)-18 in deceased donors and the onset markers of AKI would need prospective validation in a clini-
of DGF in the recipient.62,63 However, in both cases the cal trial and ideally would use serum or urine for analysis,
association was modest and, as yet, have not been intro- thereby avoiding the requirement for a biopsy.
duced into clinical practice. In a longitudinal study of 182
transplant patients, elevated urinary NGAL levels were
somewhat predictive of AKI from all causes other than
Biomarkers of Acute Rejection in
rejection (specificity 0.86 and sensitivity 0.86), whereas Renal Transplantation
if very high levels of NGAL were present it was highly pre-
dictive of acute rejection (sensitivity 1.00 and specificity Biopsy of the renal allograft remains the gold standard for the
0.93).64 Some of these molecules may ultimately be useful diagnosis of acute rejection and/or other pathologies in the
in clinical settings, but further prospective evaluation in transplanted organ—the histologic profile aids therapeutic
clinical trials is required. Other urinary molecules of AKI decisions and may provide prognostic value. Nevertheless,
have been evaluated in the transplant setting. Cystatin C biopsies are invasive with inherent risks, and variability in
showed an association with DGF but with a receiver oper- sampling and interobserver biopsy scoring raises questions
ating characteristic (ROC) area under the curve of 0.54 regarding their diagnostic accuracy.73 Serum creatinine
to 0.74, the accuracy in the transplant setting was low.65 and proteinuria remain established noninvasive mark-
The problem with many of these assays in the trans- ers used for allograft surveillance, but a change in these
plant setting is that AKI is often seen in association with parameters is neither sensitive nor specific to a specific etiol-
acute rejection. Hence, developing an assay that can reli- ogy and still necessitates a biopsy. Furthermore, subclinical
ably separate nonimmunologic causes of AKI and rejec- rejection, by definition, is not accompanied by an elevation
tion-associated AKI has been difficult. More recently, in the serum creatinine. The identification of noninvasive
genomics has been used to develop a molecular signature biomarkers of acute rejection with high diagnostic accu-
of AKI. Paradoxically it has been difficult to develop such racy based on their sensitivity, specificity, positive predic-
a signature in studies of renal disease, because AKI is self- tive value (PPV), and/or negative predictive value (NPV),
limiting and renal biopsies are not often performed. Kid- which could assist in screening patients for biopsy or to
ney transplants experience AKI and are often biopsied in replace the need for a biopsy altogether, is the holy grail in
these circumstances, and are thus ideally positioned to biomarker discovery in transplantation.
study the gene expression patterns of AKI.66 cDNA micro- The establishment and widespread application of high-
arrays were used to study the gene expression patterns of throughput omics technologies over the past two decades
14 living donor kidneys, 15 deceased donor kidneys with has generated large-scale data and provided greater under-
immediate function, and 14 deceased donor kidneys with standing of the molecular pathways and mechanisms in
AKI (defined by dialysis in first week). The gene expression various states of allograft injury, with much of the focus on
patterns of living donor kidneys were observed to cluster acute rejection.74 Furthermore, it has provided opportuni-
separately from deceased donor kidneys (132 genes). In ties to discover biomarkers in acute rejection of diagnostic,
kidney transplants with AKI immediately after transplant predictive, or prognostic relevance in blood (sera, peripheral
there were 48 transcripts related to cell cycle regulation, cell blood mononuclear cells) and/or urine. A number of earlier
growth/metabolism, and signal transduction that were sig- studies in the biomarker search in acute rejection employed
nificantly differentially regulated compared with those with more traditional laboratory techniques such as quantita-
immediate function.67 In a separate study, the same authors tive PCR (qPCR) and ELISA to measure mRNA and protein
compared gene expression patterns of AKI allografts with levels, respectively, of a select number of molecules in a tar-
carefully selected, pristine 6-week protocol biopsies, and geted, or “biased” manner. However, genomic/transcrip-
identified 394 transcripts that were differentially expressed tomic technologies such as microarray, RNA sequencing,
in AKI.68 Using the geometric mean of the fold increase in and NanoString, and myriad proteomic strategies, coupled
the top 30 differentially regulated genes (compared with with exponential advancement of computational analytical
control nephrectomies), they devised an IRRAT (injury- methods have enabled the simultaneous measurement of
repair-response-associated-transcript) score that corre- tens to hundreds of thousands of molecules in a disease state
lated inversely with renal function at the time, and directly and provided opportunities to identify potential biomarkers
with eGFR improvement thereafter. The IRRAT score was using a nontargeted approach.75 These have enabled us to
26 • Biomarkers of Kidney Injury and Rejection 423

go beyond the study of genes, RNA, and proteins as poten- Potential Urinary Biomarkers
tial biomarkers, but to evaluate even smaller molecules,
including miRNA and peptides. Being a direct product of the allograft, urine is a par-
To identify any biomarker, understanding the under- ticularly attractive biofluid for evaluating potential bio-
lying pathogenesis of the condition is essential. The basic markers. Before the widespread application of the omics
mechanisms of acute rejection in renal allografts are well disciplines, substantial progress had been made by using
understood and extensively reviewed.76,77 Important fea- qPCR to interrogate urine obtained concurrently from
tures to emerge include the need not only to appreciate the patients with biopsy-proven acute rejection for prese-
shared immune mechanisms between AMR and ACR, but lected immune activation genes of interest.76 In one of
also to recognize the biologic pathways that are unique to the early landmark studies, Suthanthiran and colleagues
each rejection type and thus confer differences in progno- selected granzyme B, perforin, and cyclophilin B mRNA,
sis and therapeutic decisions.78 This is further complicated the products of which are cytotoxic proteins implicated
by coexistence of both (mixed rejection) and/or coexistence in animal and patient studies of allograft rejection, for
with other pathologies. evaluation in urinary cell pellets from patients with
Overall, there is a lack of consistency in the identifica- biopsy-proven allograft rejection.80 Granzyme B and per-
tion of candidate biomarkers across individual published forin mRNA expression were shown to be increased in
studies. The reasons for this are multiple small sample sizes; acute rejection. Using a similar targeted approach, the
heterogeneity of patient demographics; variability of the same group showed subsequently the increased expres-
transcriptional milieu of acute rejection in the allograft, sion of a number of other immune products known to be
urine, and/or peripheral blood, which is further influenced relevant to the pathophysiology of acute rejection. These
by immunosuppressive agents; acute rejection type or sub- were selected for further evaluation in the prospective,
type and severity (further complicated by the presence of observational multicenter CTOT-4 study, one of projects
mixed rejection); the timing of the rejection episode; the conducted by the National Institutes of Health (NIH)
type/quality of the allograft; concurrent pathology (e.g., BK -sponsored Collaborative Trials in Organ Transplanta-
virus, IFTA, ATN); and the representativeness of the biopsy tion study consortium (CTOT).81 In the CTOT-4 study,
sample used for transcriptome analysis. Furthermore, the the investigators assessed whether urinary cell levels
lack of validation in large prospective, independent cohorts of mRNA encoding CD3e chain, perforin, granzyme B,
is where a number of noninvasive candidate biomarkers proteinase inhibitor 9, CD103 (a cell surface marker
falter and are delayed from proceeding toward potential expressed on alloreactive CD8+ T lymphocytes), inter-
clinic application. However substantial research has been feron (IFN) -inducible protein 10 (IP-10, also known as
undertaken and a number of promising approaches have CXCL10), and the chemokine receptor CXCR3 obtained
been identified (see detailed review79). This chapter will dis- at time of biopsy: (1) is diagnostic of acute rejection, and
cuss the more promising noninvasive biomarkers of acute (2) whether changing expression levels in sequential
rejection in renal transplantation. The larger multicenter urine specimens could predict the subsequent develop-
studies and their proposed biomarker candidates have ment of acute rejection. In this study 4300 urine speci-
been tabulated in Table 26.4. It should be highlighted that mens were collected from 485 renal transplant recipients
some of these studies adopted a targeted approach where at timed intervals and at clinically indicated biopsies. The
molecules of interest were preselected for assessment of combined measure of CD3e, IP-10, and 18s ribosomal
their biomarker potential, whereas others adopted unbi- RNA as a three-gene signature to discriminate ACR from
ased techniques to discover candidate molecules before no rejection yielded an ROC curve area under the curve
proceeding with targeted validation of a more refined set of (AUC) of 0.85 in the primary data set and 0.74 in an
molecules. external validation set. The gene set was less effective at
differentiating ACR from borderline changes (AUC 0.78)
and AMR. Subsequently, a urinary six-gene signature
TABLE 26.4 Summary of Multicenter Studies Evaluating (that was derived from measuring absolute levels of 26
Potential Biomarkers of Acute Rejection prespecified mRNAs using qPCR) from a single-center
study was shown to distinguish acute rejection from
Study Biofluid Biomarker
other causes of renal allograft dysfunction, and could
Suthanthiran Urine (mRNA) Combined 3-gene further distinguish between ACR and AMR.82
et al., CTOT-481 signature: The evaluation of urinary proteins associated with acute
CD3ε, IP-10(CXCL10),
and 18s ribosomal
rejection has also received much attention over the past
RNA decade, with CXCL10 and CXCL9 (monokines induced by
Hricik et al., Urine (protein) CXCL9, CXCL10 IFN-γ) showing the greatest promise as potential biomark-
CTOT-186 ers to date. Both these chemokines are secreted by infiltrat-
Li et al.97 a Whole blood (mRNA) DUSP1, NKTR, MAPK9, ing inflammatory cells and renal tubular and endothelial
PSEN1, PBEF1
Roedder et al., Whole blood (mRNA) kSORT (17-gene set) cells.83 Using an antibody array consisting of 120 che-
AART99 mokines and cytokines to screen the urine of recipients
Kurian et al.98 PBMCs (mRNA) 200-gene setb with allograft injury compared with those with stable
aDiscovery
function and healthy controls, with further confirma-
cohort was from single center, with external validation in a multi-
center cohort.
tion using multiple-bead array, CXCL9 and CXCL10 were
bTruGraf assay from Transplant Genomics Inc. is based on this gene set and significantly elevated in acute renal injury.84 A number
undergoing validation at the Mayo Clinic. of other groups adopted a more targeted approach using
424 Kidney Transplantation: Principles and Practice

ELISA to evaluate cytokines and tubular injury markers, approaches, which in part is because of the heightened
which supported the potential of CXCL9 and/or CXCL10 challenge posed by the large dynamic range in protein con-
as biomarkers in acute rejection (both clinical and sub- centrations in blood.91 Most studies evaluating blood as a
clinical), but, as they are elevated in other inflamma- potential biomarker source have reported on the molecu-
tory states, they may not be specific for rejection.85 The lar (mRNA) transcripts in whole blood or peripheral blood
CTOT-01 study, comprising 280 adult and pediatric first mononuclear cells (PBMCs). As was the case with evalua-
transplant recipients, evaluated a number of potential bio- tion in urine, much interest was shown in perforin and gran-
markers including CCR1, CR5, CXCR3, CCL5 (RANTES), zyme B mRNA expression in the PBMCs of acute rejection
CXCL9, and CXCL10 using ELISA.86 Importantly, whereas patients with a number of studies, albeit with small sample
the PPV for acute rejection was not high, CXCL9 urinary sizes, supporting their use as biomarkers.92,93 However, a
protein yielded a NPV of 92% (urinary CXCL9 mRNA recent a meta-analysis was conducted and suggested that
NPV was 83%), and low urinary CXCL9 urinary protein neither molecule, if evaluated alone, be it in urine, periph-
levels 6 months posttransplant identified patients unlikely eral blood lymphocytes or allograft tissue was convincing
to develop an acute rejection episode or a 30% decline in as a noninvasive biomarker of acute rejection.94
their eGFR between 6 and 24 months (92.5%–99.3%). A Along with the evolution and widespread applica-
more recent prospective, longitudinal single-center study, tion of omic technologies, particularly DNA microarray
comprising 300 kidney transplant recipients, evaluated the platforms, paralleled by advances in the bioinformatic
predictive potential of CXCL9 and CXCL10 for a subsequent analytical methods to handle the large body of data gen-
acute rejection episode.87 Early urinary CXCL10:creatinine erated, there was increasing recognition that the immune
levels predicted clinical and subclinical rejection and both response in acute rejection is complex. Hence, the expres-
ACR and AMR. sion of a combination/set of molecules, a “signature,”
Although requiring validation in large prospective rather than a single molecule, could improve the diag-
longitudinal cohorts, untargeted protein expression pro- nostic accuracy.95 Promising data have come from sin-
filing studies have also uncovered potential candidate gle-center studies showing the potential of untargeted
biomarkers both in urine and blood.79 Sidgel, Sarwal, and approaches (such as DNA microarray) to identify potential
colleagues have used high-throughput proteomic and biomarkers in acute rejection from peripheral blood.96 In
peptidomic techniques for both untargeted and targeted a relatively large pediatric study comprising 367 periph-
potential biomarker discovery in acute rejection.37,88,89 eral blood samples with paired allograft histology, Li and
In one of their larger studies, isobaric tags for relative colleagues97 used DNA microarray (across three different
and absolute quantitation (iTRAQ)-based proteomic dis- platforms, 103 unique peripheral samples in the discovery
covery and targeted ELISA validation were used in 262 set) at a single center to identify differentially expressed
renal allograft recipient urine samples from a single-center genes in acute rejection. From this, a five-gene signature
pediatric and young adult cohort (split into a training and was derived (DUSP1, PBEF1, PSEN1, MAPK9, and NKTR)
independent validation set) with biopsy-confirmed acute that classified acute rejection with high accuracy (PPV
rejection, chronic allograft injury, BK virus nephritis, sta- 83%, NPV 97%, and ROC AUC 0.955 for distinguishing
ble grafts, and nephrotic syndrome, and healthy normal acute rejection from stable grafts). These genes are mostly
controls.31 In a more recent study, the same group applied expressed by activated monocytes in the peripheral circu-
state-of-the-art MS methods (either iTRAQ-based or label- lation, reflecting injury mechanisms relating to oxidative
free LC-MS) across 264 renal allograft biopsy-matched cellular stress responses (DUSP1), apoptosis (MAPK9), IL2-
urine samples.90 Centralized and blinded histology data of dependent activation of cytolytic genes (NKTR), increased
the biopsies were used to classify urine samples into acute cell adhesion via the e-cadherin/catenin complex (PSEN1),
rejection, chronic allograft nephropathy, BK virus nephritis, and vascular smooth muscle injury (PBEF1).97 The authors
and stable grafts. The study identified 958 unique urinary performed external validation of this gene set in an indepen-
proteins, from which a set of 131 peptides was selected, and dent multicenter cohort. The subtypes of rejection were
targeted quantification using the selected reaction moni- not differentiated in the acute rejection cases, and given it
toring (SRM) assay was performed in an independent vali- is a pediatric cohort the majority of the transplants were
dation cohort. Ultimately a set of 35 proteins identified for from living donors. These studies were followed by larger
their ability to distinguish three major clinical phenotypes multicenter collaborations.98,99 The 200-gene TruGraf
(acute rejection, chronic allograft nephropathy, and BK panel, which is currently undergoing validation in a pro-
virus), with 11 of these urinary peptides for acute rejection spective cohort of 300 renal transplant patients, was devel-
yielding an ROC AUC of 0.93. As it stands, the high cost oped from a study of more than 1000 kidney transplant
and complexities of the advanced proteomic technologies recipients who were followed prospectively to identify 148
have limited its more widespread adoption and use in pro- cases (divided into a test and validation set) comprising 46
tein biomarker discovery and validation opportunities. with normal graft histology, 63 with biopsy-proven acute
rejection, and 39 with acute kidney dysfunction with-
out histologic evidence of acute rejection.98 Global gene
Potential Peripheral Blood expression profiling was performed using DNA microarray
Biomarkers of Acute Rejection on peripheral blood obtained at the time of biopsy. Mul-
tiple three-way classified tools identified the 200 highest
Unlike urine, there are fewer high-quality published stud- value probe sets with high diagnostic accuracy. However,
ies evaluating potential serum/plasma protein biomarkers as the authors acknowledged this was not a prospective
in acute rejection, particularly with untargeted proteomic study and there were no cases of pure AMR for evaluation
26 • Biomarkers of Kidney Injury and Rejection 425

in their cohort. In the Assessment of Acute Rejection in


Renal Transplantation (AART study), 558 blood samples
Biomarkers of Chronic Allograft
from 436 adult and pediatric kidney allograft recipients Injury
were included, comprising blood samples collected con-
currently with indication and for-cause biopsies (thus Despite the significant improvement in acute rejection
capturing both clinical and subclinical rejection in the rates over the past two decades, this has not translated
acute rejection cases).99 The authors preselected 43 genes into improved long-term graft survival. Chronic allograft
(based on their published original profiling studies using injury (CAI) remains a major cause of long-term graft loss
microarray, and other published work) for mRNA evalua- and represents an irreversible pattern of injury resulting
tion by microfluidic qPCR in 143 case-control samples and from a number of immunologic (acute and chronic rejec-
a 17-gene set (kSORT) was derived to discriminate acute tion, human leukocyte antigen [HLA] mismatch, DSAs),
rejection from no acute rejection (AUC 0.94, 95% confi- and nonimmunologic factors (donor age, ischemia/reper-
dence interval [CI] 0.91–0.98), and validated in several fusion injury, infection, calcineurin inhibitor (CNI) tox-
ways. Although the results are rather promising, there icity).105,106 There are differing opinions as to whether
was no discrimination between acute cellular and AMR, allograft loss is multifactorial, because some recent obser-
and a biopsy for accurate diagnosis would still be required. vational studies suggest that most cases of allograft loss
In addition, prospective validation is required to evalu- can be attributed to a single disease entity,107,108 whereas
ate its diagnostic accuracy. More recently, the authors other published findings suggest otherwise.109 Interstitial
evaluated the predictive capacity of kSORT and the IFN-γ fibrosis and tubular atrophy (IFTA) are the histopatho-
enzyme-linked immunosorbent assay (ELISPOT, discussed logic hallmarks of CAI. It has subsequently been recog-
further later) both alone or in combination to predict sub- nized that concurrent interstitial inflammation and IFTA
clinical (sc) rejection at 6 months (confirmed on protocol changes on biopsy identify recipients at greatest risk of
biopsy) posttransplant in 75 consecutive kidney trans- graft loss.110,111 Despite greater understanding of the fac-
plant recipients from the ESCAPE study.100 There were tors contributing to CAI/IFTA, the precise pathophysio-
promising data of the predictive potential when both the logic mechanisms are not fully understood. Consequently,
molecular and immune cell functional assays were com- the development of targeted therapeutic strategies has
bined to predict sc-acute rejection, scACR or scAMR, but been limited. As with subclinical rejection, the presence
the cohort size was small (17 and 5 cases of scACR and of IFTA—a histologic diagnosis—predates the measured
scAMR respectively). decline in renal function. The need for earlier detection
As in the aforementioned study, functional cell-based to allow timely therapeutic intervention has provided the
immune monitoring to evaluate circulating antidonor T cell impetus for numerous genome, transcriptome, and pro-
alloreactivity using ELISPOT has garnered interest and has teomic studies in the search for potential noninvasive,
been used increasingly in transplant research. Although predictive biomarkers in CAI.
previous single-center studies suggested its potential for The concept that early transcript changes may predate
predicting acute rejection, a more recent multicenter study the subsequent histologic phenotype of CAI has been an
from the CTOT consortium found no correlation with the appealing hypothesis.74 Using oligonucleotide microarray,
incidence of acute rejection.101 Scherer et al.112 identified differential transcript expres-
Although the focus on this chapter has been on nonin- sion between progressors and nonprogressors, with genes
vasive biomarkers of acute rejection, it is important to rec- pertinent to T and B cell activation, the immune response,
ognize the extensive research done in invasive molecular profibrotic processes, and epithelial-to-mesenchymal
markers and their role in disease reclassification and/or transition (EMT) featuring prominently in the progres-
refining the exact diagnosis in acute rejection.74 As men- sor group. In contrast, a larger study of a retrospectively
tioned previously, the allograft biopsy remains the gold selected group of 107 allograft recipients, who had their
standard for diagnosis of acute rejection (both clinical and biopsy microarray data summarized into pathogenesis-
subclinical). However, sampling error or tissue insufficiency, based transcript sets, found that the molecular pheno-
and interobserver variability in reporting have highlighted type in early (6 weeks after transplantation) transcript
the role for molecular profiles in allograft tissue to aid in the expression mainly reflected the injury-repair responses
diagnosis of ACR and AMR. Halloran and colleagues have to implantation stresses, without strong correlation with
done extensive work in recent years in this area and shown 6-month IFTA or renal function at 24 months.113 In an
that renal biopsies of AMR demonstrate transcript changes attempt to overcome these design issues, the multicenter
relating to endothelial injury, IFN-γ effects, and natural Genomics of Chronic Allograft Rejection (GoCAR) study
killer cells.102,103 In response to these data, the Banff work- prospectively recruited a large cohort of patients from five
ing group has added “increased expression of gene tran- centers. The study identified a predictive gene set capable
scripts in the biopsy tissue indicative of endothelial injury, if of classifying renal allografts at risk for progressive injury
thoroughly validated” as an AMR diagnostic criteria under because of fibrosis. Using differential gene expression to
evidence of antibody-vascular endothelium interaction.104 predict the development of chronic graft injury3 biopsies
From Halloran and colleagues, the Molecular Microscope were obtained prospectively 3 months after transplanta-
Diagnostic system (MMDx-kidney) has been devised and tion from renal allograft recipients (n = 159) with stable
assesses mRNA expression in each specimen compared renal function and were analyzed for gene expression by
with a reference set of known specimens and generates an microarray. Genes were sought that correlated with subse-
automated report.103 Prospective trials using this system quent 12-month Chronic Allograft Damage Index (CADI)
are awaited. score but not fibrosis at the time of the biopsy. Using
426 Kidney Transplantation: Principles and Practice

bioinformatics and machine learning, an optimal and min-


imal gene set for prediction of future kidney fibrosis was
Anti-HLA Antibodies as
identified. This 13-gene set was independently predictive a Biomarker of Graft Loss
for the development of fibrosis at 1 year.3 The high pre-
dictive capacity of the gene set (AUC 0.967) was superior Since the development of the complement dependent cyto-
to clinical indicators and also accurately predicted early toxicity assay, the presence of antidonor HLA antibodies
allograft loss (AUC 0.842 and 0.844 at 2 and 3 years, has been known to be a cause of hyperacute rejection and
respectively). The predictive value of this gene set was vali- graft loss.115 With refinements in our understanding of the
dated using an independent GoCAR cohort (n = 45, AUC pathogenesis of antibody-mediated graft injury and the
0.866) and two independent, publicly available, expres- emergence of solid-phase assays to detect anti-HLA anti-
sion data sets. Although not designed to identify causality bodies in the serum of patients our understanding of the
or pathogenesis, there were some clues regarding causality role of DSAs and their effect on graft injury and graft loss
because the transcripts specifically associated with CADI-3 has changed considerably.116 DSAs may be present before
were related to alloimmunity, including T cell activation, the transplant (preformed) or develop de novo after trans-
whereas genes involved in programmed cell death/apopto- plantation. Preformed antibodies are associated with early
sis and cell adhesion were associated with CADI-12 alone. acute and severe rejection, including hyperacute rejec-
This suggests that both recipient and donor graft factors tion, accelerated rejection and acute AMR. In some cases
were involved in the pathogenesis of CAI. Immune cell gene the presence of preformed antibodies may have a benign
enrichment analysis revealed that stromal cell (mostly course with little or no evidence of graft injury.117 There is
fibroblast) genes were the most significantly associated evidence to suggest that their pathogenicity may be a reflec-
with CADI-12 in addition to macrophage, dendritic, and tion of their antibody subclass, specificity, strength, and/
CD4+ T cells.3 Although the exact roles of the 13 predic- or complement-binding capacity. However, our ability to
tive genes and the associated molecular pathways remain predict their effect on the transplant outcome is imperfect
to be determined, there was a significant overrepresenta- at best. The development of de novo DSA is also an impor-
tion of genes involved in cell growth and tumor develop- tant predictor of graft outcome but its effect is different from
ment/suppression including MET (MET proto-oncogene), that of preformed DSA. The development of de novo DSA
ST5 (suppression of tumorigenicity 5), and KAAG1 (kid- (dnDSA) is associated with several clinical scenarios includ-
ney associated antigen 1); genes that are involved in ubiq- ing (1) inadequate immunosuppression either from doctor-
uitination either as E3 ligases or as interacting proteins, initiated minimization or patient noncompliance; (2) a high
including RNF149, ASB15, and KLH13; genes involved number of HLA mismatches, and epitope mismatches, espe-
in significant developmental or growth pathways such as cially to MHC class II antigens; and (3) factors that increase
the NOTCH/Wnt pathway and the RAR pathway through graft immunogenicity such as inflammation from prior
SMAD, including the genes TGIF1 (TGFB-induced fac- nonantibody-mediated rejection, infection, or ischemia.
tor homeobox 1), SPRY4 (sprouty homolog 4), WNT9A, Development of dnDSA can occur any time after transplant
RXRA, and FJX1; and genes involved in energy and mem- and is found in 15% to 30% of transplant recipients.118
brane repair, such as the mitochondrial gene CHCHD10 The proportion that develop dnDSA in the first year varies
and SERINC5, which phosphorylates membrane proteins. widely (2%–20%) depending on the cohort studied, and the
Whereas this gene set shows potential as a prognostic bio- ongoing prevalence is between 2% and 5% per year.119,120
marker for chronic graft injury, this needs to be confirmed Although dnDSA can cause acute AMR, their predominant
in a randomized control trial where the gene set is used as outcome is subclinical rejection, chronic AMR and ulti-
a tool for stratifying patients according to risk into differ- mately glomerulosclerosis and chronic graft loss. Several
ent treatment arms. Such analysis would help confirm its studies have shown that development of dnDSA is predic-
utility in clinical decision making. tive of ultimate graft loss. Wiebe et al. have shown that the
There are very few studies that have used a genomic mean time to graft failure from first detection of dnDSA in
approach to specifically develop an assay that is predictive patients where there is deterioration of graft function at
of IFTA. One such study profiled urine cell mRNA expres- time of detection is 3.3 years and 8.3 years in cases where
sion in 114 patients with allograft fibrosis, identified on dnDSA is detected in stable functioning grafts.120 This has
indication or protocol biopsy.114 The samples were divided been confirmed by several other studies where subclini-
before analysis at a 2:1 ratio into a discovery (76 patients, cal AMR was associated with increased graft loss within
32 with allograft fibrosis and 44 with normal histology) the first 8 years after transplantation.121 IgG subclass and
and validation sets (38 patients, 16 with fibrosis and 22 the presence or absence of complement binding are also
normal histology). A four-gene set comprising vimen- strong predictors of graft loss. Currently C1q-binding anti-
tin, NKCC2, E-cadherin, and 18S rRNA was developed, bodies are the best studied. There are several studies that
and diagnosis of fibrosis in the validation set was highly have shown that the presence of C1q-binding antibodies is
accurate, with an AUC of 0.89 (P < 0.0001). In the inde- associated with a greater risk of AMR, deterioration in graft
pendent validation set, this gene signature predicted the function, and graft loss. In a large French cohort study C1q-
presence of fibrosis with 77.3% sensitivity and 87.5% binding DSA was found to be an independent risk factor for
specificity. These results require confirmation in a more graft loss with a hazard ratio of 4.5 (95% CI, 2.2–9.0).122
uniform study protocol. Furthermore, whether this profile There have been a large number of retrospective and pro-
is predictive rather than diagnostic of fibrosis remains to spective studies that have confirmed the association between
be determined. complement-binding antibodies and graft loss.122–130 More
26 • Biomarkers of Kidney Injury and Rejection 427

recently there is evidence that C3d- and C4d-binding DSA in kidney transplantation. With the development of single
are better predictors of graft loss than C1q-binding antibod- antigen bead and other solid state assays for the detec-
ies.126,129 A large retrospective study by Sicard et al. showed tion of anti-HLA antibodies, there is now a reliable and
that the presence of C3d-binding antibodies was highly pre- highly specific test for the detection of DSA (both pre-
dictive of graft loss, whereas the presence of C1q-binding formed and de novo). Numerous large retrospective and
antibodies was less so.126 These findings were confirmed by prospective cohort studies have demonstrated that DSA
a small but prospectively designed study in pediatric kidney is predictive of loss of function and graft loss.118 The asso-
transplant recipients where 34 of 119 patients developed de ciation between the presence of DSA and graft damage is
novo DSA (25 were C1q binding and 9 C3d binding). This biologically plausible. Antibody binding to HLA expressed
study showed that the presence of C3d-binding DSA was on the renal transplant endothelium has been shown in
predictive of graft loss but C1q-binding antibodies was less experimental and clinical studies to activate the classical
so.129 complement pathway, which is known to be a key com-
Theoretically, the presence of C1q- and C3d-binding ponent of AMR. Besides complement-mediated damage,
antibodies and their association with graft loss would antibodies can damage graft endothelial cells via antibody-
seem to make sense from a pathogenesis perspective. Com- mediated cell cytotoxicity. In addition, neutrophils, macro-
plement activation is a well-studied cause of tissue injury phages, and natural killer cells have Fc receptors that bind
and the detection of complement-binding antidonor DSA fragments, triggering degranulation, cell activation,
antibodies by the less sensitive complement-dependent and the release of lytic enzymes causing cell lysis and tis-
cytotoxicity assay is a predictor of hyperacute rejection. sue damage. There have been numerous clinical studies
Furthermore, C1q production occurs in the first step of demonstrating a strong correlation between macrophage
the classical complement pathway whereas production accumulation within the graft and the degree of fibrosis
of C3d is indicative of complete complement activation, and tissue damage.135–139 More importantly, the presence
suggesting a stronger antibody response. However, cau- of DSA is associated with a chronic AMR where peritubu-
tion should be exercised in accepting such a simple par- lar capillary multilayering, intimal fibrosis, and sclerotic
adigm, because interpretation of these findings may be intima are seen in the context of increased thickness of the
more complex. First, the human antibody response to for- vascular intima, smooth muscle cell invasion, and trans-
eign antigens is polyclonal with multiple IgG subclasses plant glomerulopathy.140,141
with different capacities for complement binding. Second, In summary, DSAs are the most used biomarker of graft
other factors such as antibody affinity for specific epitopes injury and graft loss in clinical transplantation. There are
will also affect the strength of the antibody response and numerous retrospective and prospective cohort studies that
in theory the resulting level of graft damage. Other stud- have confirmed the association between preformed and de
ies have attempted to link IgG subclass dominance with novo DSA and graft loss. There is clinical and experimen-
the type of graft injury to identify predictive tests that tal evidence to suggest that the binding of DSA to vascu-
may be more informative clinically.131 Although IgG1 is lar endothelium leads to the pathologic changes seen in
the predominant antibody subclass and is C1q binding, chronic AMR and transplant vasculopathy. A very impor-
acute AMR was associated with IgG3 as the immuno- tant step in its uptake as a biomarker is the presence of a
dominant subclass, and subclinical AMR was associated reliable and reproducible assay in the form of the single
predominantly with the presence of IgG4 subclass DSA. antigen bead or Luminex assay. Currently many transplant
Although this could be useful to stratify prediction, the centers use it to screen patients to identify those at risk of
finding is not universal, and other studies have shown chronic AMR and accelerated graft loss. In addition, devel-
that IgG-subclass stratification did not predict patients opment of do novo DSA has potential as SEP for clinical tri-
who went on to develop AMR.132 The reason for the poor als, but further work is required to improve its utility and
correlation was that the majority of patients had a mix- reliability, and a more reliable test to evaluate the strength
ture of IgG subclasses, composed of both strong and weak and pathogenicity of DSA is required. Whereas assays of
complement-activating subclasses, which did not facili- C1q, C3d, and C4d binding have been developed, their prog-
tate the stratification of patients according to risk. nostic value has been controversial,129,130,142 and other
Perhaps a better way to consider this is to view comple- assays of antibody titer may ultimately be more reliable.141
ment binding as a marker of the strength of the antibody More importantly, there have been no effective treatments
response. It is important to remember that the mean for chronic AMR, which is essential to demonstrate that
fluorescent intensity (MFI) reported with the Luminex improvements in function are associated with reductions
assay has poor reproducibility between centers, and it is in DSA titer. To date, the use of DSAs as a biomarker have
not licensed as a quantitative assay. On the other hand, not been evaluated in randomized clinical trials. The use of
C1q binding on the single antigen bead assay correlates costimulation blockade with belatacept-based immunosup-
strongly with the density of HLA antibody on the bead sug- pression has been shown to result in reduced development
gesting that high combined titers of DSA will have a high of de novo DSA compared with cyclosporine-based therapy,
prevalence of C1q binding.133 When single antigen bead and resulted in better long-term graft survival143,144 Evi-
MFI was compared with C1q binding and antibody titra- dence of benefit from other therapies relies on case studies
tion studies, the titration studies were found to be the more only. Rituximab has been used in desensitization protocols,
predictive.134 but a systematic review found little evidence to support
Despite its shortcomings, the measurement of DSA is its use and the limited publication of well-designed clini-
probably the most advanced of the prognostic biomarkers cal trials precluded a formal meta-analysis of the available
428 Kidney Transplantation: Principles and Practice

evidence.145,146 Tocilizumab, a blocking monoclonal anti- sensitivity and 83% specificity in a test set of 12 patients
body of the IL-6R has shown efficacy at reducing HLA titers, divided equally into the two groups. The expansion of B
and facilitated transplantation in highly sensitized patients, cell transcripts was not only identified in the blood, but
with potential as a treatment for chronic AMR.147,148 As a also in the urine sediment, which exhibited upregulation
result, formal evaluation of this therapy in a randomized of CD20 mRNA in the tolerant group. Using a different
clinical trial would be expected. Although identification of microarray platform, Sagoo and colleagues were able to
DSA is a useful biomarker for stratifying patients according distinguish operationally tolerant recipients from chronic
to risk, its development as a biomarker would benefit from rejection, stable patients on immunosuppression, and nor-
a prospective analysis in clearly defined patient populations mal controls.152 Again, a predominance of B cell related
and in randomized clinical trials where its utility as a SEP genes and associated molecular pathways were identified
can be clearly defined. in their tolerance footprint. However, it is possible that the
strong representation of B cells in the aforementioned stud-
ies may reflect the absence of immunosuppression, rather
Development of Biomarkers for than the state of tolerance per se. Newell was unable to
Studies of Tolerance identify any of the three genes in their predictive set from
Sagoo’s platform, questioning the robustness of the find-
With several groups developing protocols for immune toler- ings.151 More recently, Baron and colleagues153 compared
ance, a biomarker that predicts this and allows immunosup- the gene lists derived from the five tolerance-related blood
pression to be withdrawn safely is essential. The first study transcriptomic studies, and were unable to identify a gene
published on gene expression profiling in operationally tol- signature to differentiate tolerant subjects from stable con-
erant renal graft recipients using microarray came from trols on immunosuppression.149,151,152,154,155 To enhance
Brouard and colleagues.149 A subset of patients in their study the study power, the microarray data from the 96 opera-
was stratified into three clinical phenotypes: tolerant patients tionally tolerant patients were integrated into one data set.
without immunosuppression for at least 2 years, chronic The authors made efforts to overcome systematic biases
rejection, and age-matched controls. Using a customized derived from merging microarray data obtained from dif-
cDNA microarray platform (Lymphochip), a “tolerant foot- ferent platforms by renormalization and standardization
print” of 49 genes was identified from PBMCs. Thirty-three of the data before integration, and only the 1846 consen-
of these differentiated operationally tolerant from chronic sus genes were retained for analysis. Their meta-analysis
rejection with 99% specificity and 86% sensitivity. There identified a robust differential gene signature compared
was underexpression of costimulatory genes and Th1/Th2- with subjects who were stable on immunosuppression. The
related cytokines in the tolerant group. In addition, 27% of most discriminating gene clusters were linked to B cells,
these genes were regulated by transforming growth factor-β monocytes, and CD4 T cells at various stages of differentia-
(TGF-β). In a later study from the same group, 20 of these tion. Following cross validation, a selection of the 20 top-
genes were selected and formed a gene set, which the authors ranked genes with the majority overexpressed in tolerance
proposed could potentially identify those patients at “mini- and pertaining to B cells was revalidated in an independent
mal risk” of rejection, where reduction of immunosuppres- cohort of 18 new tolerance samples of which 6 were new
sion could be considered.150 In a cohort of 144 patients who cases, with 91.7% accuracy (94.4% sensitivity, 90% speci-
were at least 5 years posttransplant with stable graft func- ficity, 85% PPV, and 96.4% NPV). This meta-analysis reaf-
tion, 3.5% displayed a tolerance profile in the peripheral firms the prominent role of B cells in the tolerant state as
blood based on this 20-gene set. However, these findings identified in earlier studies, but whether their strong pres-
were not obtained in parallel with kidney biopsies to verify ence is causative or consequent remains unanswered.153
the absence of immune reactivity; to date no study has been
published that tests their hypothesis.
The bias toward B cells in the tolerant state was also iden- The Use of Biomarkers as
tified by multicenter studies by Newell and colleagues151
and Sagoo and colleagues.152 The patients in the tolerance
an Enrichment Strategy for
cohort were from the US Immune Tolerance Network (25 Randomized Trials of
patients) and the European Indices of Tolerance Consor- Immunosuppressive Therapy in
tium (11 patients). The two studies used samples from each
other’s cohort for validation of their findings. Newell and Transplantation
colleagues used microarray for whole blood gene profiling
and found 30 genes overexpressed by at least twofold in Observational studies may provide useful predictive infor-
19 operationally tolerant patients (more than 12 months mation of a biomarker on health outcomes, such as the
off immunosuppression) versus patients with stable graft risk of renal allograft fibrosis, acute rejection, and graft
function with ongoing triple immunosuppression. Twenty- loss, but a well-designed randomized clinical trial is
two of these genes were B cell specific. However, this gene needed to estimate the health effect of a biomarker in the
expression profile could not differentiate between tolerant clinical setting. Random assignment of the biomarker in a
patients and healthy (nontransplanted) controls. Further randomized control trial assures comparability of groups,
analysis identified a three-gene signature that was able and the true unconfounded effect of the strategy can be
to distinguish operationally tolerant patients from those estimated by between-groups comparison of health out-
with stable function on immunosuppression with 100% comes. This is desperately needed in transplantation
26 • Biomarkers of Kidney Injury and Rejection 429

Biomarker Enrichment Clinical Trial

R
Enrichment Design a Treatment A (SOC)
n
d
o

Transplant
Assess +VE m

Surrogate End Point


Biomarker i

Hard Endpoint
z
e Treatment B
R
a Treatment A (SOC)
n
–VE d
o
m
i
z
e Treatment C

Fig. 26.2 Outline of potential clinical trial where biomarkers are used to stratify patients according to risk and enrich for patients based on a biomarker
validated for a particular clinical phenotype. (Adapted from Freidlin B, McShane LM, Korn EL. Randomized clinical trials with biomarkers: design issues. J Natl
Cancer Inst 2010;102(3):152–60.)

because, traditionally, new immunosuppressive agents the “biomarker” arm, novel immunosuppressive regimens
are evaluated in low- or standard-risk patients, and patient will be assigned to participants of different risk profiles,
and graft survival are assessed at 12 or 24 months. How- tailored according to the risk stratification that has been
ever, short-term graft survival is excellent in this cohort identified by the biomarker.
of patients with 1-year graft survival for deceased donor
grafts in Australia and most other countries at 94% and
patient survival at 97% (ANZDATA report 2016).156 Conclusion
This means that achieving superior 1-year outcomes in
this cohort of patients is not feasible. What is required The development of new technologies to investigate the
is the redesign of clinical trials to those that are focused transcriptome, proteome, and metabolome has opened
on improving long-term outcomes and patient-reported up new possibilities in biomarker development. Although
outcomes.1,9 To achieve this, we need to transition to this field is currently in its infancy it has the potential to
individualized therapy based on the patients’ clinical and improve the clinical management of transplant patients
genetic profiles. Recent advances in biomarker develop- by predicting individual patient’s risk profile in the early
ment have the potential to drive personalized medicine transplant period when interventions are more likely to
by enabling individualized risk stratification and targeted be successful. Not only do these new biomarkers have the
immunosuppression through identification of biomark- potential to be predictive of outcome, but they also have
ers associated with immune-mediated complications after the potential to stratify patients, according to phenotype,
transplantation. Traditional randomized clinical trials, into more individualized therapies, and be used to guide
which focus on the average treatment effect in the over- patient recruitment for clinical trials using enrichment
all transplant population, are suboptimal for evaluation strategies described earlier. What is required going for-
of these targeted treatments. Efficient development and ward is to validate biomarkers in prospectively recruited,
use of new immunosuppressive drugs requires clinical discrete patient populations that have been followed lon-
trials that are driven by predictive biomarkers to enable gitudinally over time. This will require international mul-
both evaluation of new treatments and identification of ticenter collaboration to allow for large-scale, prospective
the patient subgroups in which these treatments are indi- studies, with better study design and clinical and his-
cated. Such trials would entail a biomarker stratification/ tologic evaluation at defined time points. Only then can
enrichment design as outlined in Fig. 26.2 and has been such discoveries transition from the bench to potentially
undertaken for several new chemotherapeutic agents in become diagnostic and prognostic clinical tools that lead
oncology.157,158 These biomarkers are expected to play an to improved graft longevity and patient survival.
important role in minimizing risk of clinical trial failure
by enriching the trial populations with specific molecular Acknowledgments
subtypes that would respond better to tested therapies. In Original work by the authors was funded by the NIH
this setting, the role of the biomarker is to identify a sub- (grant 1U01AI070107) and National Health and Medi-
set of kidney transplant recipients who will be randomized cal Research Council of Australia. Karen Keung is a
to a strategy in which a biomarker is measured compared recipient of the Jacquot Research Entry Scholarship
with a strategy in which the biomarker is not measured from the Royal Australasian College of Physicians and
(see Fig. 26.2). Patients who are randomized to the “no the Australian and New Zealand Society of Nephrology.
biomarker” arm will receive standard of care immunosup- Philip O’Connell is a recipient of an NHMRC Practitioner
pressive therapies. Among those that are randomized to Fellowship.
430 Kidney Transplantation: Principles and Practice

References 24. Oghumu S, BA, Nori U, et al. Acute pyelonephritis in renal allografts:
a new role for microRNAs? Transplantation 2014;97(5):559–68.
1. O’Connell PJ, Kuypers DR, Mannon RB, et al. Clinical trials for immu- 25. Quintana LF, Sole-Gonzalez A, Kalko SG, et al. Urine proteomics
nosuppression in transplantation: the case for reform and change in to detect biomarkers for chronic allograft dysfunction. J Am Soc
direction. Transplantation 2017;101(7):1527–34. Nephrol 2009;20(2):428–35.
2. Buyse M, Sargent DJ, Grothey A, Matheson A, de Gramont A. Bio- 26. Srivastava M, Eidelman O, Torosyan Y, Jozwik C, Mannon RB, Pol-
markers and surrogate end points—the challenge of statistical vali- lard HB. Elevated expression levels of ANXA11, integrins beta3 and
dation. Nat Rev Clin Oncol 2010;7(6):309–17. alpha3, and TNF-alpha contribute to a candidate proteomic signa-
3. O’Connell PJ, Zhang W, Menon MC, et al. Biopsy transcrip- ture in urine for kidney allograft rejection. Proteomics Clin Appl
tome expression profiling to identify kidney transplants at risk 2011;5(5-6):311–21.
of chronic injury: a multicentre, prospective study. Lancet 27. Sigdel TK, Kaushal A, Gritsenko M, et al. Shotgun proteomics identi-
2016;388(10048):983–93. fies proteins specific for acute renal transplant rejection. Proteomics
4. Biomarkers Definitions Working Group. Biomarkers and surrogate Clin Appl 2010;4(1):32–47.
endpoints: preferred definitions and conceptual framework. Clin 28. Banon-Maneus E, Diekmann F, Carrascal M, et al. Two-dimensional
Pharmacol Ther 2001;69(3):89–95. difference gel electrophoresis urinary proteomic profile in the search
5. Meier-Kriesche HU, Schold JD, Srinivas TR, Kaplan B. Lack of of nonimmune chronic allograft dysfunction biomarkers. Trans-
improvement in renal allograft survival despite a marked decrease plantation 2010;89(5):548–58.
in acute rejection rates over the most recent era. Am J Transplant 29. Wu D, Zhu D, Xu M, et al. Analysis of transcriptional factors and
2004;4(3):378–83. regulation networks in patients with acute renal allograft rejection.
6. Kasiske BL, Chakkera HA, Louis TA, Ma JZ. A meta-analysis of J Proteome Res 2011;10(1):175–81.
immunosuppression withdrawal trials in renal transplantation. 30. Loftheim H, Midtvedt K, Hartmann A, et al. Urinary proteomic
J Am Soc Nephrol 2000;11(10):1910–7. shotgun approach for identification of potential acute rejection
7. Woodle ES, First MR, Pirsch J, et al. A prospective, randomized, biomarkers in renal transplant recipients. Transplant Res 2012;
double-blind, placebo-controlled multicenter trial comparing early 1(1):9.
(7 day) corticosteroid cessation versus long-term, low-dose cortico- 31. Sigdel TK, Salomonis N, Nicora CD, et al. The identification of novel
steroid therapy. Ann Surg 2008;248(4):564–77. potential injury mechanisms and candidate biomarkers in renal
8. Hricik DE, Formica RN, Nickerson P, et al. Adverse outcomes of allograft rejection by quantitative proteomics. Mol Cell Proteomics
tacrolimus withdrawal in immune-quiescent kidney transplant 2014;13(2):621–31.
recipients. J Am Soc Nephrol 2015;26(12):3114–22. 32. Freue GV, Sasaki M, Meredith A, et al. Proteomic signatures in
9. Stegall MD, Morris RE, Alloway RR, Mannon RB. Developing new plasma during early acute renal allograft rejection. Mol Cell Pro-
immunosuppression for the next generation of transplant recipients: teomics 2010;9(9):1954–67.
the path forward. Am J Transplant 2016;16(4):1094–101. 33. Hricik DE, Nickerson P, Formica RN, et al. Multicenter validation of
10. Kasiske BL, Israni AK, Snyder JJ, Skeans MA. Patient outcomes in urinary CXCL9 as a risk-stratifying biomarker for kidney transplant
renal transplantation I. the relationship between kidney function injury. Am J Transplant 2013;13(10):2634–44.
and long-term graft survival after kidney transplant. Am J Kidney 34. O’Riordan E, Orlova TN, Mei JJ, et al. Bioinformatic analysis of
Dis 2011;57:466–75. the urine proteome of acute allograft rejection. J Am Soc Nephrol
11. Hariharan S, McBride MA, Cherikh WS, Tolleris CB, Bresnahan BA, 2004;15(12):3240–8.
Johnson CP. Post-transplant renal function in the first year predicts 35. Schaub S, Rush D, Wilkins J, et al. Proteomic-based detection of
long-term kidney transplant survival. Kidney Int 2002;62(1):311–8. urine proteins associated with acute renal allograft rejection. J Am
12. Clayton PA, Lim WH, Wong G, Chadban SJ. Relationship between Soc Nephrol 2004;15(1):219–27.
eGFR decline and hard outcomes after kidney transplants. J Am Soc 36. Schaub S, Wilkins JA, Antonovici M, et al. Proteomic-based iden-
Nephrol 2016 Nov;27(11):3440–6. tification of cleaved urinary beta2-microglobulin as a potential
13. Naresh CN, Hayen A, Weening A, Craig JC, Chadban SJ. Day-to-day marker for acute tubular injury in renal allografts. Am J Transplant
variability in spot urine albumin-creatinine ratio. Am J Kidney Dis 2005;5(4 Pt 1):729–38.
2013;62(6):1095–101. 37. Ling XB, Sigdel TK, Lau K, et al. Integrative urinary peptidomics in
14. Nankivell BJ, Shingde M, Keung KL, et al. The causes, significance renal transplantation identifies biomarkers for acute rejection. J Am
and consequences of inflammatory fibrosis in kidney transplantation: Soc Nephrol 2010;21(4):646–53.
the Banff i-IFTA lesion. Am J Transplant 2018 Feb;18(2):364–76. 38. Scian MJ, Maluf DG, Mas VR. MiRNAs in kidney transplanta-
Available online at: https://doi.org/10.1111/ajt.14609. tion: potential role as new biomarkers. Expert Rev Mol Diagn
15. Prentice RL. Surrogate endpoints in clinical trials: definition and 2013;13(1):93–104.
operational criteria. Stat Med 1989;8(4):431–40. 39. Mas VR, Dumur CI, Scian MJ, Gehrau RC, Maluf DG. MicroRNAs
16. Weir CJ, Walley RJ. Statistical evaluation of biomarkers as surrogate as biomarkers in solid organ transplantation. Am J Transplant
endpoints: a literature review. Stat Med 2006;25(2):183–203. 2013;13(1):11–9.
17. Alonso A, Molenberghs G, Geys H, Buyse M, Vangeneugden T. A 40. Sui W, Dai Y, Huang Y, Lan H, Yan Q, Huang H. Microarray analysis
unifying approach for surrogate marker validation based on Pren- of MicroRNA expression in acute rejection after renal transplanta-
tice’s criteria. Stat Med 2006;25(2):205–21. tion. Transpl Immunol 2008;19(1):81–5.
18. Strehlau J, Pavlakis M, Lipman M, et al. Quantitative detection of 41. Lorenzen JM, Volkmann I, Fiedler J, et al. Urinary miR-210 as a
immune activation transcripts as a diagnostic tool in kidney trans- mediator of acute T-cell mediated rejection in renal allograft recipi-
plantation. Proc Natl Acad Sci USA 1997;94(2):695–700. ents. Am J Transplant 2011;11(10):2221–7.
19. Sharma VK, Bologa RM, Li B, et al. Molecular executors of cell 42. Wilflingseder J, Regele H, Perco P, et al. miRNA profiling discrimi-
death—differential intrarenal expression of Fas ligand, Fas, gran- nates types of rejection and injury in human renal allografts. Trans-
zyme B, and perforin during acute and/or chronic rejection of plantation 2013;95(6):835–41.
human renal allografts. Transplantation 1996;62(12):1860–6. 43. Ben-Dov IZ, Muthukumar T, Morozov P, Mueller FB, Tuschl T,
20. Akalin E, O’Connell PJ. Genomics of chronic allograft injury. Kidney Suthanthiran M. MicroRNA sequence profiles of human kidney
Int Suppl 2010;(119):S33–7. allografts with or without tubulointerstitial fibrosis. Transplantation
21. Ying L, Sarwal M. In praise of arrays. Pediatr Nephrol 2012;94(11):1086–94.
2009;24(9):1643–59. Quiz 55, 59. 44. Scian MJ, Maluf DG, David KG, et al. MicroRNA profiles in allograft
22. Geiss GK, Bumgarner RE, Birditt B, et al. Direct multiplexed measure- tissues and paired urines associate with chronic allograft dysfunc-
ment of gene expression with color-coded probe pairs. Nat Biotech tion with IF/TA. Am J Transplant 2011;11(10):2110–22.
2008;26(3):317–25. 45. Glowacki F, Savary G, Gnemmi V, et al. Increased circulat-
23. Shannon CP, Balshaw R, Ng RT, et al. Two-stage, in silico deconvo- ing miR-21 levels are associated with kidney fibrosis. PLoS One
lution of the lymphocyte compartment of the peripheral whole blood 2013;8(2):e58014.
transcriptome in the context of acute kidney allograft rejection. PLoS 46. Atzler D, Schwedhelm E, Zeller T. Integrated genomics and metabolo-
One 2014;9(4):e95244. mics in nephrology. Nephrol Dial Transplant 2014;29(8):1467–74.
26 • Biomarkers of Kidney Injury and Rejection 431

47. Weiss RH, Kim K. Metabolomics in the study of kidney diseases. Nat 70. Sellares J, Reeve J, Loupy A, et al. Molecular diagnosis of antibody-
Rev Nephrol 2012;8(1):22–33. mediated rejection in human kidney transplants. Am J Transplant
48. Bohra R, Klepacki J, Klawitter J, Klawitter J, Thurman JM, Chris- 2013;13(4):971–83.
tians U. Proteomics and metabolomics in renal transplantation— 71. Famulski KS, Broderick G, Einecke G, et al. Transcriptome analysis
quo vadis? Transpl Int 2013;26(3):225–41. reveals heterogeneity in the injury response of kidney transplants.
49. Blydt-Hansen TD, Sharma A, Gibson IW, Mandal R, Wishart DS. Am J Transplant 2007;7(11):2483–95.
Urinary metabolomics for noninvasive detection of borderline and 72. Obeidat MA, Luyckx VA, Grebe SO, et al. Post-transplant nuclear
acute T cell-mediated rejection in children after kidney transplanta- renal scans correlate with renal injury biomarkers and early allograft
tion. Am J Transplant 2014;14(10):2339–49. outcomes. Nephrol Dial Transplant 2011;26(9):3038–45.
50. Wang JN, Zhou Y, Zhu TY, Wang X, Guo YL. Prediction of acute 73. Williams WW, Taheri D, Tolkoff-Rubin N, Colvin RB. Clinical role of
cellular renal allograft rejection by urinary metabolomics using the renal transplant biopsy. Nat Rev Nephrol 2012;8(2):110–21.
MALDI-FTMS. J Proteome Res 2008;7(8):3597–601. 74. Menon MC, Keung KL, Murphy B, O’Connell PJ. The use of genomics
51. Zhao X, Chen J, Ye L, Xu G. Serum metabolomics study of the and pathway analysis in our understanding and prediction of clinical
acute graft rejection in human renal transplantation based on renal transplant injury. Transplantation 2016;100(7):1405–14.
liquid chromatography-mass spectrometry. J Proteome Res 75. Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease.
2014;13(5):2659–67. Genome Biol 2017;1(1):83.
52. Gourishankar S, Leduc R, Connett J, et al. Pathological and clinical 76. Nankivell BJ, Alexander SI. Rejection of the kidney allograft. N Engl J
characterization of the ‘troubled transplant’: data from the DeKAF Med 2010;363(15):1451–62.
study. Am J Transplant 2010;10(2):324–30. 77. Hricik DE. Transplant immunology and immunosuppression: core
53. Parikh CR, Mansour SG. Perspective on clinical application of curriculum 2015. Am J Kidney Dis 2015;65(6):956–66.
biomarkers in AKI. J Am Soc Nephrol 2017;28(6):1677–85. 78. Lefaucheur C, Loupy A, Vernerey D, et al. Antibody-mediated vas-
54. Mishra J, Ma Q, Prada A, et al. Identification of neutrophil gelatinase- cular rejection of kidney allografts: a population-based study. Lancet
associated lipocalin as a novel early urinary biomarker for ischemic 2013;381(9863):313–19.
renal injury. J Am Soc Nephrol 2003;14(10):2534–43. 79. Salvadori M, Tsalouchos A. Biomarkers in renal transplantation: an
55. Mori K, Lee HT, Rapoport D, et al. Endocytic delivery of lipocalin- updated review. World J Transplant 2017;7(3):161–78.
siderophore-iron complex rescues the kidney from ischemia- 80. Li B, Hartono C, Ding R, et al. Noninvasive diagnosis of renal-
reperfusion injury. J Clin Invest 2005;115(3):610–21. allograft rejection by measurement of messenger RNA for perforin
56. Mishra J, Dent C, Tarabishi R, et al. Neutrophil gelatinase-associated and granzyme B in urine. N Engl J Med 2001;344(13):947–54.
lipocalin (NGAL) as a biomarker for acute renal injury after cardiac 81. Suthanthiran M, Schwartz JE, Ding R, et al. Urinary-cell mRNA pro-
surgery. Lancet 2005;365(9466):1231–8. file and acute cellular rejection in kidney allografts. N Engl J Med
57. Mori K, Nakao K. Neutrophil gelatinase-associated lipocalin 2013;369(1):20–31.
as the real-time indicator of active kidney damage. Kidney Int 82. Matignon M, Ding R, Dadhania DM, et al. Urinary cell mRNA profiles
2007;71(10):967–70. and differential diagnosis of acute kidney graft dysfunction. J Am Soc
58. Ichimura T, Bonventre JV, Bailly V, et al. Kidney injury molecule-1 Nephrol 2014;25(7):1586–97.
(KIM-1), a putative epithelial cell adhesion molecule containing a 83. Panzer U, Steinmetz OM, Reinking RR, et al. Compartment-specific
novel immunoglobulin domain, is up-regulated in renal cells after expression and function of the chemokine IP-10/CXCL10 in a
injury. J Biol Chem 1998;273(7):4135–42. model of renal endothelial microvascular injury. J Am Soc Nephrol
59. van Timmeren MM, Vaidya VS, van Ree RM, et al. High urinary 2006;17(2):454–64.
excretion of kidney injury molecule-1 is an independent predic- 84. Hu H, Kwun J, Aizenstein BD, Knechtle SJ. Noninvasive detection
tor of graft loss in renal transplant recipients. Transplantation of acute and chronic injuries in human renal transplant by eleva-
2007;84(12):1625–30. tion of multiple cytokines/chemokines in urine. Transplantation
60. Reese PP, Hall IE, Weng FL, et al. Associations between deceased- 2009;87(12):1814–20.
donor urine injury biomarkers and kidney transplant outcomes. 85. Jackson JA, Kim EJ, Begley B, et al. Urinary chemokines CXCL9 and
J Am Soc Nephrol 2016;27(5):1534–43. CXCL10 are noninvasive markers of renal allograft rejection and BK
61. Puthumana J, Hall IE, Reese PP, et al. YKL-40 Associates with viral infection. Am J Transplant 2011;11(10):2228–34.
renal recovery in deceased donor kidney transplantation. J Am Soc 86. Hricik DE, Nickerson P, Formica RN, et al. Multicenter validation of
Nephrol 2017;28(2):661–70. urinary CXCL9 as a risk-stratifying biomarker for kidney transplant
62. Parikh CR, Abraham E, Ancukiewicz M, Edelstein CL. Urine injury. Am J Transplant 2013;13(10):2634–44.
IL-18 is an early diagnostic marker for acute kidney injury and 87. Rabant M, Amrouche L, Morin L, et al. Early low urinary CXCL9
predicts mortality in the intensive care unit. J Am Soc Nephrol and CXCL10 might predict immunological quiescence in clini-
2005;16(10):3046–52. cally and histologically stable kidney recipients. Am J Transplant
63. Hall IE, Coca SG, Perazella MA, et al. Risk of poor outcomes with 2016;16(6):1868–81.
novel and traditional biomarkers at clinical AKI diagnosis. Clin J Am 88. Sigdel TK, Lee S, Sarwal MM. Profiling the proteome in renal trans-
Soc Nephrol 2011;6(12):2740–9. plantation. Proteomics Clin Appl 2011;5(5-6):269–80.
64. Heyne N, Kemmner S, Schneider C, Nadalin S, Konigsrainer A, 89. Sigdel TK, Kaushal A, Gritsenko M, et al. Shotgun proteomics identi-
Haring HU. Urinary neutrophil gelatinase-associated lipocalin fies proteins specific for acute renal transplant rejection. Proteomics
accurately detects acute allograft rejection among other causes of Clin Appl 2010;4(1):32–47.
acute kidney injury in renal allograft recipients. Transplantation 90. Sigdel TK, Gao Y, He J, et al. Mining the human urine proteome for
2012;93(12):1252–7. monitoring renal transplant injury. Kidney Int 2016;89(6):1244–52.
65. Hall IE, Doshi MD, Poggio ED, Parikh CR. A comparison of alternative 91. Ong S, Mannon RB. Genomic and proteomic fingerprints of acute
serum biomarkers with creatinine for predicting allograft function rejection in peripheral blood and urine. Transplant Rev (Orlando)
after kidney transplantation. Transplantation 2011;91(1):48–56. 2015;29(2):60–7.
66. Perco P, Pleban C, Kainz A, Lukas A, Mayer B, Oberbauer R. Gene 92. Vasconcellos LM, Schachter AD, Zheng XX, et al. Cytotoxic lympho-
expression and biomarkers in renal transplant ischemia reperfusion cyte gene expression in peripheral blood leukocytes correlates with
injury. Transpl Int 2007;20(1):2–11. rejecting renal allografts. Transplantation 1998;66(5):562–6.
67. Hauser P, Schwarz C, Mitterbauer C, et al. Genome-wide gene- 93. Simon T, Opelz G, Wiesel M, Ott RC, Susal C. Serial peripheral blood
expression patterns of donor kidney biopsies distinguish primary perforin and granzyme B gene expression measurements for predic-
allograft function. Lab Invest 2004;84(3):353–61. tion of acute rejection in kidney graft recipients. Am J Transplant
68. Famulski KS, de Freitas DG, Kreepala C, et al. Molecular phenotypes 2003;3(9):1121–7.
of acute kidney injury in kidney transplants. J Am Soc Nephrol 94. Heng B, Li Y, Shi L, et al. A Meta-analysis of the significance of gran-
2012;23(5):948–58. zyme B and perforin in noninvasive diagnosis of acute rejection after
69. Reeve J, Sellares J, Mengel M, et al. Molecular diagnosis of T cell- kidney transplantation. Transplantation 2015;99(7):1477–86.
mediated rejection in human kidney transplant biopsies. Am J 95. Khatri P, Sarwal MM. Using gene arrays in diagnosis of rejection.
Transplant 2013;13(3):645–55. Curr Opin Organ Transplant 2009;14(1):34–9.
432 Kidney Transplantation: Principles and Practice

96. Flechner SM, Kurian SM, Head SR, et al. Kidney transplant rejection 120. Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Rates and determinants of
and tissue injury by gene profiling of biopsies and peripheral blood progression to graft failure in kidney allograft recipients with de novo
lymphocytes. Am J Transplant 2004;4(9):1475–89. donor-specific antibody. Am J Transplant 2015;15(11):2921–30.
97. Li L, Khatri P, Sigdel TK, et al. A peripheral blood diagnostic test 121. Loupy A, Vernerey D, Tinel C, et al. Subclinical rejection phenotypes
for acute rejection in renal transplantation. Am J Transplant at 1 year post-transplant and outcome of kidney allografts. J Am Soc
2012;12(10):2710–8. Nephrol 2015;26(7):1721–31.
98. Kurian SM, Williams AN, Gelbart T, et al. Molecular classi- 122. Loupy A, Lefaucheur C, Vernerey D, et al. Complement-binding
fiers for acute kidney transplant rejection in peripheral blood anti-HLA antibodies and kidney-allograft survival. N Engl J Med
by whole genome gene expression profiling. Am J Transplant 2013;369(13):1215–26.
2014;14(5):1164–72. 123. Sutherland SM, Chen G, Sequeira FA, Lou CD, Alexander SR,
99. Roedder S, Sigdel T, Salomonis N, et al. The kSORT assay to detect Tyan DB. Complement-fixing donor-specific antibodies identified by
renal transplant patients at high risk for acute rejection: results of a novel C1q assay are associated with allograft loss. Pediatr Trans-
the multicenter AART study. PLoS Med 2014;11(11). e1001759. plant 2012;16(1):12–7.
100. Crespo E, Roedder S, Sigdel T, et al. Molecular and functional noninva- 124. Malheiro J, Tafulo S, Dias L, et al. Determining donor-specific anti-
sive immune monitoring in the ESCAPE study for prediction of subclin- body C1q-binding ability improves the prediction of antibody-
ical renal allograft rejection. Transplantation 2017;101(6):1400–9. mediated rejection in human leucocyte antigen-incompatible kidney
https://doi.org/10.1097/TP.0000000000001287. transplantation. Transpl Int 2017;30(4):347–59.
101. Hricik DE, Augustine J, Nickerson P, et al. Interferon gamma 125. Freitas MC, Rebellato LM, Ozawa M, et al. The role of immuno-
ELISPOT testing as a risk-stratifying biomarker for kidney transplant globulin-G subclasses and C1q in de novo HLA-DQ donor-specific
injury: results from the CTOT-01 multicenter study. Am J Trans- antibody kidney transplantation outcomes. Transplantation
plant 2015;15(12):3166–73. 2013;95(9):1113–9.
102. Halloran PF, Reeve JP, Pereira AB, Hidalgo LG, Famulski KS. Anti- 126. Sicard A, Ducreux S, Rabeyrin M, et al. Detection of C3d-bind-
body-mediated rejection, T cell-mediated rejection, and the injury- ing donor-specific anti-HLA antibodies at diagnosis of humoral
repair response: new insights from the Genome Canada studies of rejection predicts renal graft loss. J Am Soc Nephrol 2015;26(2):
kidney transplant biopsies. Kidney Int 2014;85(2):258–64. 457–67.
103. Halloran PF, Famulski KS, Reeve J. Molecular assessment of dis- 127. Calp-Inal S, Ajaimy M, Melamed ML, et al. The prevalence and
ease states in kidney transplant biopsy samples. Nat Rev Nephrol clinical significance of C1q-binding donor-specific anti-HLA anti-
2016;12(9):534–48. bodies early and late after kidney transplantation. Kidney Int
104. Loupy A, Haas M, Solez K, et al. The Banff 2015 kidney meeting 2016;89(1):209–16.
report: current challenges in rejection classification and prospects for 128. Yabu JM, Higgins JP, Chen G, Sequeira F, Busque S, Tyan DB. C1q-
adopting molecular pathology. Am J Transplant 2017;17(1):28–41. fixing human leukocyte antigen antibodies are specific for predicting
105. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Allen RD, Chap- transplant glomerulopathy and late graft failure after kidney trans-
man JR. The natural history of chronic allograft nephropathy. plantation. Transplantation 2011;91(3):342–7.
N Engl J Med 2003;349(24):2326–33. 129. Comoli P, Cioni M, Tagliamacco A, et al. Acquisition of C3d-Binding
106. Nankivell BJ, Chapman JR. Chronic allograft nephropathy: current activity by de novo donor-specific HLA antibodies correlates with
concepts and future directions. Transplantation 2006;81(5):643–54. graft loss in nonsensitized pediatric kidney recipients. Am J Trans-
107. El-Zoghby ZM, Stegall MD, Lager DJ, et al. Identifying specific causes plant 2016;16(7):2106–16.
of kidney allograft loss. Am J Transplant 2009;9(3):527–35. 130. Viglietti D, Loupy A, Vernerey D, et al. Value of donor-specific anti-
108. Einecke G, Sis B, Reeve J, et al. Antibody-mediated microcircula- HLA antibody monitoring and characterization for risk stratification
tion injury is the major cause of late kidney transplant failure. Am J of kidney allograft loss. J Am Soc Nephrol 2017;28(2):702–15.
Transplant 2009;9(11):2520–31. 131. Lefaucheur C, Viglietti D, Bentlejewski C, et al. IgG donor-specific
109. Naesens M, Kuypers DR, De Vusser K, et al. The histology of kidney anti-human HLA antibody subclasses and kidney allograft antibody-
transplant failure: a long-term follow-up study. Transplantation mediated injury. J Am Soc Nephrol 2016;27(1):293–304.
2014;98(4):427–35. 132. Honger G, Hopfer H, Arnold ML, Spriewald BM, Schaub S, Amico P.
110. Park WD, Griffin MD, Cornell LD, Cosio FG, Stegall MD. Fibrosis with Pretransplant IgG subclasses of donor-specific human leukocyte anti-
inflammation at one year predicts transplant functional decline. gen antibodies and development of antibody-mediated rejection.
J Am Soc Nephrol 2010;21(11):1987–97. Transplantation 2011;92(1):41–7.
111. Torres IB, Moreso F, Sarro E, Meseguer A, Seron D. The interplay 133. Schaub S, Honger G, Koller MT, Liwski R, Amico P. Determinants
between inflammation and fibrosis in kidney transplantation. of C1q binding in the single antigen bead assay. Transplantation
Biomed Res Int; 2014:750602. 2014;98(4):387–93.
112. Scherer A, Gwinner W, Mengel M, et al. Transcriptome changes in 134. Tambur AR, Herrera ND, Haarberg KM, et al. Assessing antibody
renal allograft protocol biopsies at 3 months precede the onset of strength: comparison of MFI, C1q, and titer information. Am J
interstitial fibrosis/tubular atrophy (IF/TA) at 6 months. Nephrol Transplant 2015;15(9):2421–30.
Dial Transplant 2009;24(8):2567–75. 135. Anthony RM, Nimmerjahn F. The role of differential IgG glycosyl-
113. Mengel M, Chang J, Kayser D, et al. The molecular phenotype of 6-week ation in the interaction of antibodies with FcgammaRs in vivo. Curr
protocol biopsies from human renal allografts: reflections of prior injury Opin Organ Transplant 2011;16(1):7–14.
but not future course. Am J Transplant 2011;11(4):708–18. 136. Hirohashi T, Chase CM, Della Pelle P, et al. A novel pathway of
114. Anglicheau D, Muthukumar T, Hummel A, et al. Discovery and valida- chronic allograft rejection mediated by NK cells and alloantibody.
tion of a molecular signature for the noninvasive diagnosis of human Am J Transplant 2012;12(2):313–21.
renal allograft fibrosis. Transplantation 2012;93(11):1136–46. 137. Hidalgo LG, Sis B, Sellares J, et al. NK cell transcripts and NK cells
115. Patel R, Terasaki PI. Significance of the positive crossmatch test in in kidney biopsies from patients with donor-specific antibodies: evi-
kidney transplantation. N Engl J Med 1969;280(14):735–9. dence for NK cell involvement in antibody-mediated rejection. Am J
116. Tait BD, Susal C, Gebel HM, et al. Consensus guidelines on the testing Transplant 2010;10(8):1812–22.
and clinical management issues associated with HLA and non-HLA 138. Zhang X, Reed EF. Effect of antibodies on endothelium. Am J Trans-
antibodies in transplantation. Transplantation 2013;95(1):19–47. plant 2009;9(11):2459–65.
117. Filippone EJ, Farber JL. The humoral theory of transplantation: epit- 139. Toki D, Zhang W, Hor KL, et al. The role of macrophages in the devel-
ope analysis and the pathogenicity of HLA antibodies. J Immunol Res opment of human renal allograft fibrosis in the first year after trans-
2016;2016:5197396. plantation. Am J Transplant 2014;14(9):2126–36.
118. Zhang R. Donor-specific antibodies in kidney transplant recipi- 140. Haas M, Loupy A, Lefaucheur C, et al. The Banff 2017 kidney
ents. Clin J Am Soc Nephrol 2018;13(1):182–92. https://doi. meeting report: revised diagnostic criteria for chronic active T
org/10.2215/CJN.00700117. cell-mediated rejection, antibody-mediated rejection, and pros-
119. Devos JM, Gaber AO, Teeter LD, et al. Intermediate-term graft loss pects for integrative endpoints for next-generation clinical trials.
after renal transplantation is associated with both donor-specific Am J Transplant 2018;18(2):293–307. https://doi.org/10.1111/
antibody and acute rejection. Transplantation 2014;97(5):534–40. ajt.14625.
26 • Biomarkers of Kidney Injury and Rejection 433

141. Valenzuela NM, Reed EF. Antibody-mediated rejection across solid 150. Brouard S, Le Bars A, Dufay A, et al. Identification of a gene
organ transplants: manifestations, mechanisms, and therapies. expression profile associated with operational tolerance among
J Clin Invest 2017;127(7):2492–504. a selected group of stable kidney transplant patients. Transpl Int
142. Guidicelli G, Guerville F, Lepreux S, et al. Non-complement-binding 2011;24(6):536–47.
de novo donor-specific anti-HLA antibodies and kidney allograft sur- 151. Newell KA, Asare A, Kirk AD, et al. Identification of a B cell signature
vival. J Am Soc Nephrol 2016;27(2):615–25. associated with renal transplant tolerance in humans. J Clin Invest
143. Rostaing L, Vincenti F, Grinyo J, et al. Long-term belatacept expo- 2010;120(6):1836–47.
sure maintains efficacy and safety at 5 years: results from the 152. Sagoo P, Perucha E, Sawitzki B, et al. Development of a cross-
long-term extension of the BENEFIT study. Am J Transplant platform biomarker signature to detect renal transplant tolerance in
2013;13(11):2875–83. humans. J Clin Invest 2010;120(6):1848–61.
144. Vincenti F, Rostaing L, Grinyo J, et al. Belatacept and long-term out- 153. Baron D, Ramstein G, Chesneau M, et al. A common gene signature
comes in kidney transplantation. N Engl J Med 2016;374(4):333–43. across multiple studies relate biomarkers and functional regulation
145. Macklin PS, Morris PJ, Knight SR. A systematic review of the use of in tolerance to renal allograft. Kidney Int 2015;87(5):984–95.
rituximab for desensitization in renal transplantation. Transplanta- 154. Braud C, Baeten D, Giral M, et al. Immunosuppressive drug-free
tion 2014;98(8):794–805. operational immune tolerance in human kidney transplant recipi-
146. Roberts DM, Jiang SH, Chadban SJ. The treatment of acute antibody- ents: part I. Blood gene expression statistical analysis. J Cell Biochem
mediated rejection in kidney transplant recipients—a systematic 2008;103(6):1681–92.
review. Transplantation 2012;94(8):775–83. 155. Lozano JJ, Pallier A, Martinez-Llordella M, et al. Comparison of
147. Vo AA, Choi J, Kim I, et al. A Phase I/II trial of the interleukin-6 transcriptional and blood cell-phenotypic markers between opera-
receptor-specific humanized monoclonal (tocilizumab) + intrave- tionally tolerant liver and kidney recipients. Am J Transplant
nous immunoglobulin in difficult to desensitize patients. Transplan- 2011;11(9):1916–26.
tation 2015;99(11):2356–63. 156. ANZDATA Registry. 39th Annual Report, Chapter 8: Transplanta-
148. Choi J, Aubert O, Vo A, et al. Assessment of tocilizumab (anti- tion. Adelaide, Australia; 2017.
interleukin-6 receptor monoclonal) as a potential treatment for 157. Freidlin B, McShane LM, Korn EL. Randomized clinical trials with
chronic antibody-mediated rejection and transplant glomerulopa- biomarkers: design issues. J Natl Cancer Inst 2010;102(3):152–60.
thy in HLA-sensitized renal allograft recipients. Am J Transplant 158. Freidlin B, Jiang W, Simon R. The cross-validated adaptive signature
2017;17(9):2381–9. design. Clin Cancer Res 2010;16(2):691–8.
149. Brouard S, Mansfield E, Braud C, et al. Identification of a peripheral
blood transcriptional biomarker panel associated with operational
renal allograft tolerance. Proc Natl Acad Sci USA 2007;104(39):
15448–53.
27 Chronic Allograft Failure
BRIAN J. NANKIVELL

CHAPTER OUTLINE Introduction Transplant Glomerulopathy and Chronic-


Epidemiology Active Antibody-Mediated Rejection
Pathophysiology of Chronic Allograft Recurrent Glomerular Disease
Damage Late Acute Rejection and Intercurrent
Historical Concepts of Allograft Failure Illnesses
Clinical Risk Factors of Allograft Loss Approach to a Failing Allograft
A Unified Model of Chronic Transplant Monitoring of Renal Function
Injury Proteinuria and Urinalysis
Mechanistic Theories of Chronic Allograft Renal Transplant Imaging
Injury Immune Surveillance Tests
Inflammatory Cytokines Urinary Diagnostics
Reactive Oxygen Species and Glomerular Immune Surveillance Markers in Blood
Hyperfiltration Kidney Transplant Biopsy
Failed Resolution of Chronic Inflammation Principles Guiding Clinically Indicated Biopsy
Epithelial-Mesenchymal Transition–Induced Risk and Safety of Kidney Transplant
Fibrosis Biopsies
Donor age and Replicative Senescence Diagnostic Algorithm for a Chronically
Cortical Ischemia and Angioregression Failing Graft
Internal Structural Organ Failure Treatment of a Failing Allograft
Time Course of Allograft Damage Long-Term Immunosuppression
Donor Quality and Procurement Injury General Treatment Principles
Delayed Graft Function and Ischemic Injury Treatment Approach by Specific Diagnosis
Early Tubulointerstitial Damage 1. Interstitial Fibrosis and Tubular Atrophy
Acute Rejection and Alloimmune 2. Approach to Calcineurin Inhibitor
Mechanisms Nephrotoxicity
Subclinical Rejection 3. Chronic Antibody-Mediated Rejection
Bk Virus Nephropathy 4. Chronic Active T Cell Mediated Rejection
Progressive and Late-Stage Chronic Allograft 5. Treatment of Acute Late Rejection
Damage 6. Treatment of Recurrent Disease
Chronic-Active T Cell Mediated Rejection 7. Treatment of BK Virus Allograft
Calcineurin Inhibitor Nephrotoxicity Nephropathy
Progressive Glomerular Abnormalities Summary

Introduction are being transplanted into higher-risk recipients generat-


ing potential survival bias.3 Kidney transplantation still
EPIDEMIOLOGY offers substantial survival benefit compared with remain-
ing waitlisted for deceased donor transplantation on dialy-
Despite the success of modern kidney transplantation sis.4 Progressive transplant structural deterioration causes
where early acute rejection rates are less than 15% and renal dysfunction and graft failure, an event accompanied
1-year graft survivals exceed 90%, yearly graft-loss rates by substantial morbidity and mortality. This chapter will
have not greatly changed at 4%.1,2 Short-term improve- evaluate the causes of chronic transplant failure, its patho-
ments have incompletely translated into better long-term physiology and diagnostic pathology, and present a thera-
survival, although increased numbers of marginal donors peutic approach.

434
27 • Chronic Allograft Failure 435

Pathophysiology of Chronic nonimmune mechanisms, which cause progressive dam-


age to individual nephrons and eventually compromise
Allograft Damage the internal structure of the organ. The transplanted kid-
ney can be progressively injured from a number of non-
HISTORICAL CONCEPTS OF ALLOGRAFT mutually exclusive pathologic insults and stresses, that
FAILURE occur over the allograft’s lifetime. Cells of local anatomic
compartments (tubular, interstitial, microvascular, and
Originally, kidney allograft damage and subsequent allograft
glomerular) have a limited repertoire of response to injury,
failure was simply attributed in registry data to “chronic
expressed by stereotypical gene patterns and a limited num-
rejection,” with the corresponding pathology of interstitial
ber of histologic phenotypes. Different drivers of injury are
lymphocytic infiltration and tubulitis with tubular atrophy
expressed at different times, and the relative mixture of dis-
and chronic interstitial fibrosis (IF/TA), often accompanied
ease and pathology prevalence alters with time posttrans-
by vascular or glomerular abnormalities and redesignated
plantation, but they can operate simultaneously and be
as chronic allograft nephropathy (CAN). Although common
observed within the same biopsy.
in the prednisolone-azathioprine immunosuppression era
The histopathology of a chronically failing allograft typi-
(early acute rejection rates have fallen from 70% to 80% to
cally shows chronic interstitial fibrosis, tubular atrophy,
less than 15%),5 the clinical presentation (fever, graft tender-
glomerulosclerosis, and vascular abnormalities. Chronic
ness, oliguria) and pattern of histologic injury have altered
alloimmune activity is manifest by persistent cellular
with the introduction of potent modern regimes, incorpo-
interstitial inflammation in scarred and nonscarred areas,
rating the calcineurin inhibitors (CNIs), cyclosporine and
fibrointimal arterial hyperplasia, and transplant glomeru-
tacrolimus, and with better immunologic matching. Acute
lopathy associated with circulating donor-specific antibody
and chronic rejection nowadays presents with a rising serum
and microvascular tissue C4d. These features are end-path-
creatinine, although episodes remaining are clinically more
way phenotypes caused by the cumulative effects of tissue
severe. Rejection remains a clinically relevant problem
injury and its fibrotic healing response. They are influ-
for immunologically active patients, those with iatrogenic
enced and modulated in turn by alloimmunity, noncognate
underimmunosuppression, and noncompliant individuals.
inflammation, immunosuppression, and against intrinsic
donor, the resilience (Fig. 27.1; see Table 27.1).
CLINICAL RISK FACTORS OF ALLOGRAFT LOSS The cumulative damage hypothesis assumes that
chronic allograft damage is the end result of multiple, time-
The risk factors for chronic allograft loss include donor
dependent immune and nonimmune insults, which cause
organ quality and ischemia-reperfusion injury manifest by
irreversible loss of a finite number of transplanted neph-
delayed graft function (DGF); immune factors, including
rons.10–12 Early events include procurement, preservation,
human leukocyte antigens (HLA) mismatch, donor-specific
and implantation injury, followed by cellular and antibody-
and non-HLA antibodies, episodes of acute and chronic
mediated alloimmunity, and additional nonimmune pro-
rejection; and recipient factors such as hypertension, pro-
cesses such as CNI nephrotoxicity, infection, acute kidney
teinuria, smoking, and medication nonadherence6 Table
injury, hypertension, and diabetes.10,13 Destroyed neph-
27.1. Posttransplant risk factors for graft failure are subcat-
rons cannot be replaced once lost, although hypertrophy
egorized into immune and nonimmune causes.
of tubular cells within remaining nephrons may partially
Proteinuria (from 0.5 g/day) is a powerful composite risk
compensate for losses by hyperfiltration. The kidney gradu-
for graft loss and allograft dysfunction.7,8 It originates from
ally fails from the incremental loss of individual nephrons,
either glomerular leakage (glomerular proteinuria) or inade-
combined with internal structural derangements contribut-
quate tubular reabsorption (from atrophic tubules within IF/
ing to organ malfunction. Multiple alloimmune, ischemic,
TA; tubular proteinuria). Direct injury from tubular resorp-
and inflammatory stimuli all cause lethal or sublethal cel-
tion of filtered toxic substances, cytokines, or other mediators
lular injury, which provokes a profibrotic chronic healing
accompanying proteinuria can cause further injury.
response. (see Table 27.1, Fig. 27.2).
Posttransplant hypertension is associated with graft
failure and death, by registry analysis, and occurs in 70%
to 90% of recipients. Contributors include pretransplant
hypertension and dialysis vascular disease, glucocorti-
Mechanistic Theories of Chronic
coid and cyclosporine therapy, allograft dysfunction with Allograft Injury
impaired excretion of dietary sodium, or transplant renal
artery stenosis. Allograft hypertensive changes include Several other hypotheses of chronic allograft failure have
fibrointimal thickening of small muscular arteries with been advanced, although they may be better described as
duplication of the internal elastic lamina, arteriolar hya- pathophysiologic mediators, or pathways of injury, rather
linosis, and ischemic glomerulosclerosis.6,9 Other adverse than as principal etiologic causes (see Fig. 27.2).
factors for graft deterioration include recipient smoking,
uncontrolled dyslipidemia, and diabetes mellitus (either INFLAMMATORY CYTOKINES
preexisting or new-onset posttransplant).
The cytokine excess theory postulates that acute and
A UNIFIED MODEL OF CHRONIC TRANSPLANT repeated tissue injury induces excessive cytokine pro-
duction (e.g., interferon-γ), resulting in interstitial and
INJURY
vascular fibrosis (mediated by TGF-β1). Cytokines and che-
The pathophysiology of chronic allograft failure is best mokines can recruit, activate and traffic inflammatory cells.
conceptualized as the end result of several immune and Upregulated or altered expression of VEGF, endothelin-1,
436 Kidney Transplantation: Principles and Practice

Ischemia- Intercurrent
ETIOLOGICAL EVENTS reperfusion illness—CMV
Cold ischemia injury and ROS Recurrent
Warm ischemia Late GN

Kidney transplant dysfunction


Technical problems rejection

Acute BK CAN

TRANSPLANTATION
Donor age
Nephropathy

Graft failure
Hypertension rejection
Vascular disease
Brain death
Ionotrope use
Donor ATN

DGF
CNI
SCR
Nephrotoxicity
True chronic rejection

Hypertension, diabetes mellitus, dyslipidemia,


KIDNEY PATHOLOGY smoking, proteinuria
Lymphocytic infiltration AR/SCR
Arteriolar hyalinosis and
CNI/HT/GN
glomerulosclerosis
Tubular atrophy and
IF/TA
chronic interstitial fibrosis

Vascular or antibody-mediated
AHR/CAMR
rejection

Fig. 27.1 Immune and nonimmune events leading to allograft damage and transplant failure. ATN, acute tubular necrosis; CAN, chronic allograft
nephropathy; CMV; cytomegalovirus; CNI, calcineurin inhibitors; DGF, delayed graft function; GN; glomerulonephritis; ROS, reactive oxygen species;
SCR, subclinical rejection.

TABLE 27.1 Risks Factors and Causes of Allograft Injury plasminogen-activating factor-1, MCP-1, PDGF-A and B,
RANTES, BMP 7, hepatocyte growth factor, CTGF, and
NONIMMUNE DONOR RISKS advanced glycation end-products, have all been described
Deceased donor kidney (vs. living donor) in experimental and human “chronic rejection.”14–18
Autonomic storm, inotropic use, donor renal failure
Donation after circulatory death (DCD, vs. donation after brain death)
Donor quality: older age, female, hypertension or diabetes, vascular REACTIVE OXYGEN SPECIES AND GLOMERULAR
disease as extended criteria donor (ECD) HYPERFILTRATION
Ischemia-reperfusion injury (organ perfusion and transport)
Prolonged organ ischemia (warm and cold ischemia times) Excessive or uncontrolled production of reactive oxygen
Acute tubular necrosis (implantation biopsy finding) and delayed species (ROS) from tubular cell mitochondria can cause cel-
graft function (clinical manifestation)
lular injury, apoptosis, and senescence. Interstitial induc-
NONIMMUNE RECIPIENT RISKS AND CAUSES ible nitric oxide synthase protein expression, nitrotyrosine,
Female sex, obesity and ex vivo ROS production are increased in CAN.19
Organ size mismatch (especially pediatric recipients) The hyperfiltration theory postulates that an increased
Ascending urinary tract infection and graft pyelonephritis
Transplant ureteric obstruction
share of the kidney’s metabolic and protein reabsorption
Polyoma (BK) virus nephropathy load falls to a diminishing number of remaining nephrons,
Calcineurin inhibitor nephrotoxicity causing glomerular hypertension and secondary glomeru-
Recurrent renal disease or de novo glomerulonephritis losclerosis. However, evidence in human transplantation
Recipient hypertension, hyperlipidemia, smoking, and proteinuria is contradictory, with many registry studies showing no
Preexisting or posttransplantation diabetes mellitus
effect on graft survival in donor-recipient size-mismatched
ALLOIMMUNE FACTORS pairs.20 The classical hyperfiltration lesions of secondary
Young recipient age focal segmental glomerulosclerosis (FSGS) are uncom-
Race and ethnicity with altered disposition of immunosuppressive mon. Hyperfiltration is relevant with substantial nephron
agents, and differing socioeconomic circumstances
Variable medication trough levels (suggests noncompliance or pos- loss, or when an infant kidney is transplanted into a large
sibly malabsorption) adult.
Recipient’s genetic alloimmune and inflammatory response
Histoincompatibility (HLA or eplet mismatches)
Recipient presensitization (panel reactive antibodies, cPRA) or donor FAILED RESOLUTION OF CHRONIC
specific antibodies INFLAMMATION
Acute rejection that is severe, steroid-resistant, vascular, antibody-
mediated, or late occurring rejection Wound healing after acute injury is normally self-lim-
Subclinical rejection, chronic T cell mediated rejection, late de novo ited, with complete resolution of inflammation and fibro-
anti-HLA antibody formation (donor-specific antibodies) and chronic genesis. Allografts may be repeatedly subject to episodic
antibody-mediated rejection with transplant glomerulopathy
Therapy nonadherence acute injury, where resolution of inflammation is partial
or incomplete. A self-perpetuating cycle of tubular injury,
27 • Chronic Allograft Failure 437

TRANSPLANTED Donor organ quality


KIDNEY Hight KDPI, older ECD kidney
Donor vascular disease

Ischemic injury
Procurement Immunosuppressive
Preservation therapy RECIPIENT
DCD ALLOIMMUNITY
Warm & cold ischemia Regraft
Donor ATN HLA/eplet mismatch
cPRA level
Pretransplant DSA
Mononuclear cell DSA Recipient age
DGF inflammation C4d

Fibrointimal
Tubular damage REJECTION hyperplasia
Ischemia/reperfusion Acute, subclinical and chronic
Arteries
BK viral infection
Nephrotoxins TCMR AMR
Allergic nephritis Chronic vascular
disease
Chronic
TCMR Microvascular
inflammation
i-IF/TA

DSA
Tubular atrophy Interstitial Nonimmune factors
fibrosis Hypertension
Diabetes
Dyslipidemia/smoking
Profibrotic mediators Chronic Vascular senescence
growth factors AMR
cytokines/EMT Remodelling

Transplant
IF/TA glomerulopathy Arteriolar
hyalinosis CNI

Nephron attrition Afferent


atubular glomeruli Ischemia arterioles
tubular collapse Hyperfiltration Glomerulosclerosis
structural disruption

Inflammation

↓ GFR Glomerular injury


ALLOGRAFT Proteinuria Glomerulonephritis
FAILURE Diabetic glomerulopathy
HUS & ischemia

Fig. 27.2 Pathophysiology of chronic allograft injury. The transplanted kidney of variable donor organ quality is subjected to a teim-dependent number
of immune and nonimmune insults which are expressed within the anatomic microcompartments of the kidney. Renal tubules are injured by ischemia-
reperfusion, acute T cell rejection, and BK viral infection leading to tubular atrophy and a healing response of chronic interstitial fibrosis. Glomeruli
are injured by antibody-mediated rejection, recurrent glomerulonephritis, and vascular ischemia from calcineurin inhibitor nephrotoxicity and other
recipient vascular diseases (e.g., hypertension, diabetes, donor disease) resulting in glomerulosclerosis. Malfunction of either glomeruli or renal tubules
results in failure of the entire nephron.

enhanced allorecognition, and further immune-mediated impairment, greater structural damage by sequential histol-
injury may result. ogy, and death-censored graft failure.21,24–28
The total inflammatory burden, irrespective of location, is
a strong predictor of graft survival and superior to the Banff EPITHELIAL-MESENCHYMAL TRANSITION–
i-score alone.21 Intragraft Foxp3+ T (regulatory, Treg) cells INDUCED FIBROSIS
have been associated with better graft function, sugges-
tive of a protective effect, although other data report mixed Epithelial-mesenchymal transition (EMT) describes a
results.22 Tregs may be important for transplant tolerance; sequence of phenotypical changes where tubular epithelial
however, their ability to recognize allo- or self-antigens is cells are transformed into spindle-shaped mesenchymal or
unclear.23 Inflammation within IF/TA areas (i-IFTA) is com- myofibroblast-like cells. Excepting distal collecting ducts,
mon, and follows prior subclinical and acute T-cell-­mediated tubular cells originate from fetal mesenchyme and transition
rejection (TCMR). i-IFTA is associated with functional to an epithelial phenotype during embryonal development.
438 Kidney Transplantation: Principles and Practice

They retain their ability to back-differentiate into mesen-


chymal cells with appropriate stimuli, such as sublethal
tubular hypoxic injury, TGF-β1, or IL-1. Clinical drivers
of EMT include subclinical rejection, cyclosporine nephro-
toxicity, and oxidative stress.29–31 EMT is also reversible;
surviving cells can repopulate injured and denuded tubules
by mesenchymal-to-epithelial transition.32 The genetically
programmed steps of EMT begin with impaired cell-to-cell
adhesion, loss of tight and adherent junctions, desmosomes,
and E-cadherin (an epithelial marker); this is followed by
reorganization of F-actin stress fibers and expression of
smooth muscle actin (a mesenchymal marker), filopodia,
and lamellipodia capable of cellular migration (Fig. 27.3).
Transitioned myofibroblasts can then secrete interstitial
matrix proteins, collagen, and fibronectin.
Evidence that EMT provides the interstitial fibroblasts
(type II EMT) responsible for IF/TA is limited. These fibro-
blasts are predominantly of recipient descent consistent
with resident or infiltrating fibroblast origin (using Y chro- Fig. 27.3 Epithelial mesenchymal transition illustrated by dual stain-
mosomal DNA analysis of sex-mismatched donor-recipient ing of E-cadherin (blue) and α-smooth-muscle actin (brown) in a tubular
pairs).33 Cell lineage tracing studies even suggest some epithelial cell.
fibroblasts originate from recipient capillary endothelial
cells (via endothelial-to-mesenchymal transition).34 EMT is
not uncommon in cross-sectional studies, with transitional theorized to drive normal cells into a senescent state with
tubular cells expressing both epithelial and mesenchymal exhausted repair, and this would lead to graft failure.39
markers (e.g., β-catenin and vimentin).31,35–38 Sequential Subsequent research has disproved replicative senescence
histologic studies correlating EMT with subsequent pro- as the cause of allograft failure.42
gression of interstitial fibrosis are mixed.30,31 However, the
EMT is likely to be incomplete in many cases, where trans- CORTICAL ISCHEMIA AND ANGIOREGRESSION
formed tubular cells without cell-adhesion molecules are
sloughed into the tubular lumen, instead of crossing into Tubular epithelial cells are metabolically active with
the interstitial compartment. abundant mitochondria powering electrolyte pumps and
endocytotic protein reabsorption. Supplied by a network
of peritubular capillaries (PTC) downstream from glomer-
DONOR AGE AND REPLICATIVE SENESCENCE ular efferent arterioles, they are susceptible to ischemia
Replicative senescence is the normal cellular aging pro- from upstream vascular and glomerular disease. Partial
cess that ultimately leads to irreversible growth arrest or total glomerulosclerosis, CNI-induced vasoconstric-
and has been postulated to cause allograft failure and an tion, arteriolar hyalinosis, and small arterial vascular
explanation for reduced survival from older donor kid- disease (from donor disease, hypertension, or fibrointimal
neys.39–41 Preexisting structural abnormalities includ- hyperplasia), all reduce postglomerular capillary blood
ing vascular disease and glomerulosclerosis, and the flow. Injured capillary endothelial cells display nuclear
reduced regenerative capacity of older kidneys also swelling, fenestrae loss, and apoptotic detachment from
contribute.42,43 Senescent cells are enlarged and flat- the PTC basement membrane. Hypoxic tubular cells
tened from altered cytoskeletal collagen, with lipofuscin switch to anaerobic glycolysis and activate antiapoptotic
deposition and age-dependent p53 and p16 gene activa- molecular programs as an adaptive survival response.
tion.41 Cultured somatic cells stop cycling after a fixed Hypoxia-inducible factors (e.g., HIF1α) regulate and acti-
number of doublings (Hayflick limit), which in humans vate multiple genes that control glucose metabolism, cel-
is regulated by telomeres that shorten after each mitotic lular proliferation and survival, angiogenesis, chemotaxis
division. Although shorter telomeres are found in older of inflammatory cells, and regulate extracellular matrix
native and transplanted kidneys, the “mitotic clock” via profibrotic cytokines, such as TGFα, PDGF, CTGF, and
(and p53 tumor-suppressor pathway) is not accelerated VEGF.44
by transplantation nor responsible for IF/TA-associated The microvascular capillary and small muscular arte-
graft failure. A separate telomere-independent (p16INK4a) rial networks become progressively attenuated with
senescence pathway operates dependent on oxidative chronic damage and have been reported in chronic TCMR,
stress. The senescent phenotype of chronic allograft C4d+ chronic antibody-mediated rejection (AMR), and
nephropathy resembles aging and reflects intrinsic alter- sclerosing CAN. Capillary rarefaction with loss of PTC
ations of cell-cycling pathways.40,42 surface area correlated with IF/TA, allograft dysfunction,
The input-stress model describes the interplay between and proteinuria.45 Cross-sectional transplant studies limit
donor organ quality subject to ongoing stress factors from causal inferences of angioregression-associated tubular
immune-mediated or nonimmune (load) mechanisms, ischemia, versus microvascular loss being a consequence
including hypertension, hyperfiltration, proteinuria, dys- of reduced supportive angiogenic factors from injured
lipidemia, nephrotoxic drugs, and infection. Stressors were tubules.45–47
27 • Chronic Allograft Failure 439

INTERNAL STRUCTURAL ORGAN FAILURE tubular collapse or luminal obstruction from debris) causes
functional failure of the whole unit. Global or partial glo-
Transplant renal function is reduced by the summated dam- merulosclerosis, and transplant glomerulopathy, all limit
age to individual nephrons, or by disruption of the kidney’s ultrafiltrate generation. Atubular glomeruli develop after
internal structure, which is essential to form urine. Local disconnection of their downstream collapsed tubules.48
compromise along the length of the intact nephron (such as Internal disruption reduces the kidney’s ability to modify
tubular ultrafiltrate into concentrated and acidified urine.
Time-course of functional decline A segmentally injured glomerulus with synechiae attached
100 to the Bowman’s capsule can misdirect ultrafiltrate into
Late events:
acute rejection severe paraglomerular or paratubular channels, leading to the
Transplant GFR (mL /min)

ATN noncompliance
interstitial space. Inflammatory necrosis with obliterative
Early events:
fibrosis destroys the integrity of the tubule.48 Allograft renal
severe rejection failure can be considered as the summated losses of indi-
50
BKVAN
Slow decline:
vidual nephrons combined with disturbance of the organ’s
chronic rejection Return internal organization.
CNI nephrotoxicity to
recurrent GN dialysis

Marginal donor / DGF Time Course of Allograft Damage


10
Transplant renal failure level
0 The pathway of progression from implanted donor kidney
0 1 5 10 to failed transplant comprises a series of time-dependent
Years after transplantation insults causing progressive histologic injury (Figs. 27.4
Fig. 27.4 Different clinical patterns of the time-course of renal func- and 27.5, see Table 27.1). In addition to inherited donor
tional deterioration provide clues toward the etiologic diagnosis. Kid- disease, two broad overlapping patterns of allograft damage
neys with poor initial function (from a marginal donor or early ischemic can be discerned by sequential biopsy studies. Tubulointer-
injury with delayed graft function) are compared with kidneys with stitial injury dominates the early posttransplant period, fol-
initial satisfactory function injured by early severe rejection or BK viral
infection) or late acute graft impairment (late rejection or acute kid-
lowed by microvascular and glomerular abnormalities and
ney injury), and are contrasted with transplants that show a slow and slowly increasing IF/TA.10,49
steady decline. (From chronic rejection, calcineurin inhibitor nephrotoxic- Substantial numbers of tubules are lost early post-
ity, or recurrent renal disease.) transplant from ischemia-reperfusion injury, early severe

Early Under-immunosuppression
posttransplant nonadherence
events iatrogenic Late acute Recurrent / de novo
rejection glomerulonephritis

Acute Inter-current
illness / sepsis

Allograft failure
rejection
de novo DSA
ATN
BKVAN
TRANSPLANTATION

Mixed rejection
Procurement Chronic AMR
Ischemia tion
ic rejec
Chron
DGF

Chronic TCMR
SCR CNI nephrotoxicity
Donor
quality

Surgical Hypertension, NODAT, dyslipidemia, proteinuria


complications
Declining GFR

Time after transplantation


Allograft pathology
Cellular infiltration & i-IF/TA
Microvascular inflammation & transplant glomerulopathy

Tubular atrophy & interstitial fibrosis


IF/TA
Arteriolar hyalinosis & glomerulosclerosis
Fig. 27.5 Risks factors and causes of allograft injury by time after transplantation causes cumulative damage. Early posttransplant injury is mediated by
donor factors, ischemia-reperfusion, and early acute rejection. Later damage to the kidney is mediated by subacute persistent diseases (e.g., chronic rejec-
tion, calcineurin inhibitor nephrotoxicity, recurrent disease, hypertensive vascular disease) with superimposed acute events (e.g., acute kidney injury or late
acute rejection from underimmunosuppression). Allograft failure occurs due to summated loss of the finite number of transplanted nephrons.
440 Kidney Transplantation: Principles and Practice

acute rejection, persistent subclinical rejection (SCR), or to 100% (the worst) is half that of a living- or standard-
polyomavirus (BK virus allograft nephropathy [BKVAN]). criteria deceased donors (SCD) scoring 0% to 20% (the best
Subsequent tubular injury is often less intense and driven kidneys). Kidneys from donors with preexisting diabetes or
by residual alloimmunity and inflammation, accompa- hypertension display marginally lower aggregate surviv-
nied by glomerular and microvascular changes. Later als (but still provide benefit over waitlisting), however they
disease drivers include late acute rejection from nonad- require careful vetting before acceptance.
herence, CNI nephrotoxicity, chronic AMR or chronic- Brain death indirectly influences graft outcome by poten-
active CA-TCMR, recurrent glomerulonephritis, acute tiation of graft immunogenicity and other nonspecific
kidney injury (AKI) from intercurrent illness, and patho- pathophysiologic changes. Registry survival rates of liv-
logic effects of hypertension, dyslipidemia, and diabetes ing unrelated and one haplotype-matched living related
mellitus (Table 27.2). Chronic histologic damage and donor kidneys are identical, despite greater genetic and
specific diseases exert additive and independent effects on HLA differences, and superior compared with deceased
graft outcome.49 donor transplants. The transplanted organ is immunologi-
cally altered by proinflammatory mediators released during
donor brain death, resulting in increased acute rejection
DONOR QUALITY AND PROCUREMENT INJURY
rates. Injured tissues express innate danger receptors or
Inherited donor abnormalities strongly influence posttrans- Toll receptor system ligands (normally signaling intracel-
plant histology, function, and response to injury which lular infection), which promotes immune cell maturation,
ultimately determine long-term graft survival. Implanta- activation, and rejection. Experimental brain death releases
tion biopsy histology accurately defines the contribution of a cascade of chemokines, cytokines, proinflammatory lym-
donor disease relative to subsequent factors in later biopsy phokines (TNF-α, interferon-γ), and adhesion molecules,
samples and is recommended. Important donor pathologic with increased expression of major histocompatibility com-
features include the extent of glomerulosclerosis (>20% is plex (MHC) antigens, which amplify the host’s alloimmune
severe, and these kidneys are often discarded), glomeru- response.51
lomegaly (with implied nephron loss and hyperfiltration), The “autonomic storm” from donation after brain death
and microvascular disease (a persistent abnormality asso- (DBD) causes chaotic blood pressure fluctuation. An early
ciated with donor age, diabetes, hypertension, and death hypertensive phase from brainstem herniation and mas-
from cerebrovascular disease). Semiqualitiative scoring sive circulating catecholamine release, is followed by
systems help determine potential suitability for transplan- hypotension from hypothalamic-pituitary dysfunction
tation, with fair predictive capacity against outcomes.50 (and reduced thyroid and cortisol levels), central diabetes
Kidney Donor Profile Index (KPDI) is a single numeri- insipidus with electrolyte disturbances, and hypothermia
cal measure reflecting 10 (adverse) clinical donor disease from core temperature dysregulation. Systemic hypoten-
and demographic parameters, which summarizes relative sion, cardiovascular instability, adrenergic vasoconstric-
organ quality (actually inferiority), with a modest C-statis- tion, and coagulopathies cause donor AKI. Histologic
tic. Matching recipients for longevity with the most optimal abnormalities associated with brain death include glomer-
KPDI kidneys is used within the US allocation algorithm. ular hyperemia, glomerulitis, periglomerulitis, endothelial
The projected graft half-life of KPDI kidneys scoring 86% cell proliferation, tubular vacuolation (from administered
osmotic agents such as mannitol), followed by tubu-
TABLE 27.2 Differential Diagnosis of Renal Allograft lar degeneration, acute tubular necrosis (ATN; seen on
Dysfunction implantation biopsy), and tubular atrophy. Transplant
dysfunction is worst from hemodynamically unstable
STRUCTURAL/INFECTIVE
donors with prolonged hypotension after brain death. The
Ureteric obstruction clinical sequela is DGF with the need for posttransplant
Lower urinary tract obstruction (bladder outlet, prostate)
Transplant renal arterial stenosis dialysis. Long-term transplant outcomes are comparable
Recurrent pyelonephritis or vesicoureteric reflux for kidneys with and without AKI, provided cortical necro-
BK virus nephropathy sis is excluded.52
ALLOIMMUNE INJURY
Late acute rejection (iatrogenic or patient noncompliance) DELAYED GRAFT FUNCTION AND ISCHEMIC
Chronic-active T cell rejection with i-IFTA INJURY
Chronic-active antibody-mediated rejection with transplant glomeru-
lopathy DGF, defined by need for dialysis within the first posttrans-
Mixed phenotype of late acute rejection (antibody-mediated and
TCMR with IF/TA, glomerulosclerosis and fibrointimal hyperplasia)
plant week, has gradually increased from 15% to about
22% over two decades53 as kidneys from older deceased
OTHER PATHOPHYSIOLOGIES donors with vascular comorbidity (expanded criteria
Nonspecific sclerosing tubulointerstitial damage (formally designated donors, ECD) or after circulatory death (DCD, or non-
as chronic allograft nephropathy)
Chronic calcineurin inhibitor nephrotoxicity
heartbeating donors) are being transplanted because of
Thrombotic microangiopathy organ shortages. Although “marginal” kidneys result in
Recurrent or de novo glomerulonephritis inferior initial function rates and patient and allograft sur-
Poorly controlled hypertension with hypertensive nephrosclerosis vival relative to standard criteria donors (SCD), the over-
Metabolic disorders (e.g., hypercalcemia, metabolic acidosis) all recipient survival is better than remaining on dialysis.
Concomitant drugs (e.g., ACE-I, ARB, NSAID, COX-2 inhibitors)
Acute kidney injury from severe medical illness DGF incidence correlates with donor age, organ size and
quality (older donor, diabetes, AKI, and hypertension),
27 • Chronic Allograft Failure 441

and donor hypotension or ischemic times (prolonged from ACUTE REJECTION AND ALLOIMMUNE
shipping or perioperative surgical reperfusion times). DGF MECHANISMS
increases long-term graft loss by up to 41% using system-
atic meta-analysis.54 Acute rejection is a risk factor of graft half-life and actu-
Renal tubular epithelial cells are metabolically active arial graft survival (especially for deceased donors), and
and vulnerable to ischemia where hypoxia reduces cel- although its incidence has decreased with modern immu-
lular metabolism and Na/K ATPase exchanger function. nosuppression, those rejection episodes remaining are more
Reperfusion injury from oxygenated blood generates ROS severe.
causing DNA breakdown, lipid peroxidation, apoptosis Other alloimmune risk factors for graft loss include recip-
and necrosis of tubular cells, and vascular endothelial cell ient sensitization and HLA matching (see Chapter x). Reg-
injury. Limiting procurement and perioperative ischemic istry graft survival is reduced by HLA mismatching, despite
injury may improve organ quality and ameliorate graft more potent immunosuppression. Pretransplant antibodies
immunogenicity. Better organ preservation techniques and specific to donor HLA antigens (DSA) can result from blood
solutions, optimal intensive care unit management, pulsa- transfusion, pregnancy/miscarriage, or prior transplanta-
tile ex vivo machine perfusion, rapid transfer of organs, and tion. Posttransplant de novo DSA formation correlates with
prompt implantation reduce organ stresses. Ischemic pre- allograft failure from chronic rejection in retrospective and
conditioning using vasodilators or antiapoptotic molecules prospective studies. A causal role for antibody-mediated
is investigational.55 graft loss is supported by microvascular inflammation with
Ischemic tubules can recover if sufficient residual tubu- C4d and transplant glomerulopathy on biopsy. Rarely, DSA
lar cells survive to replenish nephron losses, and the base- against nonclassical HLA antigens (e.g., endothelial cells
ment membrane remains intact. Repair mechanisms are as AECA; AT1 receptor; perecan, a heparin sulfate; MICA,
initiated by inflammatory and fibrogenic signals, causing a polymorphic nonclassical class I antigen; and basement
interstitial infiltration of fibroblasts, mononuclear cells, and membrane molecules, collagen, vimentin, Kα-tubulin) are
macrophages. Fibroblast proliferation with extracellular pathogenic.
matrix deposition remodels tissue contributing to the IF/ The effect of rejection depends on its type, timing, sever-
TA pattern: the sequela of tubular injury combined with ity, and persistence. When diagnosed and treated promptly,
an injury-repair response. IF/TA is accompanied by low- acute interstitial cellular rejection usually resolves without
level proteinuria, hypertension, allograft dysfunction, and sequelae. In contrast, episodes of vascular or steroid-resis-
reduced graft survival. tant rejection, recurrent rejection episodes, untreated SCR,
chronic-active CA-TCMR, or late (mixed) rejection causes
substantial allograft damage.10,58 Persistent interstitial CA-
EARLY TUBULOINTERSTITIAL DAMAGE
CMR increases later IF/TA, clinical or subclinical AMR may
Tubular cell injury during the early posttransplant period result in transplant glomerulopathy, and vascular rejection
results from ischemia-reperfusion injury with ATN, acute causes arterial vasculopathy.
severe rejection and persistent SCR, BKVAN, and CNI
nephrotoxicity, superimposed on donor disease. Acute SUBCLINICAL REJECTION
functional CNI nephrotoxicity causes tubular cell injury
characterized by isometric vacuolization, microcalcifica- SCR is defined as histologically proven acute or borderline
tion, and rarely, by cytoplasmic inclusion bodies (from giant rejection, but without concurrent functional deteriora-
abnormal mitochondria). The histologic features of tubular tion. It is only diagnosed by protocol biopsy and is clinically
atrophy are reduced tubular cell height, nuclear loss, and distinct from acute rejection, with functional impairment
tubular luminal dilation. Alloimmune mononuclear infil- prompting an indication-biopsy. It can be present in the
tration increases profibrotic factors including TGF-β and interstitial or microvascular compartments and mediated
the TIMP family of enzymes, further increasing interstitial by T cells or DSA, respectively. The reported variation in
fibrosis.26,56 prevalence relates to differences in transplant population,
The extracellular matrix is a dynamic network of HLA mismatch, prior acute rejection, ethnicity, baseline
proteins and proteoglycans, which accumulate from immunosuppression protocol, transplant era, and biopsy
increased synthesis and/or decreased breakdown. Medi- timing. The prevalence of acute subclinical TCMR (Banff
ators include TGF-β1, angiotensin, and immunosup- grade 1A) in 3-month protocol biopsy specimens ranged
pression. Cyclosporine increases profibrotic cytokines from 3% to 31%, with borderline SCR occurring in 11% to
(TGF-β1 and TIMP-1) in humans and in experimental 41%.10,59
models of CNI nephrotoxicity with interstitial fibrosis. Interstitial SCR has been associated with IF/TA, allograft
Angiotensin II blockade is a potential treatment target, dysfunction, and reduced graft survival59–61 (Fig. 27.6).
but it is not used in clinical practice. Cell cycle inhibi- SCR is followed by IF/TA mediated by TCMR and can cause
tors (including mycophenolate and mammalian target of persistent CA-TCMR with i-IFTA in some, mediated by lym-
rapamycin [mTOR] inhibitors) reduce myofibroblast pro- phocytes and activated macrophages, inflammatory medi-
liferation and collagen deposition in vivo and in experi- ators, and profibrotic signals (IL-1, IL-6, TNF-α, adhesion
mental chronic rejection. Sirolimus may limit tubular molecules, and TGF-β). More potent immunosuppression
atrophy, vascular hyperplasia, and interstitial fibrosis, is associated with fewer acute rejection episodes, reduced
although a confounding increase in early alloimmune SCR, and subsequent IF/TA in cohort studies.
injury (compared with potent CNI controls) make con- Several controlled trials have evaluated treatment of
clusions less certain.57 interstitial SCR. The first randomized controlled trial (RCT)
442 Kidney Transplantation: Principles and Practice

Fig. 27.7 BK virus nephropathy infecting a renal tubule. Tubular cells


Fig. 27.6 Subclinical rejection with interstitial lymphocytic infiltration are abnormal with some “ground-glass” nuclear changes, smudging,
with low-level tubulitis, but unchanged renal transplant function. tubular necrosis, and sloughing into the lumen, eventually forming uri-
nary decoy cells.

demonstrated that corticosteroid therapy significantly


decreased acute rejection episodes and 6-month IF/TA
scores and improved 2-year function and trend to 4-year
survival. The cyclosporine-based immunosuppression pro-
duced an SCR prevalence of 30%.62 A repeat trial using
tacrolimus in low-immune risk patients could not demon-
strate benefit of treatment (SCR prevalence was 4.7%).63
Another study using cyclosporine or tacrolimus (28% SCR
prevalence) showed better eGFR at 6 and 12 months with
treatment.64
Subclinical AMR rejection in protocol biopsies (with
microvascular inflammation and C4d) is more common
in desensitized positive-crossmatch or untreated sensitized
recipients, and after late AMR treatment.65,66 Sublethal
microcirculatory injury from circulating DSA and comple-
ment activates glomerular endothelial cells, produces a
widened subendothelial space with fibrillary deposition,
glomerular basement membrane (GBM) reduplication and
Fig. 27.8 Immunoperoxidase stain (SV40T) of BK-infected renal tubular
eventually, chronic transplant glomerulopathy. PTC multi- cells (brown) diagnostic of polyoma viral nephropathy.
lamination develops below PTC endothelial cells.65,67

BK VIRUS NEPHROPATHY and apoptosis or necrosis, then slough into the tubular
lumen—visualized as diagnostic urinary “decoy” cells.
BK virus is an endemic polyomavirus infection of high preva- Multifocal viral activation advances and provokes an
lence, low morbidity, and long latency that can reactivate in acute antiviral cytopathic inflammatory response of
immunocompetent individuals.68 After primary childhood monocytes, polymorphonuclear leukocytes, and plasma
infection, BK virus can establish a lifelong, subclinical infection cells, which resembles acute TCMR (but lacks arteritis,
within the renal cortex and medulla, and it can be transmit- C4d deposition, or HLA-DR expression).71 Chronic-active
ted within a transplanted kidney. Asymptomatic reactivation tubulointerstitial inflammation with persistent SV40T
occurs in 10% to 68% of recipients using CNI-based immu- and tubular necrosis ensues, and this is commonly fol-
nosuppression, where 1.1% to 10.3% progress to BKVAN.69 lowed by progressive nephron destruction and functional
Severe BKVAN with tubulointerstitial nephritis leads to pro- deterioration.71
gressive renal dysfunction and graft loss (5% incidence, where Suspicious pathology should be confirmed by tissue
46% fail).70 Polyomavirus allograft infection encompasses BK SV40T staining and viremia quantification (Fig. 27.8).
virus (but rarely JC virus cases). Asymptomatic BK viremia Characteristic 35 to 38 nm intranuclear paracrystal-
may occur within 2 to 6 months posttransplant, evolving into line viral arrays by electron microscopy are distinguished
a stage of renal impairment and clinical disease. from adenovirus (70–90 nm), cytomegalovirus (CMV),
Viral replication within tubular epithelial cells forms and enveloped herpes simplex (120–160 nm). Late viral
intranuclear inclusions, which enlarge with smudgy infection demonstrates chronic tubulointerstitial scarring,
nuclear chromatin, cellular atypia, and anisocytosis dystrophic microcalcification, and sometimes low-grade
(Fig. 27.7). Cells degenerate with rounding, detachment, (plasma cell rich) inflammation that is SV40T-negative.72
27 • Chronic Allograft Failure 443

PROGRESSIVE AND LATE-STAGE CHRONIC


ALLOGRAFT DAMAGE
Late phenotypical changes develop in the glomerular and
microvascular compartments, overlaying progressive tubu-
lointerstitial damage.10 Drivers for ongoing tubular injury
(see Fig. 27.1) include residual CA-TCMR, late acute rejec-
tion (often associated with DSA), CNI nephrotoxicity, BK
viral infection, and episodes of late AKI from an intercurrent
illness. Acute late rejection often results in severe inflamma-
tory tubular loss and initiation of persistent chronic inflam-
mation, followed by progressive renal dysfunction and graft
failure (see Fig. 27.1).10,45 The causes of late glomerular and
vascular disease includes CNI, chronic AMR with transplant
glomerulopathy, CA-TCMR, recurrent glomerulonephritis,
diabetic nephropathy, hypertensive nephrosclerosis, and
vascular effects of smoking and dyslipidemia (see Fig. 27.1).
Fig. 27.9 Early vascular changes of chronic antibody-mediated rejec-
tion in a small muscular artery. Intimal and endothelial cells are abnor-
CHRONIC-ACTIVE T-CELL-MEDIATED REJECTION mal with edema and early neointimal formation present. A small,
partially adherent thrombus is seen in the lumen.
The expanded definition of CA-TCMR now includes two
phenotypes: grade 1 as persistent interstitial mononuclear
infiltration with i-IFTA, and uncommonly, a vascular vari-
ant of arterial fibrointimal hyperplasia (grade 2).73
Interstitial CA-TCMR (type involves T cells, CD4+ or CD8+)
and macrophages in the interstitial compartment resulting
in tubulitis and inflammatory injury characterized by i-IFTA
(>25% i-IFTA areas with tubulitis). i-IFTA itself strongly pre-
dicts graft failure independent of renal function, IF/TA, and
inflammation scores, and associated with progressive IF/TA
with tissue injury/repair-associated molecular transcripts.
i-IFTA is preceded by TCMR and underimmunosuppression,
and it is followed by progressive IF/TA, fibrointimal hyperpla-
sia, renal dysfunction, and allograft loss.24,27,73–75 Persistent
i-IFTA is the pathologic consequence of CA-TCMR, although
it is a nonspecific phenotype of active tubular destruction
(where other causes such as BKVAN, pyelonephritis, and
obstruction should be excluded).
Vascular chronic TCMR (grade 2) presents as medial
fibrointimal hyperplasia within small muscular arteries, Fig. 27.10 More advanced subacute vascular changes with extensive
associated with focal destruction of the internal elastic neointimal formation (within the internal elastic lamina boundary)
characterized by invading myofibroblasts, deposition of matrix pro-
lamina, smooth muscle infiltration forming a “neointima” teins, collagen, and edema, resulting in near occlusion of the vascular
with vascular narrowing (the Banff cv score; Figs. 27.9 lumen. Masson trichrome stain.
and 27.10). Donor disease, past vascular rejection, hyper-
lipidemia, hypertension, and smoking also cause similar
arterial changes (distinguished by subendothelial hyalino- all produce a stereotypical constellation of renal histologic
sis, elastic lamina reduplication, and medial hyperplasia abnormalities. The classically described histologic features
in small arteries on histology and by clinical verification; of CNI nephrotoxicity include de novo or increasing arte-
Fig. 27.11) riolar hyalinosis (Fig. 27.12), striped cortical fibrosis (Fig.
27.13), isometric tubular vacuolization (Fig. 27.14), and
tubular microcalcification (unrelated to other causes, such
CALCINEURIN INHIBITOR NEPHROTOXICITY
as ATN or hyperparathyroidism). Other reported abnormal-
The introduction of cyclosporine revolutionized kidney ities include peritubular and glomerular capillary conges-
transplantation, markedly improving 1-year graft survival tion (diagnostically unreliable), diffuse interstitial fibrosis
rates and permitting transplantation of nonrenal solid (important but nonspecific), toxic tubulopathy (in high-
organs. CNIs are generally well tolerated and underpin dose cyclosporine therapy), and juxtaglomerular hyper-
modern immunosuppression regimens (see Chapters 17), plasia (rare and nonspecific). Tacrolimus and cyclosporine
but are pleomorphic nephrotoxins, causing multiple histo- show similar histology, although lesion frequencies vary by
logic abnormalities—a significant diagnostic and manage- CNI. Mild acute arteriolopathy, striped interstitial fibrosis,
ment issue for long-term use. glomerular congestion, and tubular microcalcification are
CNI exposure to patients with autoimmune diseases, more frequent with cyclosporine (compared with tacroli-
nonrenal transplants, or rodent models of nephrotoxicity mus). Chronic moderate to severe arteriolar hyalinosis was
444 Kidney Transplantation: Principles and Practice

Fig. 27.11 Chronic fibrointimal hyperplasia in chronic rejection


(severe) with concentric layers of smooth muscle cells and collagen and
near occlusion of the vessel.

Fig. 27.13 Striped fibrosis. Demarcated areas of striped fibrosis near


adjacent normal cortex associated with interstitial fibrosis. Masson tri-
chrome stain stains collagen green.

Fig. 27.12 Arteriolar hyalinosis with a large, eccentrically located nod-


ule within the media of the arteriole.

comparable between CNI and very common by 10 years


Fig. 27.14 Isometric vacuolation in the proximal tubular cells from
posttransplant, being irreversible once established. Tacro- cyclosporine tubulopathy.
limus offers more potent immunosuppression with compa-
rable nephrotoxicity.76
The pathologic diagnosis of CNI nephrotoxicity is hin- cristae are rare. Chronic diffuse tubulointerstitial damage
dered by the paucity of specific and reliable diagnostic mark- from CNI is common in humans and experimental models
ers. Striped fibrosis (cortical fibrosis demarcated against (but diagnostically nonspecific).
normal adjacent cortex in a striped pattern) likely originates The most reliable diagnostic abnormality is de novo or
from watershed infarction of interlobular or arcuate arter- increasing arteriolar hyalinosis, classically described as
ies, but lacks diagnostic sensitivity and specificity (see Fig. peripheral and nodular pattern (vs. subendothelial and
27.13). Tubular microcalcification is described in chronic diffuse), but imperfect. CNI arteriolopathy is from arte-
cyclosporine nephrotoxicity, but can also be secondary to riolar smooth muscle cell loss and replacement by hyaline
residual hyperparathyroidism or localized cellular necrosis deposits. Early acute arteriolar hyalinosis is often mild and
from any cause (including severe rejection and ATN). Iso- patchy, and reversible with dosage reduction, but alterna-
metric vacuolation of proximal tubular cells (corresponding tively, may be from age-related donor disease. Late arterio-
to dilated and stressed endoplasmic reticulum in proximal lar hyalinosis lesions are more severe and less reversible,
straight tubules) was frequent with high-dose cyclosporine, with microvascular narrowing causing ischemic glomeru-
and it can still represent acute CNI toxicity (although tubu- losclerosis and associated IF/TA78 (Fig. 27.15). Hyalinized
lar vacuolation also occurs in ATN). Cytoplasmic inclusion arterioles are unable to maintain their structural integrity,
bodies from abnormal giant mitochondria with deranged collapsing inwards with luminal narrowing and reduced
27 • Chronic Allograft Failure 445

Fig. 27.16 Transplant glomerulopathy by methenamine silver stain


Fig. 27.15 Severe arteriolar hyalinosis of the feeding afferent arteriole light microscopy, showing double contours of the glomerular capillary
leading to collapse of the glomeruli from calcineurin nephrotoxicity. loops (seen as a parallel pair of lines).

blood flow (to 20% of normal). Arteriolar hyalinosis corre- or endothelial cell injury from CNI, hypertension, or alloim-
lates with downstream glomerulosclerosis from ischemia.76 mune injury results in vascular narrowing of afferent arte-
Arteriolar hyalinosis on biopsy is assessed for a nodular- rioles, causing downstream ischemic glomerulosclerosis.
ity (imperfect for CNI etiology) and histologic progression Glomerular ischemia and podocyte injury may also lead to
(compared with prior or baseline histology; see Fig. 27.12). proteinuria and glomerulosclerosis.
Donor disease, ischemic arteriolar injury, dyslipidemia, Severe tubular injury can disconnect the downstream
diabetes mellitus, and hypertensive vascular disease are proximal tubule from its perfused glomerulus, designated
alternative causes of hyalinosis.77–79 Categorizing arterio- as “atubular glomeruli.” They are often small or contracted
lopathy by circular or noncircular involvement marginally within an enlarged glomerular cyst surrounded by peri-
improved reproducibility, but at the expense of usable clini- glomerular fibrosis. Bowman’s space is filled by inspissated
cal grading.77,80 Even nodular arteriolopathy (thought to proteinaceous material from residual glomerular filtration
be specific to CNI toxicity) can be observed in 28% with- and local reabsorption.85 Atubular glomeruli are common
out cyclosporine exposure (and is likely related to vascular in native tubulointerstitial kidney diseases (e.g., lithium
aging or other causes).81 and cisplatin nephrotoxicity), and comprise 1% to 2% of
CNI nephrotoxicity with hyalinosis and glomeruloscle- glomeruli from living and deceased donor implantation
rosis is a common secondary diagnosis (in 30% of indica- biopsies, 17% to 18% in CAN, and 29% with cyclosporine
tion biopsies),82 and it becomes increasingly common after nephrotoxicity.85
chronic CNI exposure and use of older donor kidneys. The
contribution of CNI nephrotoxicity to chronic graft injury TRANSPLANT GLOMERULOPATHY AND
is suggested by unchanged long-term graft attrition rates CHRONIC-ACTIVE ANTIBODY-MEDIATED
despite suppression of early acute rejection, appearance of REJECTION
characteristic histologic lesions from longitudinal histopa-
thology studies,76,78 and by data from clinical trials of CNI Chronic transplant glomerulopathy is the principal mor-
avoidance, early withdrawal and dose reduction, or late phologic expression of persistent CA-AMR within the glom-
withdrawal; which demonstrate functional, structural, and erulus.86 Its features by light microscopy include capillary
survival benefits.83,84 activation, capillaritis, double contour formation on silver
staining, and mesangial interposition (Figs. 27.16 and
27.17). It is semiquantitated, scored by the Banff schema
PROGRESSIVE GLOMERULAR ABNORMALITIES
by worst involved peripheral capillary loop (designated cg),
Microvascular and glomerular compartments are becom- and reported in 5% to 15% of failing and failed grafts.87–90
ing more commonly affected in late histology with wors- Electron microscopy (EM) demonstrates glomerular endo-
ening arteriolar hyalinosis and severe vascular narrowing thelial swelling and activation, subendothelial widening
from CNI nephrotoxicity, diabetes mellitus, hypertension, with deposition of flocculent or fibrillary material GBM
dyslipidemia, smoking, or vascular aging. Glomerular duplication, and mesangial matrix expansion (and desig-
abnormalities include ischemic glomerular loss, formation nated cg1a if only on EM; Fig. 27.18) Transplant glomeru-
of atubular glomeruli formation, recurrent glomerular dis- lopathy is associated with proteinuria, renal impairment,
ease, and transplant glomerulopathy (see later). and reduced graft survival.56,89
Morphometric analysis of CAN finds two separate popu- Transplant glomerulopathy may be accompanied by
lations of glomeruli. Smaller glomeruli display ischemic glomerular inflammation (by monocytes/macrophages,
wrinkling and extracapillary fibrotic material are con- natural killer [NK] cells, and activated T cells), and endo-
trasted with larger, hyperfiltering glomeruli. Late vascular thelial C4d deposition (in glomeruli and/or PTCs), or
446 Kidney Transplantation: Principles and Practice

Fig. 27.19 Peritubular multilamination of the basement membrane


in chronic antibody-mediated rejection (transplant glomerulopathy).
Note multiple layers of reduplicated basement membrane.
Fig. 27.17 Light microscopic appearance of transplant glomerulopa-
thy showing increased mesangial matrix, thickened capillary loops, and
partial closure of the capillary loops. glomerulonephritis (excluded by serology, viremia, and
absence of GBM deposits). A T-cell-mediated glomerulitis
with typical glomerulopathy is rarely reported.
Multilamination (or multilayering) of PTC basement
membranes (Fig. 27.19) is the counterpart of CA-AMR,
occurring within PTC from DSA and visualized by EM.
Endothelial cell thickening with fenestrae loss reflect
endothelial injury, with excess production of basement
membrane forming additional layers, as multilamination.
Normally, the PTC endothelium sits on a single (occasion-
ally double) layer of basement membrane. Multilamination
occurs in 38% of failed transplants from “chronic rejection.”
Moderate disease is five to six layers, a severe abnormality
is seven or more layers. Minor (three layers) multilamina-
tion may signify antibody; however, its specificity at this
threshold is uncertain: it also occurs in native kidney dis-
eases (obstructive uropathy, analgesic nephropathy, radia-
tion nephritis, reflux-dysplastic syndrome, and thrombotic
microangiopathy).95
Endothelial C4d is a marker of activation of the classical
complement cascade by deposited DSA, appearing in glo-
Fig. 27.18 Subendothelial fibrillary material in transplant glomerulop- merular capillaries and PTCs. Its prevalence in PTCs var-
athy by electron microscopic examination.
ies from 36% to 91% with transplant glomerulopathy,89
12% to 61% diagnosed with chronic rejection and renal
dysfunction,91,96 and 2% of protocol specimens.97 AMR
circulating DSA (predominantly anti-HLA antibodies).91,92 without C4d occurs in 20% to 30% (termed “C4d-nega-
The molecular signature of AMR finds markers of NK cells tive,” although C4d immunohistochemistry may be mod-
(e.g., FGFBP2 and GNLY) and endothelial cell genes (e.g., erate or strongly positive in glomerular capillaries).98 The
ROBO4, DARC).93 Late chronic AMR cases with de novo prevalence of C4d varies by center, clinical scenario, meth-
DSA and transplant glomerulopathy often display pro- odology, and definition of C4d positivity (Fig. 27.20). Occa-
teinuria, mixed histology (with cellular interstitial T cell sionally, linear C4d staining occurs without microvascular
infiltration), and more IFNγ-inducible, NK and T cell tran- inflammation (glomerulitis or “PTCitis”) and is of uncertain
scripts (fewer AKI transcripts) relative to early AMR from significance (DSA should be checked). ABO-incompatible
preformed DSA.94 allografts commonly display harmless C4d staining (and is
The differential diagnoses of transplant glomerulopathy ignored). Testing for anti-HLA DSA is negative in 10% to
include thrombotic microangiopathy, which may produce 15% of CA-AMR cases (usually a false negative, occasion-
similar glomerular histology and requires clinical exclusion ally from non-HLA DSA).
by blood film examination, haptoglobin levels, lactate dehy- Banff criteria require all three primary criteria be met to
drogenase levels (differential diagnosis includes infection, confirm the diagnosis of CA-AMR (see later); however, an
recurrent atypical HUS, and anticardiolipin antibody throm- incomplete clinicopathologic expression of AMR and CA-
botic microangiopathy), and hepatitis C mesangiocapillary AMR is common in clinical practice.89
27 • Chronic Allograft Failure 447

Fig. 27.20 Immunoperoxidase stain for C4d in glomerular capillary


loops and peritubular capillaries in chronic antibody-mediated rejection.
Fig. 27.21 Recurrence of diabetic nephropathy in a renal transplant
showing thickened tubular basement membranes and diffuse diabetic
The diagnostic triad of CA-AMR in the context of organ glomerulopathy with massively increased mesangial matrix and thick-
ened glomerular basement membrane (green).
dysfunction includes the following features.73
  

1. Morphology of chronic tissue injury as either:


transplant glomerulopathy (double contours on light
□ 

microscopy Banff score cg1 or by EM score cg1a, sup- glomerulonephritis is diagnosed by exclusion of de novo
ported by activated glomerular endothelial cells and/ and donor-transmitted disease, and it contributes to 8.4%
or widened subendothelial space containing fibrillary of allograft losses by 10 years.99 The relative effect of recur-
material); rent glomerulonephritis is greater in some populations
severe PTC basement membrane multilamination (by
□  where primary glomerulonephritis is prevalent or severe,
EM); and broadly, as graft survival lengthens. Its clinical course
or new-onset fibrointimal hyperplasia (especially leu-
□  and severity often recapitulates the original disease.100
kocytes within sclerotic intima); Patients with vasculitis or lupus nephritis are normally
unexplained acute thrombotic microangiopathy or
□  controlled by transplant immunosuppression (medication
acute tubular injury without apparent cause. noncompliance needs review).101 Focal segmental glo-
2. Current or recent antibody interaction with vascular merulosclerosis (recurs in 20%–50%) and dense deposit
endothelium as either: disease (50%–90% recurrence) have the worst prognosis.
linear C4d deposition in peritubular capillaries (“focal”
□  Membranous glomerulonephritis recurs in 29% to 50%,
C4d1 score as 10% positive by immunohistochemis- membranoproliferative glomerulonephritis type 1 recurs
try or C4d2/C4d3 by more sensitive immunofluores- in 20% to 33% (reclassified as a C3 glomerulopathy with
cence), as the “footprint” of classical complement acti- IgG), and IgA nephropathy recurs in 58% (with minimal
vation provoked by antibody; early effect but increased with time).100 Diabetic glomer-
microvascular inflammation (MVI) comprising mononu-
□  ulopathy recurs with variable clinical expression (Fig.
clear cell adhesion within capillaries as either glomerulitis 27.21).
and/or peritubular capillaritis (summed g+ptc scores ≥2);
validated molecular markers associated with antibody-
□ 
LATE ACUTE REJECTION AND INTERCURRENT
mediated endothelial injury (e.g., endothelial genes, DSA ILLNESSES
or AMR) as a related gene-set or mathematically derived
diagnostic classifier. These significantly strengthen the Acute rejection occurring beyond 3 months posttrans-
diagnosis of AMR (independently of C4d and DSA re- plantation presents with subacute or acute renal dys-
sults), but are of limited availability and not approved function. It usually follows medication noncompliance
by regulatory bodies. Molecular technologies applicable or iatrogenic underimmunosuppression (e.g., after
to formalin-fixed paraffin-embedded tissue optimized to severe infection or cancer diagnosis). Biopsy usually
derive a molecular signature are under development. shows established mixed rejection with extensive inter-
3. Circulating donor-specific antibody to donor HLA epit- stitial lymphocytic infiltration, IF/TA, and glomerulo-
opes or other endothelial antigens. sclerosis, frequently accompanied by markers of AMR
(circulating DSA, usually class II, endothelialitis, and tis-
sue C4d expression) and/or vascular rejection. Late AKI
RECURRENT GLOMERULAR DISEASE
can follow medical or surgical events (including myocar-
Glomerular disease (including glomerulonephritis and dial infarction, sepsis, or abdominal emergencies), where
diabetes) causes most end-stage renal failure, and some AKI is the dominant acute histologic diagnosis, but often
allografts develop recurrence of these diseases. Recurrent with chronic background changes.
448 Kidney Transplantation: Principles and Practice

Approach to a Failing Allograft formulas, although inexpensive and simple, are imperfect
compared with the more expensive and accurate isotopic
The clinical approach to a patient with a failing kidney GFR methods. Errors relate to differential creatine genera-
transplant involves the integrated synthesis of clinical tion (e.g., muscle loss from corticosteroids, malnutrition,
history and examination, biochemical information, renal and sepsis), variable tubular secretion of creatinine, the
imaging, and histopathology (Table 27.3, Fig. 27.22) to nonlinear relationship with GFR, and inaccuracies and lab-
allow formulation of a clinicopathologic diagnosis con- oratory differences in biochemical measurement (modified
taining the etiologic cause of dysfunction, potential revers- Jaffe method or enzymatic measurement). Serum creatinine
ibility, and evaluation of intrinsic structural damage. An underestimates extent of nephron losses, and early biopsy
accurate etiologic diagnosis then allows formulation of should be considered before severe and irreversible loss of
potential treatment strategies. function has occurred.

MONITORING OF RENAL FUNCTION PROTEINURIA AND URINALYSIS


Transplant renal dysfunction depends predominantly on Proteinuria is a powerful and independent adverse risk fac-
nephron loss reflected by IF/TA, with a lesser effect from tor for graft and patient survival, which represents several
percentage glomerulosclerosis and intrinsic glomeru- diagnostic groupings, including transplant glomerulopathy,
lar abnormalities. Serum creatinine and calculated GFR recurrent FSGS, and glomerulonephritis (causing glomeru-
lar proteinuria). Severe IF/TA results in modest tubular pro-
teinuria from impaired tubular resorption of filtered proteins.
TABLE 27.3 Clinical Investigation of Chronic Transplant Proteinuria increases with uncontrolled hypertension,
Dysfunction glomerular hyperfiltration, morbid obesity, and mTOR inhib-
itor therapy; it is reduced by renin-angiotensin blockade, CNI
1. Clinical examination for blood pressure, hydration status, kidney therapy at therapeutic levels, renal ischemia, and poor trans-
transplant palpation, and auscultation for bruit
2. Repeat serum creatinine after adequate hydration and elimination
plant function, per se. Residual proteinuria originating from
of any extrinsic nephrotoxins (e.g., NSAID or COX-2 inhibitors) native kidneys may obscure interpretation; however, this
3. Review blood levels of calcineurin inhibitor for toxicity (higher usually declines within the first few months after transplan-
levels suggest nephrotoxicity vs. low or variable levels indicating tation. Proteinuria that fails to decrease or increases post-
inadequate dosing or noncompliance with risk of rejection) transplant (quantified by serial urine protein-to-creatinine
4. Renal transplant ultrasound to evaluate arterial blood flow and
exclude ureteric obstruction. MAG3 scan when ultrasound incon- ratio) occurs in 31% to 45% and predicts a worse prognosis.
clusive Persistent, increasing or de novo high-grade or nephrotic-
5. BK evaluation by serum nucleic acid testing, with quantification if range proteinuria, or hematuria with proteinuria should
positive (or urinary “decoy” cells if otherwise unavailable) prompt a diagnostic biopsy glomerulonephritis.
6. Urinalysis for proteinuria and hematuria (suggests glomerulone-
phritis or transplant glomerulopathy)
7. Transplant biopsy for diagnosis of different subtypes of acute RENAL TRANSPLANT IMAGING
and chronic rejection, IF/TA, calcineurin nephrotoxicity, recurrent
glomerulonephritis, or BK virus nephropathy Diagnostic ultrasonography is important for initial evalua-
tion of transplant dysfunction, where it can exclude ureteric

Elevated serum creatinine Pattern: Acute, subacute, or chronic

Clinical evaluation Fluid push


Dehydration/diarrhea IV fluids or oral hydration Serum creatinine
Sepsis/fever & repeat creatinine remains elevated
Hypotension
Nephrotoxins CNI level high Reduce CNI dose
Acute nephrotoxicity & repeat creatinine
MSU: MC&S With:
CNI level low Possible late Transplant C4d
Variable / absent acute rejection biopsy SV40T
Noncompliance check DSA IF
EM
Transplant Urinalysis Establish
ultrasound hematuria Possible glomerulonephritis etiological
for obstruction proteinuria or transplant glomerulopathy diagnosis

Obstruction: BK virus NAT Possible BKVAN Treat pathophysiological


urological management >10,000 copies/mL cause of dysfunction

Fig. 27.22 Clinical workflow of diagnostic evaluation of a transplant recipient with an elevated serum creatinine. Clinical history, examination, urinaly-
sis, and other investigations, such as transplant imaging and diagnostic biopsy, are often undertaken in parallel without undue delay.
27 • Chronic Allograft Failure 449

obstruction and surgical complications, evaluate the cortical aminopeptidase, γ-glutamyl transpeptidase, and alkaline
parenchymal quality, and evaluate the allograft’s vascular phosphatase), α1-microglobulin, N-acetyl-d-glucosamini-
supply (by Doppler) for cortical hypoperfusion or infarction dase (NAG), and neutrophil gelatinase-associated lipocalin
(e.g., from a thrombosed polar artery or venous thrombosis), (NGAL), are markers of proximal tubular injury. Urinary
or renal artery stenosis. Ultrasound imaging is suboptimal for β2-microglobulin strongly associates with acute rejection.
the diagnosis of acute rejection (features include increased Urinary mRNA levels of FOXP3 (a specific marker for Treg),
renal volume, reduced cortical echogenicity, loss of cortico- were increased with acute rejection compared with CAN
medullary differentiation, and splayed medullary pyramids) and normal biopsies (lower levels identified graft failure
or chronic allograft damage. The chronic changes of irregu- risk), but overlap between groups limited clinical utility.106
lar cortical outline, reduced cortical width, increased paren- Advanced proteomic methods to detect biomarkers of rejec-
chymal echogenicity, and loss of corticomedullary junction tion, including cytokines and their receptors, are under
differentiation are late features. evaluation (see later).
The Doppler resistance index (RI) is a noninvasive mea- Urinary mRNA derived from sloughed tubular cells or
sure of intrarenal compliance calculated from early seg- excreted inflammatory cells appear more specific for intra-
mental transplant arteries as the index of peak systolic blood renal events compared with proteinuria. mRNA is mea-
velocity (Vmax) relative to the minimal diastolic velocity sured by PCR, microarray, or RNA-seq methods, but it
(Vmin), expressed as 1 − (Vmin/Vmax). Higher RI values degrades quickly in urine. Acute rejection correlates with
imply decreased diastolic blood flow and augmented down- multiple molecules, including cytotoxic T lymphocyte
stream vascular resistance, correlating with intraabdominal markers, CD3 (a T cell marker), CD103 (CD8 cytotoxic T
pressure, older age, and pulse pressure profile (and inversely lymphocyte intraepithelial homing marker), perforin, gran-
with pulse rate). An RI exceeding 0.80 correlates with renal zyme A, TIM-3, and CD25 (T cell activation markers), che-
impairment, graft failure, and fractional interstitial fibrosis mokine receptor CXCR3, and ligand interferon-inducible
on biopsy.102,103 Renal transplant angiography of chronic protein 10 (IP10 or CXCL10), interferon-γ, and chemokine
rejection demonstrate severe (“pruned”) vascular attenu- CXCL9, but not with chronic rejection or normal biopsy
ation with rejection. Power Doppler or contrast-enhanced results.106–109 Urinary TGF-β, detected by coculture with
ultrasound quantify reduced intragraft blood flow with luciferase-expressing cells (or by TGF-β1 mRNA), increased
parenchymal damage. Phase-sensitive, two-dimensional with chronic renal rejection or CAN. A three-gene mRNA
speckle tracking or sonic elastography also shows altered signature of CD3ε, CXCL10, and 18S RNA levels, sepa-
parenchymal properties with fibrosis and IF/TA; however, all rated acute TCMR from nonrejection (AUC 0.85, sensitivity
techniques are relatively insensitive for early damage. 79%, specificity 78%) and distinguished acute TCMR from
Other investigational techniques include magnetic borderline and AMR. It became abnormal several weeks
resonance imaging (MRI) for quantification of allograft before clinical presentation and appears the most promis-
volume loss, microstructural changes, and blood flow altera- ing potential biomarker.109
tions reflecting parenchymal damage. T1-weighted pulse An alternative to mRNA is microRNAs (miRNAs), which
sequences can distinguish acute rejection from CNI nephro- are short (18–22 nucleotides) RNA molecules that bind to
toxicity using intensity differences at the corticomedullary and inhibit mRNA, down-regulating protein translation.
demarcation are sensitive for cortical disease but nonspecific, They are not translated into proteins (noncoding). A major
with poor biopsy diagnosis correlations.104 MRI of acute advantage over mRNA is their remarkable stability, surviv-
rejection displays lower cortical enhancement and delayed ing in blood, urine, or paraffin tissue blocks. Circulating
renal excretion. Gadolinium-enhanced dynamic turbo fast- miRNAs are released after cell death or sublethal injury as
low-angle-shot (FLASH) imaging, gadolinium MRI perfu- larger shedding microvesicles (100 nm to 1 μm), or actively
sion, and blood-oxygenation-level-dependent (BOLD) MRI secreted smaller exosomes (30–120 nm diameter). Acute
(reduced medullary perfusion distinguishes acute rejection rejection initiates substantial alterations in mRNA and
from ATN), gadolinium-free diffusion-weighted imaging, their corresponding miRNA targets, originating from infil-
contrast-free arterial spin labeling have been tested in trans- trating cells and injured parenchyma. miRNA alterations in
plantation. MRI offers high-contrast resolution without radi- rejection differed from ischemic injury and IF/TA (panels of
ation and with fair sensitivity for rejection but suboptimal miRNAs were superior). More technical and clinical studies
specificity. Positron emission tomography (e.g., 18F-FDG) are needed before their use as urinary biomarkers.110,111
coregistration with low-dose computed tomography (PET/
CT) can detect infiltration of activated leukocytes in experi- Immune Surveillance Markers in Blood
mental TCMR, but lacks specificity in human allografts.105 Noninvasive markers of renal immune activity are being
developed as potential replacements for an invasive trans-
IMMUNE SURVEILLANCE TESTS plant biopsy. Assays of activated lymphocytes secreting
IFN-γ correlate with dysfunction112 and mitogen-stimu-
Urinary Diagnostics lated CD4 T cell reactivity,113 and quantify the risks of infec-
Urinary protein biomarkers have been advocated to detect tion and rejection; however, diagnostic overlap, moderate
disease earlier than serum creatinine, with superior speci- specificity, and lack of independent validation obscures
ficity than blood testing. Most tests (see later) are at an their clinical value. Serum neopterin (from activated
investigational stage and not yet ready for routine clinical macrophages) is a sensitive marker of acute alloimmune
practice. activity, but nonspecific and elevated in CMV infection.
Urinary excretion of low molecular weight proteins Serum-soluble CD30 levels (a Th2 marker) correlated with
(including β2-microglobulin) or tubular enzymes (alanine chronic rejection but lacked specificity (being increased by
450 Kidney Transplantation: Principles and Practice

infection and altered by CNI therapy). These concerns of chrome or Sirius Red staining detects collagen and is
specificity and technical issues limit their current use. used in research settings.
Gene transcripts of circulating mononuclear cells have 5. Implantation or postperfusion biopsy specimens are
been evaluated as potential biomarkers. Gene-expression needed to distinguish preexisting donor pathology from
profiling of mononuclear cell samples collected with a newer changes in subsequent samples. A temporal
diagnostic kidney biopsy found increased IL-4, IL-5, IL-6, sequence of changing histology should be interpreted
IFN-γ, perforin and granzyme B mRNA, Fas ligand, and with clinical events and therapy.
the costimulatory molecule OX4O in small studies of acute 6. The tissue from a chronically failing graft should be pro-
rejection.107 Many of these “omic” technologies lack the cessed with additional special techniques. Light micros-
specificity and sensitivity for early disease detection and copy assesses the presence, extent, and grade of chronic
are beset by methodologic limitations, cost, inadequate IF/TA and nephron loss, and it helps define specific etio-
standardization, and validation by prospective multicenter logic diagnoses (e.g., acute and chronic rejection, CNI
trials. The transplant kidney biopsy still remains the most nephrotoxicity, hypertensive nephrosclerosis, BKVAN,
reliable diagnostic test (see later). or transplant glomerulonephritis). Periodic acid–Schiff
stain highlights arteriolar hyalinosis, silver stains
detect double contours of transplant glomerulopathy,
KIDNEY TRANSPLANT BIOPSY and trichrome stains collagen. Immunofluorescence or
Principles Guiding Clinically Indicated Biopsy immunoperoxidase techniques are usually negative or
nonspecific, but they help diagnose glomerulonephritis,
Chronic allograft deterioration is best evaluated by diagnos-
allograft viral infection (e.g., BK virus for SV40T or CMV
tic transplant histology (see Table 27.3; Table 27.4), and
stains), or CA-AMR (C4d deposition). EM detects early
biopsy is recommended with the following caveats:
   transplant glomerulopathy or deposits of glomerulone-
1. Biopsy after exclusion of clinical causes of acute dys- phritis.
function (e.g., obstruction hydronephrosis, dehydration 7. Provide adequate clinical information to the patholo-
with AKI, graft pyelonephritis, see later). gist, such as function, donor quality, relevant events
2. Biopsy early: late histology is often nonspecific (hamper- (such as DGF or prior acute rejection with treatments);
ing a specific etiologic diagnosis), and established neph- cause of recipient end-stage renal failure; and presence
ron loss is irreversible and less responsive to potential of proteinuria or BK viremia. A collaborative clini-
therapy. copathologic diagnosis formulated toward etiologic
3. Ensure an adequate sample for histological assessment causes of graft dysfunction should be made (see Table
(containing at least 10 glomeruli and two arteries) and 27.4).
preferably from two cores of cortex (some features are
patchy). Samples also should include arterioles (defined Risk and Safety of Kidney Transplant Biopsies
as two or fewer muscle layers and absent/incomplete Transplant needle core biopsy has an excellent safety profile
internal elastic lamina) for assessment of CNI-induced with a low risk of graft loss or major morbidity. Major com-
hyalinosis, and muscular arteries for immune-medi- plications, such as macroscopic hematuria with ureteric
ated fibrointimal hyperplasia (Banff cv). Glomerular obstruction, peritonitis, or graft loss, approximates 1%.
and microvascular abnormalities provide important Minor complications reported are gross hematuria in 3.5%,
diagnostic clues. IF/TA is appreciated easily on smaller perirenal hematomas in 2.5%, and asymptomatic arterio-
samples, reflects summated nephron loss, and correlates venous fistulas in 7.3%.114 The risk of graft loss from proto-
with GFR, guiding therapy and salvageability. Some old col biopsy is 0.03%. Risk is increased with indication-driven
transplanted kidneys can be surrounded by a fibrotic biopsy, with extraperitoneal or a transperitoneal position-
capsule that needs careful penetration by biopsy. ing of adult kidneys transplanted into infants, or with larger
4. Fibrosis (and IF/TA) may be problematic to quantify in needles (exceeding 18 gauge). Safety should be maximized
a standardized fashion, especially if patchy (e.g., striped with a skilled operator employing real-time ultrasound
fibrosis or variably diffuse IF). Image analysis using tri- guidance and an automated gun.

TABLE 27.4 Clinical Scenarios and Kidney Transplant Pathology


Clinical Disease Key Defining Features Scenario and Associated Features
Extended or marginal donor (ECD) Arterial and arteriolar vascular disease, glomerulosclerosis DGF donor IF/TA
Early ischemia-reperfusion injury Acute tubular necrosis edema or TA DGF oliguria
Subclinical TCMR Interstitial infiltration of mononuclear cells with tubulitis Stable GFR, late IF/TA
Subclinical AMR Microvascular inflammation C4d deposition Stable GFR, late chronic AMR
Circulating DSA Glomerulopathy
Chronic TCMR iIFTA and tIFTA fibrointimal hyperplasia ↓ GFR late IF/TA
Chronic AMR Transplant glomerulopathy double contours, mesangial ↓ GFR hypertension, proteinuria, noncompliance
matrix PTC-BM ML (by EM) C4d+ (may be negative)
donor specific antibody
CNI nephrotoxicity Progressive arteriolar hyalinosis, glomerulosclerosis, IF/TA ↓ GFR striped fibrosis
BK virus nephropathy Inflammatory tubular necrosis Blood BK NAT+
27 • Chronic Allograft Failure 451

DIAGNOSTIC ALGORITHM FOR A CHRONICALLY TABLE 27.5 Management of Chronic Allograft


FAILING GRAFT Deterioration
A gradually deteriorating serum creatinine should be evaluated PREVENTION AND SCREENING
by initial exclusion of reversible causes such as dehydration Minimize ischemia-reperfusion damage (shortest ischemic times;
(fluid state examination, history of diarrhea, or diuretic use), optimal procurement, preservation, and transport measures)
acute CNI nephrotoxicity (doses and blood levels), nephrotoxins Minimize donor-recipient histoincompatibility (especially class II)
Rapid histologic diagnosis and effective treatment of acute rejection
(e.g., ACE inhibitors, angiotensin receptor II blockers, NSAIDs, Early optimal immunosuppression (including early CNI levels and IL2R
or COX-2 inhibitors), recurrent glomerular disease (urinalysis blocker with medium to high immune risk recipients)
for hematuria and proteinuria), and ureteric obstruction or Control of early subclinical rejection (first 3–6 months)
vascular impairment (by Doppler ultrasound imaging). Early prophylaxis for CMV with (val)ganciclovir or valacyclovir
Transplant biopsy can provide a specific etiologic diagnosis Early BK screening
Regular monitoring of renal function (with serum creatinine)
of an acute or chronically failing graft and is for rational treat- Interval urinalysis and imaging (for development of recurrent glo-
ment planning directed toward the underlying pathophysi- merulonephritis or late ureteric obstruction)
ologic cause(s). Because IF/TA is the final outcome of multiple Regular follow-up for compliance review (clues include missed follow-
prior insults, assigning a specific etiologic diagnosis can pres- up appointments)
ent a practical difficulty if the allograft is severely damaged. CONTROL OF PROGRESSION FACTORS
The principal cause(s) of transplant deterioration should Control hypertension (calcium channel blockers ideal but theoretical
be classified as either (1) nonimmune causes of tubular advantages for ACEI or ARB therapy, careful diuretic use may be
injury (e.g., early ischemia-reperfusion damage with delayed needed)
Avoid added salt, stop smoking, control lipids, limit weight gain, life-
function or donor disease); (2) alloimmune causes (acute style modifications (reduction risk of metabolic syndrome)
TCMR, acute AMR, CA-AMR, CA-TCMR or mixed rejection Control diabetes and urinary tract infections
patterns); or (3) a specific diagnostic entity (e.g., recurrent Consider minimization of long-term CNI (by dose reduction, elimina-
glomerulonephritis, BKVAN, CNI nephrotoxicity, hyperten- tion, or substitution) in low immune risk recipients or with chronic
sive nephrosclerosis). Many late-indication biopsies show allograft injury or development of CNI nephrotoxicity (monitor any
medication conversions for acute rejection)
mixed rejection, secondary to noncompliance or iatrogenic Avoid underimmunosuppression (risk of subclinical, chronic, or acute
underimmunosuppression. The dominant pathologic cause late rejection)
of graft dysfunction should be reported, along with other Match the level of immunosuppressive therapy against individual
secondary diagnoses. Pointers for primary treatment assign- immunologic risk of rejection (from HLA-mismatch or eplet load
scores, prior rejection history, presence of DSA, regraft recipient,
ment (either increasing or decreasing immunosuppression) patient reliability, and compliance)
should include recent graft pathology (alloimmune histo-
logic clues include fibrointimal hyperplasia of small arteries, DIAGNOSE AND MANAGE INTERVAL ACUTE DETERIORATION IN
FUNCTION
interstitial lymphocytic infiltration, lymphocytes in peritu-
Manage acute recipient events (such as acute kidney injury from
bular capillaries, transplant glomerulopathy, or positive C4d severe life-threatening sepsis) with transient immunosuppression
staining), the recipient’s prior rejection and noncompliance minimization (keeping corticosteroids used), followed by restitu-
history, and circulating DSA status. tion optimal immunosuppression after stabilization after the acute
insult
Prompt and accurate diagnosis of acute-on-chronic renal deteriora-
tion with treatment based on etiologic causes
Treatment of a Failing Allograft
LONG-TERM IMMUNOSUPPRESSION
Fig. 27.1), indicating a single “magic bullet” is unlikely
Induction and maintenance regimens of conventional immu- to suffice. Instead, several therapeutic approaches
nosuppression produce good early results; however, evi- may be required to address multiple specific etiologic
dence for the optimal combination of medications supported threats (Tables 27.2 and 27.3). Adequate mainte-
by long-term clinical studies is more limited (Table 27.5). nance therapy is of prime importance; avoidance of
Most units use CNI-based triple therapy, some withdraw cor- underimmunosuppression and limiting nonadher-
ticosteroids routinely, most undertake dosage adjustments ence are central issues. Indirect clinical approaches,
or switch therapy according to changing clinical circum- such as control of hypertension, lipids, infections, and
stances. Ideal long-term therapeutic agents should effectively smoking are suggested.
suppress the alloimmune response, be nonnephrotoxic, well 2. Drivers of injury and potential threats vary by time post-
tolerated with simple oral dosing regimes, and devoid of risk transplant. Treatments exert different windows of bene-
for diabetes, cardiovascular disease, or neoplasia. No current fit, and therapy should ideally be initiated at early stages
immunosuppressive agent fulfills these criteria—all have of injury.
side effects affecting substantial numbers of patients. 3. Prevention is better than cure; lost nephrons are not
replaced. Early injury should be minimized by procuring
GENERAL TREATMENT PRINCIPLES an optimal donor organ, limiting ischemia-reperfusion
injury, and using adequate initial and subsequent main-
Key concepts applicable to treatment of chronic dysfunc- tenance immunosuppression with appropriate moni-
tion include the following: toring. Late rejection from nonadherence should be
  

1. Chronic allograft damage is commonly the end result minimized by optimal medical and psychological care,
of multiple pathophysiologic pathways of injury (see and follow-up monitoring.
452 Kidney Transplantation: Principles and Practice

4. 
Therapeutic flexibility of immunosuppression should when proteinuria exceeding 0.5 g/day.117 Other controlled
allow tailored changes according to altered clinical cir- trials of CNI elimination with sirolimus substitution or initial
cumstances and immunologic risk. Examples would avoidance, slowed or reduced vascular and tubulointerstitial
be CNI minimization strategies with DGF or late CNI damage.116,119 Belatacept (a selective blocker of T cell activa-
nephrotoxicity, or conversely, strengthening of immu- tion targeting the CD28-CD80/86 costimulation pathway)
nosuppression when subclinical, chronic, or late rejec- improved transplant function, patient, and graft survival
tion develops, optimal immunosuppression appropriate (43% reduction over 7 years) in a randomized trial against
to each individual patient’s immunologic risk category, cyclosporine-based therapy.84
and implementing early (biopsy) diagnosis and adequate
treatment for severe or resistant rejection. 2. Approach to Calcineurin Inhibitor
5. 
Technical advances in tissue pathology, such as Nephrotoxicity
gene-complementary DNA microarrays, microRNA, Isolated CNI nephrotoxicity (either acute cyclosporine-
proteomics, and metabolomics, are expected to yield diag- mediated glomerular vasoconstriction, or chronic struc-
nostic advances and mechanistic insights. Allograft tran- tural nephrotoxicity without rejection), if recognized early,
scriptome changes occur before histologic fibrosis and the can be treated by CNI sparing regimes with relatively good
pathologic expression of disease. Molecular signatures outcomes.82 CNI withdrawal reverses the functional decline
may improve diagnostic disease classification and provide and improves eGFR in failing transplants.119 Randomized
detailed information to better target treatment. trials of reduction or withdrawal of CNI improved renal
function in 90%, with a small risk of rejection and graft
TREATMENT APPROACH BY SPECIFIC loss.120 Patients interconverted from cyclosporine to low-
DIAGNOSIS dose tacrolimus, showed improved renal function, lipids,
hypertension, and cardiovascular markers, but unchanged
An accurate and specific etiologic diagnosis of the patho- 5-year graft survival.121
physiologic cause(s) of transplant dysfunction is the critical When deteriorating function from CNI nephrotoxicity
initial step toward rational treatment planning. occurs in low immune risk patients (and subclinical rejec-
tion is excluded by biopsy), then CNI withdrawal and main-
1. Interstitial Fibrosis and Tubular Atrophy tenance with concentration-controlled mycophenolate
The most common pathology of slowly progressive graft fail- mofetil and corticosteroids are recommended.122 When CNI
ure is chronic IF/TA, often accompanied by vascular abnor- nephrotoxicity occurs in recipients of higher immune risk,
malities and glomerulosclerosis.10 Previously designated as retransplants, or prior rejection episodes, then CNI can be
sclerosing CAN,115 this pattern occurs in 27% to 45% of late either minimized or substituted with sirolimus, everolimus,
graft losses,82,87 and the majority of late biopsies.a CAN is best or belatacept. The serum creatinine should be monitored as
considered as a final common pathway of nephron injury the rejection risk for CNI withdrawal approximates 10% to
with its fibrotic healing response, rather than a specific diag- 15%.123 mTOR inhibitors provide weaker immunosuppres-
nostic entity. Although multiple pathogenic pathways cause sion compared with CNI and are poorly tolerated at higher
IF/TA, the alloimmune response is the most important.10,26,49 doses with side effects including mouth ulcers, peripheral
The clinical approach to a failing graft with IF/TA should edema, proteinuria, anemia, or thrombocytopenia (discon-
consider potential immune and nonimmune etiologies, and tinuation rates are 30%–45%).
collate clinical, biologic, and histologic information. IF/TA Newer nonnephrotoxic immunosuppressive agents are
should be classified as primarily attributable to (1) nonimmune limited by incomplete long-term clinical data. Belatacept
causes, such as donor disease or early ischemia-reperfusion or tofacitinib (a JAK 2/3 inhibitor of cytokine signaling)
damage; (2) immune injury from acute, severe, persistent, when combined with mycophenolate decreased graft fail-
or late acute rejection; or (3) other specific diagnostic entities ure (odds ratio [OR] = 0.61, 95% confidence interval [CI]
including recurrent disease, BKVAN, hypertensive nephro- 0.39–0.96) in a meta-analysis of CNI-sparing trials.124
sclerosis, and CNI nephrotoxicity. Several causes of injury Tofacitinib provides effective immunosuppression in renal
may simultaneously coexist or be differentially expressed over transplantation, with increased risk of viral activation, ane-
the graft’s lifetime, and multiple diseases and pathologies may mia, and EBV-related lymphoproliferative disease, but has
be present on a single biopsy (see Table 27.2). been diverted away from transplantation into rheumatol-
One treatment strategy advocated for the IF/TA-CAN ogy. Voclosporine is a cyclosporine analog that is immu-
pattern is CNI minimization, based on the (often incorrect) nologically noninferior to tacrolimus and claims reduced
assumption than CNI nephrotoxicity is its primary cause. CNI nephrotoxicity. Sotrastaurin (a protein kinase C inhibitor
sparing regimes supplemented by mycophenolate increase reducing T-lymphocyte activation and cytokine release)
transplant glomerular filtration rate (GFR) in meta-analysis failed to provide adequate immunosuppression. ASKP1240
of RCTs and tend to improve graft survival. Acute rejection is (a humanized anti-CD40 monoclonal antibody targeting
increased in trials of mandated CNI elimination, but gener- the CD40/CD154 costimulatory pathway) is in preclinical
ally not for avoidance or dysfunction studies.118 A large ran- development with a CNI-free arm.125
domized study of transplant dysfunction (CAN was present
in 91.3%) that substituted sirolimus for CNI failed its primary 3. Chronic Antibody-Mediated Rejection
eGFR difference endpoint against continued CNI. Conversion Chronic AMR can occur from uncontrolled preformed DSA
was futile with severe dysfunction (eGFR < 40 mL/min) or or de novo DSA developing after transplantation.90,126
Treatment strategies for chronic AMR are adapted from
aReferences 10,49,56,82,116,117 acute AMR; however, evidence is poor and largely limited to
27 • Chronic Allograft Failure 453

uncontrolled cohort studies. Strengthened baseline immu- exclusion of interfering agents (e.g., St. John’s Wort or
nosuppression with increased tacrolimus and mycophe- phenytoin-inducing metabolizing enzymes and reducing
nolate dosages (suppressing B and T cell expansion) and CNI levels) are prudent. It is unknown whether CA-TCMR
corticosteroid use (re-added if patients are steroid-free or is responsive to increased immunosuppression or antilym-
upscaled) are suggested. ACEI or ARB reduce proteinuria phocyte therapies.
without changing the immunologic cause. Plasmapheresis
to remove circulating DSA is reasonable for acute AMR but 5. Treatment of Acute Late Rejection
futile as chronic treatment (DSA quickly return). Intrave- Acute rejection can develop late after transplantation in
nous immunoglobulin (IVIG to a cumulative total of 1–2 g/ sensitized patients, from medication noncompliance or
kg) is used in small uncontrolled trials of late rejection, with iatrogenic underimmunosuppression. Clinically severe,
uncertain or modest effectiveness. frequently steroid-resistant with limited reversibility, the
Rituximab, a chimeric monoclonal antibody against histology displays widespread tubulointerstitial damage,
CD20-bearing B lymphocytes, rapidly depletes B cells (but mononuclear interstitial infiltration (with lymphocytes,
not DSA-producing plasma cells), is well-tolerated with plasma cells, eosinophils, and macrophages), and is fre-
occasional acute infusion reactions, and a minor risk of bac- quently associated with C4d staining, de novo donor spe-
terial infection or latent viral reactivation. A multicenter, cific antibodies (typically class II), and/or acute or chronic
double-blind, placebo-controlled (RITUX ERAH) trial of vascular rejection.
biopsy-proven AMR in 38 patients, showed no difference Initial treatment by pulsed corticosteroids and strength-
of rituximab (375 mg/m2), when added to pulse steroids, ening baseline immunosuppression has limited success.
plasmapheresis, and IVIG in the composite endpoint of graft Antithymocyte globulin can be used for dominant cellu-
loss and day-12 functional improvement. A modest trend of lar rejection. Plasma exchange with IVIG (postexchange),
improved 6-month Banff scores favored rituximab.127 and possibly rituximab or bortezomib have been used for
Bortezomib (a proteasome inhibitor directed against antibody-dominant mixed rejection with modest results
plasma cells) slowly reduces DSA levels (IgG half-life is in uncontrolled cohort studies. Extent of therapy requires
7–21 days, according to subclass), when administered at a careful consideration of recipient risk against graft salvage-
typical single myeloma cycle dose (1.3 mg/m2 for 3–5 infu- ability. Uncontrolled chronic rejection leading to progres-
sions) with plasmapheresis as rescue therapy. Bortezomib sive graft failure is common.
successfully reduced DSA and reversed AMR in case reports
and small case series, but randomized trials are awaited. 6. Treatment of Recurrent Disease
Side effects include peripheral neuropathy (about 30%), Recurrent or de novo glomerulonephritis occurs in up to
myelosuppression, and herpes reactivation.90 8.4% of grafts99,133,134 with clinical clues being hematuria
Tocilizumab (an anti-IL-6 cytokine receptor blocker) or proteinuria. Control of hypertension and blockade of the
modulates proinflammatory and immune regulatory cas- renin angiotensin system are suggested and appear benefi-
cades involved in TCMR, AMR, and chronic vasculopathy. cial in cohort studies. Specific treatment varies by type of
Initial results in desensitization programs and AMR preven- recurrent glomerulonephritis.
tion and treatment studies are promising.128 Tocilizumab
rescue therapy (monthly infusions) of 36 recipients with Recurrent Focal Segmental Glomerulosclerosis. FSGS
chronic AMR who failed rituximab and IVIG (and variable exerts a disproportionate clinical effect because of its high
plasmapheresis) produced 6-year graft survival rates of recurrence rate, poor intermediate outcome, and the
80% (patient survival 91%), stabilized renal function, and numbers of young patients transplanted with renal fail-
reduced DSA levels; this betters historical control results.129 ure caused by FSGS. FSGS affects podocytes either from
genetic mutations in critical podocyte proteins (familial) or
4. Chronic Active T Cell Mediated Rejection secondary to an unknown circulating protein “toxic per-
Chronic-active TCMR is characterized by persistent inter- meability factor,” which increases glomerular permeabil-
stitial T lymphocyte infiltration with associated tubulitis, B ity (idiopathic). Idiopathic FSGS recurs in 20% to 50% of
cells and macrophages, and nephron loss which represent patients with about half failing within 5 years. Adverse risk
a failure of maintenance immunosuppression. The diagno- factors of FSGS recurrence include early childhood onset,
sis is supported by i-IFTA and tubulitis within IF/TA areas rapid progression to dialysis, diffuse mesangial hypercellu-
(i-IFTA2-3 and t-IFTA2-3 thresholds are proposed), where larity or glomerular collapse, or fulminant recurrence in
alternative diagnoses are excluded (e.g., BKVAN).73 i-IFTA previous transplant (where risk of graft loss for retransplant
strongly predicted graft failure (independent of renal func- is 70%).
tion, IF/TA, and inflammation scores) in the DEKAF study, Proteinuria may recur within hours posttransplant, but
and was associated with tissue injury and repair-associated typically develops by 1 to 2 weeks. Recurrence is suspected
transcripts from molecular studies.130 Fibrointimal arterial by an increasing weekly spot urine protein/creatinine ratio,
hyperplasia progressing to vascular narrowing is a rarer and confirmed by podocyte foot process effacement on EM
phenotype of CA-TCMR. (the earliest marker before segmental glomerulosclerosis on
Suggested treatment involves strengthened immu- light microscopy). Plasma exchange to remove circulating
nosuppression, such as conversion from cyclosporine to protein, high-dose cyclosporine, corticosteroid, and ACE
tacrolimus,131 azathioprine to mycophenolate mofetil,132 inhibitor (or ARB) use achieves complete or partial remis-
and addition of corticosteroids to dual-therapy or steroid- sion in 80% to 90% of patients.135 mTOR or VEGF inhibi-
free regimes, and strengthened to 10 mg/day. Compliance tors directly affect podocytes and worsen proteinuria; they
checks, review of appropriate target blood drug levels, and should be avoided or replaced.
454 Kidney Transplantation: Principles and Practice

Membranous Glomerulonephritis. Membranous glo- of nephron loss with a fibrotic healing response. Allograft
merulonephritis recurs in 10% to 30% of patients, but also damage is common, progressive, and remains clinically
constitutes the most common cause of de novo posttrans- important, despite improvements in immunosuppression
plant glomerular disease, presenting with subnephrotic and the control of early acute rejection episodes.
proteinuria or nephrotic syndrome. The 10-year graft loss An early phase of tubulointerstitial damage occurs
rate approximates 50%, with increased risk in male recipi- soon after transplantation and secondary to causes such
ents, in active original disease, and living donor transplants. as ischemia-reperfusion injury, ATN, acute rejection and
Immunosuppression with mycophenolate mofetil or aza- SCR, polyomavirus nephropathy, and CNI tubular neph-
thioprine, and corticosteroids reduce antibody formation. rotoxicity, superimposed on any preexisting donor disease.
Rituximab can be used for failed primary therapy. Subsequently, cellular infiltration and alloimmune injury
gradually lessens although subclinical chronic TCMR, and
Other Recurrent Diseases. Immunosuppression usually AMR can persist in some. Microvascular and glomerular
controls most immune-mediated renal diseases including abnormalities progress with time from CNI nephrotoxicity,
ANCA vasculitis, lupus nephritis, and Goodpasture’s dis- hypertension, chronic AMR with transplant glomerulopa-
ease. IgA nephropathy commonly recurs but generally with thy, and glomerulonephritis. Late acute dysfunction from
a milder clinical effect. Treatment including corticosteroids mixed acute rejection, BKVAN, and ATN from illness can
and mycophenolate is suggested. Rapid corticosteroid with- occur.
drawal triples the risk of recurrent IgA glomerulonephri- These pathogenic insults are associated with disruption
tis.136 Light chain deposition disease often recurs. Dense of the internal architecture of the transplanted kidney,
deposit disease recurs in 50% to 90% leading to graft fail- cortical ischemia from microvascular attenuation, and
ure in most, but eculizumab is helpful in case reports. Fabry persistent chronic inflammation that fails to resolve, and
disease recurs late after transplantation, requiring specific cytokine, chemokine, and growth factor production pro-
agalsidase treatment. Transplant diabetic nephropathy also moting genetic and phenotypical glomerular and tubular
reappears late with proteinuria and graft dysfunction and changes with excessive fibrosis and tissue remodeling and
characteristic tissue pathology. repair.
Progressive allograft dysfunction is detected by serial
7. Treatment of BK Virus Allograft Nephropathy monitoring of serum creatinine and investigated by evalu-
Current antiviral agents are relatively ineffective against ation of therapeutic drug levels, urinalysis, Doppler ultra-
BK, and correspondingly, preventive strategies such as sound imaging, and a timely diagnostic biopsy. A careful
avoidance of excessive immunosuppression and regular collaborative etiologic diagnosis should consider the rela-
viral surveillance are recommended. Quantitative testing tive effects of acute or chronic rejection against nonim-
of BK viremia (>10,000 copies/mL is high-level infection) mune factors (including preexisting donor quality and
is superior69,137 to urinary “decoy cell” cytology.137,138 vascular disease, early ischemia-reperfusion injury, recur-
Early detection of viremia by screening programs is man- rent glomerular disease, BKVAN, and CNI nephrotoxicity)
aged by cautious reduction of overall immunosuppression, and accelerating factors such as hypertension, proteinuria,
which allows reconstitution of anti-BK virus cell mediated dyslipidemia, and smoking. Treatment should be targeted
immunity with viral clearance and prevention of destruc- toward the dominant pathophysiologic diagnosis.
tive parenchymal infection.
Established BKVAN is more difficult to eradicate, espe- Acknowledgments
cially with active interstitial inflammation.70,71,139 Cipro- I am grateful for the excellent photomicrographs provided
floxacin, low-dose cidofovir, elimination or substitution by Dr. Rajathurai Murugasa and Professor Ranjit S. Nanra
of mycophenolate with leflunomide (all weak antiviral of John Hunter Hospital, Newcastle, NSW, Australia.
agents) or azathioprine, conversion from tacrolimus to
low-dose cyclosporine or 50% reduction of the CNI dose, References
and regular intravenous immunoglobulin therapy (pre- 1. Meier-Kriesche HU, Schold JD, Kaplan B. Long-term renal allograft
ferred for BK infection coexisting with “rejection”), are all survival: have we made significant progress or is it time to
rethink our analytic and therapeutic strategies? Am J Transplant
potential therapies demonstrating benefit in small cohort 2004;4(8):1289–95.
studies.123,140,141 Late rejection after reduced immuno- 2. Lamb KE, Lodhi S, Meier-Kriesche HU. Long-term renal allograft
suppression and sustained viral clearance is a risk affect- survival in the United States: a critical reappraisal. Am J Transplant
ing 15.8% of BKVAN kidneys, and modest strengthening 2011;11(3):450–62.
of maintenance immunosuppression should be considered 3. Segev DL. Renal allograft survival: getting better all the time. Am J
Transplant 2011;11(3):422–3.
after sustained viral clearance (especially for high immu- 4. Orandi BJ, Luo X, Massie AB, et al. Survival benefit with kidney
nologic risk patients) but regularly monitored for viral transplants from HLA-incompatible live donors. N Engl J Med
relapse.71 2016;374(10):940–50.
5. Hume DM, Merrill JP, Miller BF, Thorn GW. Experiences with renal
homotransplantation in the human: report of nine cases. J Clin
Invest 1955;34(2):327–82.
Summary 6. Nankivell BJ, Kuypers DR. Diagnosis and prevention of chronic kid-
ney allograft loss. Lancet 2011;378(9800):1428–37.
Progressive late allograft failure and chronic allograft dam- 7. Fernandez-Fresnedo G, Plaza JJ, Sanchez-Plumed J, Sanz-Guajardo
age is best conceptualized as the consequence of cumula- A, Palomar-Fontanet R, Arias M. Proteinuria: a new marker of long-
term graft and patient survival in kidney transplantation. Nephrol
tive transplant damage from time-dependent immune and Dial Transplant 2004;19(Suppl. 3):47–51.
nonimmune insults resulting in a final common pathway
27 • Chronic Allograft Failure 455

8. Veronese FV, Noronha IL, Manfro RC, Edelweiss MI, Goldberg J, Gon- 30. Hertig A, Anglicheau D, Verine J, et al. Early epithelial phenotypic
calves LF. Prevalence and immunohistochemical findings of subclin- changes predict graft fibrosis. J Am Soc Nephrol 2008;19(8):1584–
ical kidney allograft rejection and its association with graft outcome. 91.
Clin Transplant 2004;18(4):357–64. 31. Vitalone MJ, O’Connell PJ, Jimenez-Vera E, et al. Epithelial-to-
9. Ponticelli C, Cucchiari D, Graziani G. Hypertension in kidney trans- mesenchymal transition in early transplant tubulointerstitial
plant recipients. Transpl Int 2011;24(6):523–33. damage. J Am Soc Nephrol 2008;19(8):1571–83.
10. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Allen RD, Chap- 32. Carew RM, Wang B, Kantharidis P. The role of EMT in renal fibrosis.
man JR. The natural history of chronic allograft nephropathy. N Cell Tissue Res 2012;347(1):103–16.
Engl J Med 2003;349(24):2326–33. 33. Grimm PC, Nickerson P, Jeffery J, et al. Neointimal and tubulointer-
11. Nankivell BJ, Chapman JR. Chronic allograft nephropathy: cur- stitial infiltration by recipient mesenchymal cells in chronic renal-
rent concepts and future directions. Transplantation 2006;81(5): allograft rejection. N Engl J Med 2001;345(2):93–7.
643–54. 34. Grgic I, Duffield JS, Humphreys BD. The origin of interstitial myofibro-
12. Nankivell BJ, Kuypers DR, Fenton-Lee CA, Allen RD, O’Connell PJ, blasts in chronic kidney disease. Pediatr Nephrol 2012;27(2):183–
Chapman JR. Histological injury and renal transplant outcome: 93.
the cumulative damage hypothesis. Transplant Proc 2001;33(1- 35. Djamali A, Reese S, Yracheta J, Oberley T, Hullett D, Becker B. Epi-
2):1149–50. thelial-to-mesenchymal transition and oxidative stress in chronic
13. Naesens M, Khatri P, Li L, et al. Progressive histological damage in allograft nephropathy. Am J Transplant 2005;5(3):500–9.
renal allografts is associated with expression of innate and adaptive 36. Hazzan M, Hertig A, Buob D, et al. Epithelial-to-mesenchymal transi-
immunity genes. Kidney Int 2011;80(12):1364–76. tion predicts cyclosporine nephrotoxicity in renal transplant recipi-
14. Benigni A, Bruzzi I, Mister M, et al. Nature and mediators of renal ents. J Am Soc Nephrol 2011;22(7):1375–81.
lesions in kidney transplant patients given cyclosporine for more 37. Hertig A, Verine J, Mougenot B, et al. Risk factors for early epithe-
than one year. Kidney Int 1999;55(2):674–85. lial to mesenchymal transition in renal grafts. Am J Transplant
15. Evans NJ, White SA, Bicknell GR, Furness PN, Nicholson ML. The 2006;6(12):2937–46.
expression of endothelin and inducible nitric oxide synthase in 38. Vongwiwatana A, Tasanarong A, Rayner DC, Melk A, Halloran PF.
human renal allografts and their role in chronic renal allograft Epithelial to mesenchymal transition during late deterioration of
nephropathy. Transplant Proc 2001;33(1-2):1181. human kidney transplants: the role of tubular cells in fibrogenesis.
16. Grandaliano G, Di Paolo S, Monno R, et al. Protease-activated recep- Am J Transplant 2005;5(6):1367–74.
tor 1 and plasminogen activator inhibitor 1 expression in chronic 39. Halloran PF, Melk A, Barth C. Rethinking chronic allograft nephrop-
allograft nephropathy: the role of coagulation and fibrinolysis in athy: the concept of accelerated senescence. J Am Soc Nephrol
renal graft fibrosis. Transplantation 2001;72(8):1437–43. 1999;10(1):167–81.
17. Pilmore HL, Dittmer ID. Calcineurin inhibitor nephrotoxicity: reduc- 40. Melk A. Senescence of renal cells: molecular basis and clinical impli-
tion in dose results in marked improvement in renal function in cations. Nephrol Dial Transplant 2003;18(12):2474–8.
patients with coexisting chronic allograft nephropathy. Clin Trans- 41. Melk A, Schmidt BM, Takeuchi O, Sawitzki B, Rayner DC, Halloran
plant 2002;16(3):191–5. PF. Expression of p16INK4a and other cell cycle regulator and
18. Song E, Zou H, Yao Y, et al. Early application of Met-RANTES ame- senescence associated genes in aging human kidney. Kidney Int
liorates chronic allograft nephropathy. Kidney Int 2002;61(2):676– 2004;65(2):510–20.
85. 42. Naesens M. Replicative senescence in kidney aging, renal disease,
19. Albrecht EW, van Goor H, Smit-van Oosten A, Stegeman CA. Long- and renal transplantation. Discov Med 2011;11(56):65–75.
term dietary L-arginine supplementation attenuates proteinuria and 43. Schmitt R, Melk A. Molecular mechanisms of renal aging. Kidney Int
focal glomerulosclerosis in experimental chronic renal transplant 2017;92(3):569–79.
failure. Nitric Oxide 2003;8(1):53–8. 44. Kaelin Jr WG, Ratcliffe PJ. Oxygen sensing by metazoans: the cen-
20. Gourishankar S, Hunsicker LG, Jhangri GS, Cockfield SM, Halloran tral role of the HIF hydroxylase pathway. Mol Cell 2008;30(4):393–
PF. The stability of the glomerular filtration rate after renal trans- 402.
plantation is improving. J Am Soc Nephrol 2003;14(9):2387–94. 45. Ishii Y, Sawada T, Kubota K, Fuchinoue S, Teraoka S, Shimizu A.
21. Mengel M, Reeve J, Bunnag S, et al. Scoring total inflammation is Injury and progressive loss of peritubular capillaries in the develop-
superior to the current Banff inflammation score in predicting out- ment of chronic allograft nephropathy. Kidney Int 2005;67(1):321–
come and the degree of molecular disturbance in renal allografts. 32.
Am J Transplant 2009;9(8):1859–67. 46. Ishii Y, Shimizu A, Sawada T, et al. Injury of peritubular capillaries
22. Bestard O, Cunetti L, Cruzado JM, et al. Intragraft regulatory T correlates with graft function in chronic renal allograft nephropa-
cells in protocol biopsies retain foxp3 demethylation and are pro- thy. Transplant Proc 2001;33(1-2):1213–4.
tective biomarkers for kidney graft outcome. Am J Transplant 47. Steegh FM, Gelens MA, Nieman FH, et al. Early loss of peritubu-
2011;11(10):2162–72. lar capillaries after kidney transplantation. J Am Soc Nephrol
23. Alessandrini A, Turka LA. FOXP3-positive regulatory T cells and 2011;22(6):1024–9.
kidney allograft tolerance. Am J Kidney Dis 2017;69(5):667–74. 48. Bonsib SM, Abul-Ezz SR, Ahmad I, et al. Acute rejection-associated
24. Mannon RB, Matas AJ, Grande J, et al. Inflammation in areas of tubular basement membrane defects and chronic allograft nephrop-
tubular atrophy in kidney allograft biopsies: a potent predictor of athy. Kidney Int 2000;58(5):2206–14.
allograft failure. Am J Transplant 2010;10(9):2066–73. 49. Naesens M, Kuypers DR, De Vusser K, et al. The histology of kidney
25. Moreso F, Ibernon M, Goma M, et al. Subclinical rejection associated transplant failure: a long-term follow-up study. Transplantation
with chronic allograft nephropathy in protocol biopsies as a risk fac- 2014;98(4):427–35.
tor for late graft loss. Am J Transplant 2006;6(4):747–52. 50. Wang CJ, Wetmore JB, Crary GS, Kasiske BL. The donor kidney
26. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Chapman JR, Allen biopsy and its implications in predicting graft outcomes: a systematic
RD. Delta analysis of posttransplantation tubulointerstitial damage. review. Am J Transplant 2015;15(7):1903–14.
Transplantation 2004;78(3):434–41. 51. Gasser M, Waaga AM, Laskowski IA, Tilney NL. The influence of
27. Nankivell BJ, Shingde M, Keung KL, et al. The causes, significance donor brain death on short and long-term outcome of solid organ
and consequences of inflammatory fibrosis in kidney transplanta- allografts. Ann Transplant 2000;5(4):61–7.
tion: the Banff i-IFTA lesion. Am J Transplant 2018;18(2):364–76. 52. Heilman RL, Mathur A, Smith ML, Kaplan B, Reddy KS. Increas-
https://doi.org/10.1111/ajt.14609. ing the use of kidneys from unconventional and high-risk deceased
28. Lefaucheur C, Gosset C, Rabant M, et al. T cell-mediated rejection donors. Am J Transplant 2016;16(11):3086–92.
is a major determinant of inflammation in scarred areas in kid- 53. Siedlecki A, Irish W, Brennan DC. Delayed graft function in the kid-
ney allografts. Am J Transplant 2018;18(2):377–90. https://doi. ney transplant. Am J Transplant 2011;11(11):2279–96.
org/10.1111/ajt.14565. 54. Yarlagadda SG, Coca SG, Formica Jr RN, Poggio ED, Parikh CR.
29. Galichon P, Vittoz N, Xu-Dubois YC, et al. Epithelial phenotypic Association between delayed graft function and allograft and patient
changes detect cyclosporine in vivo nephrotoxicity at a reversible survival: a systematic review and meta-analysis. Nephrol Dial
stage. Transplantation 2011;92(9):993–8. Transplant 2009;24(3):1039–47.
456 Kidney Transplantation: Principles and Practice

55. Perez-Saez MJ, Montero N, Redondo-Pachon D, Crespo M, Pascual J. 77. Brocker V, Schubert V, Scheffner I, et al. Arteriolar lesions in renal
Strategies for an expanded use of kidneys from elderly donors. Trans- transplant biopsies: prevalence, progression, and clinical signifi-
plantation 2017;101(4):727–45. cance. Am J Pathol 2012;180(5):1852–62.
56. Cosio FG, Grande JP, Wadei H, Larson TS, Griffin MD, Stegall MD. 78. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Chapman JR, Allen
Predicting subsequent decline in kidney allograft function from early RD. Calcineurin inhibitor nephrotoxicity: longitudinal assessment
surveillance biopsies. Am J Transplant 2005;5(10):2464–72. by protocol histology. Transplantation 2004;78(4):557–65.
57. Flechner SM, Kurian SM, Solez K, et al. De novo kidney transplanta- 79. Horike K, Takeda A, Yamaguchi Y, et al. Is arteriolar vacuolization
tion without use of calcineurin inhibitors preserves renal structure a predictor of calcineurin inhibitor nephrotoxicity? Clin Transplant
and function at two years. Am J Transplant 2004;4(11):1776–85. 2011;25(Suppl. 23):23–7.
58. McDonald S, Russ G, Campbell S, Chadban S. Kidney transplant 80. Sis B, Dadras F, Khoshjou F, Cockfield S, Mihatsch MJ, Solez K.
rejection in Australia and New Zealand: relationships between rejec- Reproducibility studies on arteriolar hyaline thickening scoring in
tion and graft outcome. Am J Transplant 2007;7(5):1201–8. calcineurin inhibitor-treated renal allograft recipients. Am J Trans-
59. Shishido S, Asanuma H, Nakai H, et al. The impact of repeated plant 2006;6(6):1444–50.
subclinical acute rejection on the progression of chronic allograft 81. Snanoudj R, Royal V, Elie C, et al. Specificity of histological mark-
nephropathy. J Am Soc Nephrol 2003;14(4):1046–52. ers of long-term CNI nephrotoxicity in kidney-transplant recipi-
60. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Allen RD, Chap- ents under low-dose cyclosporine therapy. Am J Transplant
man JR. Natural history, risk factors, and impact of subclinical rejec- 2011;11(12):2635–46.
tion in kidney transplantation. Transplantation 2004;78(2):242–9. 82. Gourishankar S, Leduc R, Connett J, et al. Pathological and clinical
61. Rush DN, Jeffery J, Nickerson P. Subclinical acute rejection: Is it a characterization of the “troubled transplant”: data from the DeKAF
cause of chronic rejction in renal transplantation? J Am Soc Nephrol study. Am J Transplant 2010;10(2):324–30.
2000;14(3):131–7. 83. Durrbach A, Pestana JM, Florman S, et al. Long-term outcomes in
62. Rush D, Nickerson P, Gough J, et al. Beneficial effects of treatment belatacept- versus cyclosporine-treated recipients of extended crite-
of early subclinical rejection: a randomized study. J Am Soc Nephrol ria donor kidneys: final results from BENEFIT-EXT, a phase III ran-
1998;9(11):2129–34. domized study. Am J Transplant 2016;16(11):3192–201.
63. Rush D, Arlen D, Boucher A, et al. Lack of benefit of early protocol 84. Vincenti F, Rostaing L, Grinyo J, et al. Belatacept and long-term
biopsies in renal transplant patients receiving TAC and MMF: a ran- outcomes in kidney transplantation. N Engl J Med 2016;374(4):
domized study. Am J Transplant 2007;7(11):2538–45. 333–43.
64. Kurtkoti J, Sakhuja V, Sud K, et al. The utility of 1- and 3-month pro- 85. Gibson IW, Downie TT, More IA, Lindop GB. Atubular glomeruli
tocol biopsies on renal allograft function: a randomized controlled and glomerular cysts—a possible pathway for nephron loss in the
study. Am J Transplant 2008;8(2):317–23. human kidney? J Pathol 1996;179(4):421–6.
65. Lerut E, Naesens M, Kuypers DR, Vanrenterghem Y, Van Damme 86. Colvin RB, Smith RN. Antibody-mediated organ-allograft rejection.
B. Subclinical peritubular capillaritis at 3 months is associated with Nat Rev Immunol 2005;5(10):807–17.
chronic rejection at 1 year. Transplantation 2007;83(11):1416– 87. El-Zoghby ZM, Stegall MD, Lager DJ, et al. Identifying specific causes
22. of kidney allograft loss. Am J Transplant 2009;9(3):527–35.
66. Loupy A, Hill GS, Suberbielle C, et al. Significance of C4d Banff 88. Matas AJ, Leduc R, Rush D, et al. Histopathologic clusters dif-
scores in early protocol biopsies of kidney transplant recipients ferentiate subgroups within the nonspecific diagnoses of CAN or
with preformed donor-specific antibodies (DSA). Am J Transplant CR: preliminary data from the DeKAF study. Am J Transplant
2011;11(1):56–65. 2010;10(2):315–23.
67. Sis B, Jhangri GS, Bunnag S, Allanach K, Kaplan B, Halloran PF. 89. Sis B, Campbell PM, Mueller T, et al. Transplant glomerulopathy,
Endothelial gene expression in kidney transplants with alloantibody late antibody-mediated rejection and the ABCD tetrad in kidney
indicates antibody-mediated damage despite lack of C4d staining. allograft biopsies for cause. Am J Transplant 2007;7(7):1743–52.
Am J Transplant 2009;9(10):2312–23. 90. Stegall MD, Gloor JM. Deciphering antibody-mediated rejection: new
68. Hirsch HH, Steiger J, Polyomavirus BK. Lancet Infect Dis insights into mechanisms and treatment. Curr Opin Organ Trans-
2003;3(10):611–23. plant 2010;15(1):8–10.
69. Hirsch HH, Knowles W, Dickenmann M, et al. Prospective study 91. Regele H, Bohmig GA, Habicht A, et al. Capillary deposition of com-
of polyomavirus type BK replication and nephropathy in renal- plement split product C4d in renal allografts is associated with base-
transplant recipients. N Engl J Med 2002;347(7):488–96. ment membrane injury in peritubular and glomerular capillaries: a
70. Johnston O, Jaswal D, Gill JS, Doucette S, Fergusson DA, Knoll GA. contribution of humoral immunity to chronic allograft rejection. J
Treatment of polyomavirus infection in kidney transplant recipients: Am Soc Nephrol 2002;13(9):2371–80.
a systematic review. Transplantation 2010;89(9):1057–70. 92. Sijpkens YW, Bruijn JA, Paul LC. Chronic allograft nephropathy
71. Nankivell BJ, Renthawa J, Sharma RN, Kable K, O’Connell PJ, Chap- categorised in chronic interstitial and vascular rejection. Transplant
man JR. BK virus nephropathy: histological evolution by sequential Proc 2001;33(1-2):1153.
pathology. Am J Transplant 2017;17(8):2065–77. 93. Halloran PF, Famulski KS, Reeve J. Molecular assessment of dis-
72. Menter T, Mayr M, Schaub S, Mihatsch MJ, Hirsch HH, Hopfer H. ease states in kidney transplant biopsy samples. Nat Rev Nephrol
Pathology of resolving polyomavirus-associated nephropathy. Am J 2016;12(9):534–48.
Transplant 2013;13(6):1474–83. 94. Aubert O, Loupy A, Hidalgo L, et al. Antibody-mediated rejection
73. Haas M, Loupy A, Lefaucheur C, et al. The Banff 2017 kidney due to preexisting versus de novo donor-specific antibodies in kidney
meeting report: revised diagnostic criteria for chronic active t cell- allograft recipients. J Am Soc Nephrol 2017;28(6):1912–23.
mediated rejection, antibody-mediated rejection, and prospects 95. Ivanyi B, Fahmy H, Brown H, Szenohradszky P, Halloran PF, Solez K.
for integrative endpoints for next-generation clinical trials. Am J Peritubular capillaries in chronic renal allograft rejection: a quanti-
Transplant 2018;18(2):293–307. Available online at: https://doi. tative ultrastructural study. Hum Pathol 2000;31(9):1129–38.
org/10.1111/ajt.14625. 96. Mauiyyedi S, Pelle PD, Saidman S, et al. Chronic humoral rejection:
74. Loupy A, Haas M, Solez K, et al. The Banff 2015 kidney meeting identification of antibody-mediated chronic renal allograft rejec-
report: current challenges in rejection classification and prospects tion by C4d deposits in peritubular capillaries. J Am Soc Nephrol
for adopting molecular pathology. Am J Transplant 2017;17(1):28– 2001;12(3):574–82.
41. 97. Mengel M, Bogers J, Bosmans JL, et al. Incidence of C4d stain in pro-
75. Sellares J, de Freitas DG, Mengel M, et al. Inflammation lesions in tocol biopsies from renal allografts: results from a multicenter trial.
kidney transplant biopsies: association with survival is due to the Am J Transplant 2005;5(5):1050–6.
underlying diseases. Am J Transplant 2011;11(3):489–99. 98. Orandi BJ, Alachkar N, Kraus ES, et al. Presentation and outcomes of
76. Nankivell BJ, P’Ng CH, O’Connell PJ, Chapman JR. calcineurin C4d-negative antibody-mediated rejection after kidney transplanta-
inhibitor nephrotoxicity through the lens of longitudinal histology: tion. Am J Transplant 2016;16(1):213–20.
comparison of cyclosporine and tacrolimus eras. Transplantation 99. Briganti EM, Russ GR, McNeil JJ, Atkins RC, Chadban SJ. Risk of
2016;100(8):1723–31. renal allograft loss from recurrent glomerulonephritis. N Engl J Med
2002;347(2):103–9.
27 • Chronic Allograft Failure 457

100. Chadban S. Glomerulonephritis recurrence in the renal graft. J Am failure: 60-month results of the CRAF Study. Transplantation
Soc Nephrol 2001;12(2):394–402. 2008;85(9):1261–9.
101. Cosio FG, Cattran DC. Recent advances in our understanding of 122. Hazzan M, Labalette M, Copin MC, et al. Predictive factors of acute
recurrent primary glomerulonephritis after kidney transplantation. rejection after early cyclosporine withdrawal in renal transplant
Kidney Int 2017;91(2):304–14. recipients who receive mycophenolate mofetil: results from a pro-
102. Naesens M, Heylen L, Lerut E, et al. Intrarenal resistive index after spective, randomized trial. J Am Soc Nephrol 2005;16(8):2509–16.
renal transplantation. N Engl J Med 2013;369(19):1797–806. 123. Kasiske BL, Zeier MG, Chapman JR, et al. KDIGO clinical practice
103. Radermacher J, Mengel M, Ellis S, et al. The renal arterial resistance guideline for the care of kidney transplant recipients: a summary.
index and renal allograft survival. N Engl J Med 2003;349(2):115– Kidney Int 2010;77(4):299–311.
24. 124. Sharif A, Shabir S, Chand S, Cockwell P, Ball S, Borrows R. Meta-
104. te Strake L, Schultze Kool LJ, Paul LC, et al. Magnetic resonance analysis of calcineurin-inhibitor-sparing regimens in kidney trans-
imaging of renal transplants: its value in the differentiation of plantation. J Am Soc Nephrol 2011;22(11):2107–18.
acute rejection and cyclosporin A nephrotoxicity. Clin Radiol 125. Webster AC, Ruster LP, McGee R, et al. Interleukin 2 receptor antag-
1988;39(3):220–8. onists for kidney transplant recipients. Cochrane Database Syst Rev
105. Hanssen O, Erpicum P, Lovinfosse P, et al. Non-invasive approaches 2010;(1):CD003897.
in the diagnosis of acute rejection in kidney transplant recipients. 126. Terasaki PI, Cai J. Human leukocyte antigen antibodies and
Part I. In vivo imaging methods. Clin Kidney J 2017;10(1):97–105. chronic rejection: from association to causation. Transplantation
106. Muthukumar T, Dadhania D, Ding R, et al. Messenger RNA for 2008;86(3):377–83.
FOXP3 in the urine of renal-allograft recipients. N Engl J Med 127. Sautenet B, Blancho G, Buchler M, et al. One-year results of the
2005;353(22):2342–51. effects of rituximab on acute antibody-mediated rejection in renal
107. Erpicum P, Hanssen O, Weekers L, et al. Non-invasive approaches transplantation: RITUX ERAH, a multicenter double-blind random-
in the diagnosis of acute rejection in kidney transplant recipients, ized placebo-controlled trial. Transplantation 2016;100(2):391–9.
part II: omics analyses of urine and blood samples. Clin Kidney J 128. Jordan SC, Choi J, Kim I, et al. Interleukin-6, A cytokine critical to
2017;10(1):106–15. mediation of inflammation, autoimmunity and allograft rejection:
108. Hricik DE, Nickerson P, Formica RN, et al. Multicenter validation of therapeutic implications of IL-6 receptor blockade. Transplantation
urinary CXCL9 as a risk-stratifying biomarker for kidney transplant 2017;101(1):32–44.
injury. Am J Transplant 2013;13(10):2634–44. 129. Choi J, Aubert O, Vo A, et al. Assessment of tocilizumab (anti-
109. Suthanthiran M, Schwartz JE, Ding R, et al. Urinary-cell mRNA pro- interleukin-6 receptor monoclonal) as a potential treatment for
file and acute cellular rejection in kidney allografts. N Engl J Med chronic antibody-mediated rejection and transplant glomerulopa-
2013;369(1):20–31. thy in HLA-sensitized renal allograft recipients. Am J Transplant
110. Nankivell BJ. microRNA in transplantation: small in name only. 2017;17(9):2381–9.
Transplantation 2015;99(9):1754–5. 130. Halloran PF, Venner JM, Madill-Thomsen KS, et al. Review: the
111. van de Vrie M, Deegens JK, Eikmans M, van der Vlag J, Hilbrands transcripts associated with organ allograft rejection. Am J Trans-
LB. Urinary MicroRNA as biomarker in renal transplantation. Am J plant 2017.
Transplant 2017;17(5):1160–6. 131. Webster AC, Woodroffe RC, Taylor RS, Chapman JR, Craig JC. Tacro-
112. Bestard O, Nickel P, Cruzado JM, et al. Circulating alloreactive T cells limus versus ciclosporin as primary immunosuppression for kidney
correlate with graft function in longstanding renal transplant recipi- transplant recipients: meta-analysis and meta-regression of ran-
ents. J Am Soc Nephrol 2008;19(7):1419–29. domised trial data. BMJ 2005;331(7520):810.
113. Kowalski RJ, Post DR, Mannon RB, et al. Assessing relative risks of 132. Knight SR, Russell NK, Barcena L, Morris PJ. Mycophenolate mofetil
infection and rejection: a meta-analysis using an immune function decreases acute rejection and may improve graft survival in renal
assay. Transplantation 2006;82(5):663–8. transplant recipients when compared with azathioprine: a system-
114. Schwarz A, Gwinner W, Hiss M, Radermacher J, Mengel M, Haller atic review. Transplantation 2009;87(6):785–94.
H. Safety and adequacy of renal transplant protocol biopsies. Am J 133. Golgert WA, Appel GB, Hariharan S. Recurrent glomerulonephri-
Transplant 2005;5(8):1992–6. tis after renal transplantation: an unsolved problem. Clin J Am Soc
115. Solez K, Axelsen RA, Benediktsson H, et al. International stan- Nephrol 2008;3(3):800–7.
dardization of criteria for the histologic diagnosis of renal allograft 134. Ivanyi B. A primer on recurrent and de novo glomerulonephritis in
rejection: the Banff working classification of kidney transplant renal allografts. Nat Clin Pract Nephrol 2008;4(8):446–57.
pathology. Kidney Int 1993;44(2):411–22. 135. Canaud G, Zuber J, Sberro R, et al. Intensive and prolonged treat-
116. Mota A, Arias M, Taskinen EI, et al. Sirolimus-based therapy fol- ment of focal and segmental glomerulosclerosis recurrence in
lowing early cyclosporine withdrawal provides significantly adult kidney transplant recipients: a pilot study. Am J Transplant
improved renal histology and function at 3 years. Am J Transplant 2009;9(5):1081–6.
2004;4(6):953–61. 136. Kukla A, Chen E, Spong R, et al. Recurrent glomerulonephri-
117. Schena FP, Pascoe MD, Alberu J, et al. Conversion from calcineu- tis under rapid discontinuation of steroids. Transplantation
rin inhibitors to sirolimus maintenance therapy in renal allograft 2011;91(12):1386–91.
recipients: 24-month efficacy and safety results from the CONVERT 137. Nankivell BJ, Renthawa J, Jeoffreys N, et al. Clinical utility of uri-
trial. Transplantation 2009;87(2):233–42. nary cytology to detect BK viral nephropathy. Transplantation
118. Moore J, Middleton L, Cockwell P, et al. Calcineurin inhibitor sparing 2015;99(8):1715–22.
with mycophenolate in kidney transplantation: a systematic review 138. Drachenberg RC, Drachenberg CB, Papadimitriou JC, et al. Morpho-
and meta-analysis. Transplantation 2009;87(4):591–605. logical spectrum of polyoma virus disease in renal allografts: diagnos-
119. Flechner SM, Kobashigawa J, Klintmalm G. Calcineurin inhibitor- tic accuracy of urine cytology. Am J Transplant 2001;1(4):373–81.
sparing regimens in solid organ transplantation: focus on improving 139. Nickeleit V, Singh HK, Mihatsch MJ. Polyomavirus nephropathy:
renal function and nephrotoxicity. Clin Transplant 2008;22(1):1– morphology, pathophysiology, and clinical management. Curr Opin
15. Nephrol Hypertens 2003;12(6):599–605.
120. Abramowicz D, Del Carmen Rial M, Vitko S, et al. Cyclosporine with- 140. Hilton R, Tong CY. Antiviral therapy for polyomavirus-associated
drawal from a mycophenolate mofetil-containing immunosuppres- nephropathy after renal transplantation. J Antimicrob Chemother
sive regimen: results of a five-year, prospective, randomized study. J 2008;62(5):855–9.
Am Soc Nephrol 2005;16(7):2234–40. 141. Kable K, Davies CD, O’Connell PJ, Chapman JR, Nankivell BJ. Clear-
121. Shihab FS, Waid TH, Conti DJ, et al. Conversion from cyclospo- ance of BK virus nephropathy by combination antiviral therapy with
rine to tacrolimus in patients at risk for chronic renal allograft intravenous immunoglobulin. Transplant Direct 2017;3(4):e142.
28 Vascular and Lymphatic
Complications After Kidney
Transplantation
SIMON R. KNIGHT and RICHARD D.M. ALLEN

CHAPTER OUTLINE Introduction Vascular Access Thrombosis


Technical Complications and Their Deep Vein Thrombosis
Prevention Vascular Causes of Ureteric Complications
Informed Consent Mycotic Aneurysm
Preoperative Assessment Biopsy-Related Vascular Complications
Right or Left Donor Kidney Transplant Renal Artery Stenosis
Back Table Preparation Definition and Incidence
Transplant Renal Vein Anastomosis Pathogenesis
Transplant Renal Artery Anastomosis Pathophysiology: “One Kidney, One Clip”
Reperfusion Imaging
Positioning the Kidney and Wound Closure Conservative Treatment
Postoperative Recovery Angioplasty and Stenting
Drain Tube Surgical Correction
Compartment Syndrome Lymphocele
Hematoma Incidence
Vascular Thrombosis and Thrombophilia Etiology
Thrombophilic Factors Presentation
Contribution of Immunosuppressive Agents Diagnosis
Renal Vein Thrombosis Treatment
Renal Artery Thrombosis Lymphocutaneous Fistula
Segmental Arterial Thrombosis Conclusions
Thrombosis Prevention Strategies
Torsion

Introduction example of the magnitude of the renal blood flow is the sim-
ple temporary occlusion of the transplant renal vein with a
The enduring techniques of vascular anastomoses described pair of forceps at the time of surgery, described clinically as
by Alexis Carrel more than a century ago have not changed the Hume test, which results in rapid and pulsatile engorge-
significantly (see Chapter 1). His simple test of satisfactory ment of a well-perfused kidney transplant. Equally, a breach
vascular anastomosis was observation of a viable kidney in the continuity of the transplanted artery or vein can result
transplant producing urine within minutes of completion. in catastrophic blood loss and circulatory failure within
No matter how many times it has been witnessed before, this minutes, particularly in the presence of a recipient left ven-
observation will always put a smile on the face of everyone tricle already compromised by coexisting coronary artery
in the kidney transplant operating room. However, the pro- disease, long-term effects of systemic hypertension, or uremic
gressive improvement in kidney graft survival has focused cardiomyopathy.
greater emphasis on surgeon-related causes of kidney graft The kidney is also unforgiving of interruption of blood
loss. Surgical misadventure after kidney transplantation, flow, with the cortex more sensitive to hypoxia than the
once ranked low as a cause of graft loss in the first 6 months medulla. The magnitude of the effect of acute and complete
after transplantation, is now three times more likely to interruption of blood flow during the transplantation pro-
occur than graft loss as a result of rejection (Fig. 28.1). cedure depends on the quality of the donor kidney, length
The transplanted kidney is a highly vascular organ. At of ischemia time, temperature of the kidney, and extent of
rest, 10% to 15% of cardiac output, accounting for between intrarenal thrombosis that might occur during a period
500 and 750 mL/min, passes through the kidney. A graphic of stasis or reduced renal blood flow. Irreversible cortical
458
28 • Vascular and Lymphatic Complications after Kidney Transplantation 459

20 deceased donor, are not new engine parts that can be taken
off a spare parts shelf. They are preowned and have no
18 regenerative capacity. The good kidney transplant sur-
1970–1974 geon is one who recognizes the small margin for surgical
16 2005–2009 error and avoids difficult situations by careful preparation
and anticipation of the potential pitfalls. The incidence of
14 vascular complications will vary according to the quality
of the recipient evaluation, the donor kidney, and surgical
12 technique of implantation. These are discussed in detail in
Chapters 4, 8, and 11, but some of these points relating to
10 prevention of vascular complications are worth reiterating
here. When complications do occur, the surgeon attempts
8
salvage of the situation, balancing risks to both the recipi-
ent and the donor kidney.
6

4 PREOPERATIVE ASSESSMENT
2
An accessible, patent iliac artery and vein with unimpeded
proximal blood flow that are able to be sutured are essential,
0 and should be evaluated by a senior member of the trans-
Rejection Technical Death Other plant surgery team before the patient is placed on the trans-
Fig. 28.1 Causes of graft loss in the first 6 months after kidney trans-
plant waiting list. Surgical access can be difficult because
plantation reported to ANZDATA Registry in Australia for recipients of a polycystic kidney, obesity, or a failed previous kidney
of living and deceased donors, comparing 5-year time periods 1970 transplant. Extensive mural arterial calcification can make
through 1974 (n = 1118 transplants) and 2005 through 2009 (n = 4014 clamping and suturing impossible without disruption of
transplants). the artery. Placement of a patient on a kidney transplant
waitlist without surgical assessment and resolution of
necrosis can occur within minutes, and even in the most identified problems, and lack of systems in place to ensure
favorable situations, it is inevitable by the 20-minute mark. access to results of the assessment at all times, is considered
Incomplete interruption of blood flow has a more subtle medically negligent in the event of an avoidable vascular
effect. Arterial pressure sensors within the kidney detect complication.
pressures capable of triggering a cascade of autoregula-
tory changes to increase systemic pressures to satisfy the
requirements of the kidney, sometimes at the expense of the
RIGHT OR LEFT DONOR KIDNEY
recipient’s well-being. Impaired venous drainage is prob- In general, transplant surgeons always prefer the left donor
ably better tolerated, although sudden occlusion of a pre- kidney because of its longer renal vein and shorter artery.
viously well-perfused kidney can lead to dramatic rupture When given the choice of a living left donor kidney with
of the cortex with uncontrolled bleeding from intrarenal two arteries or a right kidney with one artery, most sur-
veins. geons choose the former.1 The longer left renal vein is less
fragile and more easily sutured to the more deeply situated
external iliac vein than the short right renal vein. Equally,
Technical Complications and the right renal artery anastomosis is more difficult to site
Their Prevention because of its propensity to kink. Objective evidence to sup-
port the greater ease of transplantation of the left kidney is
INFORMED CONSENT found in Australian registry data that compared outcomes
of left and right deceased donor kidney pairs.2 The data of
A description of the possible complications of kidney trans- paired kidneys from 2396 heart-beating brain-dead donors
plantation given to a patient when obtaining informed showed that recipients of right-sided kidneys were at signifi-
consent before surgery can cause alarm. The possibilities cantly greater risk of developing delayed graft function and
should be put into the context of the individual transplant had inferior graft function because of greater risk of graft
center’s own published results of patient and graft survival loss in the 3 months after transplantation, but principally
at 1 year. Ideally, the individual transplant surgeon’s own because of surgical misadventure. Recent registry data
results will be peer-reviewed on a regular basis both within from the Netherlands, however, refute these findings, sug-
and outside the surgeon’s own transplant center. gesting that although the risk of technical complications
Kidney transplantation involves placement of a kidney for deceased donor implants is not different for right or left
in a heterotopic position. By comparison, cardiothoracic kidneys, technical graft failure rate is higher with the right
and liver transplant surgeons have an easier task, placing kidney from a living donor.3
size-matched donor organs into an orthotopic position after Anastomosis of the deceased donor right renal vein can be
removal of the failed recipient organ. In deceased donor facilitated by vein elongation using the adjacent donor infe-
kidney transplantation, the surgeon must cope with the rior vena cava (Fig. 28.2) or a donor iliac vein extension graft.
computer-allocated pairing of the donor kidney and recipi- Alternatively, as is frequently the need in living donor right
ent. Donor kidneys, particularly from an extended criteria kidneys, the recipient external iliac vein can be mobilized by
460 Kidney Transplantation: Principles and Practice

A B

C
Fig. 28.2 Extension of the right renal vein using adjacent deceased donor inferior vena cava (IVC). (A) Marking the portion of IVC proposed for
elongation. (B) Fashioning of the IVC. (C) Elongated renal vein ready for transplantation.

dividing the internal iliac veins. Creation of a vein extension


using the gonadal vein in the live donor kidney has also been
described, but in practice is rarely necessary.4

BACK TABLE PREPARATION


All donor kidneys require back table preparation, and
failure of the surgeon to examine the deceased donor
kidney before starting the recipient procedure can cre-
ate problems if the kidney is not “as advertised” by the
retrieval surgeon. Accessory arteries may have been
missed or divided (Fig. 28.3). Atheromatous plaque, clot,
or an intimal flap may be impinging on the lumen of the
renal artery. Inadvertent traction or a donor surgeon’s
wayward scissor may have torn or injured the donor
renal vein. If problems are identified and corrected before
surgery, operating and anastomosis times are kept to
a minimum and surgical options are retained, such as Fig. 28.3 Right-sided deceased donor kidney with two renal arteries. The
preservation of the inferior epigastric artery for anasto- lower artery was divided at the time of multiorgan donor retrieval surgery.
mosis to a lower-pole artery. For living donor kidneys,
a missed accessory artery in the living donor kidney is the gonadal vein ligated and removed and, in the case
apparent at the time of initial cold perfusion at the back of a deceased donor kidney, the adrenal gland removed.
table. This is not the case for the in situ, cold-perfused, Hemostasis after revascularization of the transplanted
deceased donor kidney. Donor artery and vein are mobi- kidney is easier if vein tributaries and small hilar vessels
lized as necessary, with perirenal adipose tissue trimmed, associated with trimmed tissue are ligated.
28 • Vascular and Lymphatic Complications after Kidney Transplantation 461

A B
Fig. 28.4 Marking of the vessels of a left-sided living donor kidney transplant before transplantation for the purpose of assisting their orientation at the
time of surgery. (A) Superior margin of the renal artery. (B) Inferior margin of the renal vein.

Repeat flushing of a deceased donor kidney with a small vol-


ume of preservation solution has several advantages. Residual
venous blood, if present, can be cleared. Leaking vessels can
be identified and ligated before revascularization. There is also
clinical evidence that the subsequently “freshened” deceased
donor kidney is more likely to avoid graft dysfunction.5 Finally,
the kidney vasculature is accurately oriented. The superior
and inferior margins of the artery and vein can be marked to
reduce the risk of twisting the vessels at the time of anastomo-
sis (Fig. 28.4). To reduce handling of the donor kidney during
the surgical procedure and for ease of surgery, the kidney can
be placed in a temporary stocking, or a surgical glove sur-
rounded by ice slush. The back table with ice slush and preser-
vation fluid should be available until the fascia is closed, in the
event that it is necessary to cool the kidney again.

TRANSPLANT RENAL VEIN ANASTOMOSIS


The technical details of the anastomosis have been described
in Chapter 11, but one or two points are worth reiterating.
Unless there is a recipient history of factors predisposing to
venous thrombosis, systemic heparinization for the vascular
anastomoses is unnecessary in a dialysis-dependent patient,
even in the era of widespread use of synthetic erythropoi-
etin agents. The site of the iliac vein anastomosis can be
marked with a sterile surgical marking pen before applying
the venous clamps to reduce the risk of vein rotation during
clamp application. Accurate sizing of the venotomy length
prevents stretching of the end of the transplant renal vein to
accommodate a venotomy that is too long. Stretching leads
to a long, stenosed anastomosis. After opening the vein, Fig. 28.5 Ascending venogram shows a long stenosis of the right iliac
the surgeon searches for pairs of valve cusps and disrupts vein in a patient with a history of temporary hemodialysis cannulas and
them if they are adjacent to the anastomosis. A stay suture a previous thigh arteriovenous fistula.
is applied to the midpoint of at least one of the sides of the
venotomy to reduce the risk of catching the opposite wall of result in massive blood loss within minutes from the large and
the anastomosis with the continuous running vein suture. thin-walled labyrinth of pelvic and presacral veins that are
The external iliac veins in an obese recipient or a short, prone to retracting into the depths of the surgical wound. In
muscular male patient with a deep pelvis and almost vertically such instances, bleeding is best managed by carefully packing
disposed external iliac vein can be challenging, particularly for the depths of the wound and applying pressure, a request for
right-sided donor kidneys placed in the left iliac fossa. For ease blood products, and systematic control of venous bleeding by
of access, it is tempting to place the venous anastomosis close application of metal clips or polypropylene (Prolene) sutures.
to the inguinal ligament, but there is a risk of compression of A thrombosed or stenosed external iliac vein is prefer-
the renal vein during wound closure. Options include length- ably identified by color Doppler ultrasound scanning (CDUS)
ening of the donor renal vein or the sometimes difficult task of before surgery and should be considered in patients with a
mobilization of the external iliac vein by dividing the internal history of deep vein thrombosis (DVT), previous transplant
iliac vein and its tributaries. The surgeon should ensure that surgery, unilateral leg swelling, or emergency dialysis access
there are long stumps on the ligated veins. Lost ligatures can via the femoral vein (Fig. 28.5). When encountered at the
462 Kidney Transplantation: Principles and Practice

A B
Fig. 28.6 Computed tomography scan of male patient with avascular right living donor kidney transplant that failed because of chronic rejection. The
arterial anastomosis was to the right internal iliac artery. (A) Axial view of small avascular kidney transplant (arrow) adjacent to the common iliac artery.
(B) Oblique perspective of vascular reconstruction of aortoiliac arteries demonstrating proximal stump of right internal iliac artery (PRIIA), retrograde
filling of distal right iliac artery (DRIIA), and patent left internal iliac artery (LIIA).

time of surgery, the common iliac vein often has a preserved comparatively straightforward but dual arteries to a right-
lumen and can be used for the transplant renal vein anasto- sided kidney make positioning of the kidney difficult with-
mosis. Alternatively, the surgeon can close the wound and out kinking one or the other artery.
transplant the kidney into the opposite iliac fossa. Individual transplant surgeons will have their own
views about how best to manage multiple arteries of a liv-
ing kidney donor. Multiple arteries can be pantalooned on
TRANSPLANT RENAL ARTERY ANASTOMOSIS
the back table to create a single arterial orifice, minimizing
The extent of dissection of the iliac artery should be lim- anastomosis time. Alternatively, separate anastomoses can
ited to diminish the risk of disruption of adjacent lymphatic be formed, although warm ischemic time will inevitably
channels. If the internal iliac artery is to be used, the sur- be longer. Although vascular multiplicity leads to longer
geon fully mobilizes the bifurcation of the common iliac warm ischemic times and higher rates of delayed graft func-
artery and carefully examines the origin for an athero- tion, no difference in longer-term outcomes is seen, and this
matous plaque. Use of the internal iliac artery is avoided should not discourage the use of such kidneys.7
if the opposite side artery has been involved in a previous
transplant (Fig. 28.6). The bifurcation or trifurcation of the REPERFUSION
internal iliac artery should be preserved to reduce the risk
of buttock claudication. If both internal iliac arteries have Reperfusion is the high point of the transplant procedure—
been used for transplantation, claudication is inevitable as there is no turning back. Before completing the arterial
is impotence. anastomosis, air is excluded from the clamped vessels by
Arterial clamps are applied with care to avoid damage or injecting heparinized saline. Fixed retractors that might
rupturing plaques. Endarterectomy can often be avoided by compress proximal iliac vessels are reviewed and individ-
carefully selecting a soft segment of artery. To avoid kink- ual anastomoses are tested before revascularization of the
ing during wound closure, the renal artery length can be transplanted kidney. Imperfect anastomoses are managed
adjusted by resecting the donor aortic patch and implant- more easily beforehand (Fig. 28.7). The proximal arte-
ing as in a live donor kidney. Equally, the shortened artery rial clamp and venous clamps are released first. The last
of the right kidney can be anastomosed to the end of the clamp removed is the distal iliac artery clamp after systemic
internal iliac artery. This has the added advantage of deeper blood pressure has stabilized after reperfusion of the kidney.
placement of the transplant anastomoses, less tension on Observation of urine within a couple of minutes is a reas-
the short right renal vein, and easy positioning of the kid- suring sight—a pink, firm, and well-perfused kidney is the
ney after revascularization. next best thing. If neither is observed, the surgeon should
Multiple renal arteries are encountered more commonly actively look for problems. Kidneys from marginal donors
with the increasing popularity of laparoscopic living kid- or with long renal ischemia times may have a “blotchy” or
ney donation and the preference for the left kidney.1 At mottled appearance with dark, less well-perfused areas. An
least 20% of left kidneys have more than one artery after encouraging sign is the gradual reduction in extent of the
living donation. Small accessory renal arteries, particularly dark areas until the kidney is uniformly pink (Fig. 28.8).
at the upper pole, can be ligated without problem, but not A flaccid, poorly perfused kidney is reason for concern.
lower-pole arteries that often contribute blood supply to the Modern tissue typing and crossmatching techniques have
donor ureter.6 Anastomosis of two arteries close together essentially excluded hyperacute rejection as a cause (see
on an aortic patch of a left-sided deceased donor kidney is Chapter 10). The surgeon starts with inspection of the renal
28 • Vascular and Lymphatic Complications after Kidney Transplantation 463

segmental. The kidney is soft but not necessarily discolored.


Anecdotal reports suggest that placement of a swab gener-
ously soaked in papaverine around the artery and excision
of affected artery adventitial tissue may help. Alternatively,
and with permission of the anesthetist, the iliac artery distal
to the arterial anastomosis is clamped and diluted glycerol
trinitrate injected into the proximal iliac artery. Spasm is
usually self-limiting but can cause concern and warrant
systemic heparinization. If no arterial inflow problem can
be identified and systemic blood pressure is satisfactory, the
surgeon should be patient, particularly if the kidney swells
when the renal vein is temporarily occluded (Hume test).
A small incision into the capsule of the kidney followed by
evidence of bright arterial bleeding can also be reassuring,
as can an on-table ultrasound.
Catastrophic bleeding after removing all vascular clamps
is unlikely to occur if the anastomoses have been assessed
before revascularization. If present, however, it is usually
Fig. 28.7 Testing the integrity of the end-to-end right internal iliac venous in nature and either from a tributary vein, or worse,
artery to transplant renal artery anastomosis before revascularization from a disrupted venous anastomosis because of traction
of the kidney transplant. on a thin-walled and right-sided living donor kidney renal
vein. Because of the continuous nature of the suture, sim-
ple repair is usually impossible and, if attempted, results
in excessive blood loss or anastomotic stenosis, or both.
Removal of the kidney, reperfusion with preservation solu-
tion, and calmly starting all over is a sensible solution.
The observation of a tense, engorged, and pulsatile kid-
ney with marked capsular bleeding is indicative of venous
outflow obstruction. Causes can be a rotated or compressed
renal vein, apposition of the sides of the venous anastomosis
because of imperfect suturing, or inclusion of external iliac
vein valve cusps in the anastomosis. Because of the rela-
tively controlled situation, revision of the anastomosis usu-
ally can be undertaken within 10 minutes, after systemic
heparinization, clamping the renal artery, and exsanguina-
tion of the transplant. This can also be achieved by reclamp-
ing the iliac vessels, removing the ligature from the gonadal
or adrenal vein stump, and cannulating the iliac artery to
Fig. 28.8 Sigmoid colon separating the dual kidney transplants from flush the kidney. An uncommon cause of venous outflow
a marginal cadaver donor. The left-sided kidney was transplanted first obstruction is compression of the left common iliac vein as
and is of uniform appearance. The right kidney is of mottled appear-
ance 10 minutes after revascularization. Ten minutes later, it had the
it passes under the right common iliac artery, described as
same appearance as the left kidney transplant. the May-Thurner syndrome.8 Probably the extra 500 to
750 mL of blood per minute from the transplanted kidney
is enough to compromise the narrowed iliac vein at that
artery to exclude kinking or twisting and resolves it if pos- point. If recognized at time of surgery, a more proximal
sible by repositioning the kidney. Next is confirmation of venous anastomosis can be attempted or the internal iliac
pulsatility of the iliac artery proximal to the anastomosis artery divided to allow mobilization of the right common
and its continuity into the transplant renal artery to the iliac artery. If recognized after transplantation, endovascu-
hilum of the kidney. Interruption of flow can be due to an lar stenting of the iliac vein may be an option.
intimal flap, particularly in recipients with underlying arte- A disappointing observation on completion of the vascu-
rial disease and kidneys from older donors. Management is lar anastomoses is the finding of a well-perfused transplant
not easy. The most likely site of an intimal flap would be at but with the ureter pointing in the wrong direction, away
the anastomosis or the proximal clamp site. from the bladder. The kidney has been transplanted upside
The decision to revise the arterial anastomosis or the down, and this is more likely to occur with a living donor
“safety-first option” of removing the transplanted kidney kidney in the absence of the full length of renal vein and
and reperfusing with preservation solution can be difficult. the aortic patch to assist with orientation. One option is
If revision is undertaken, the recipient is heparinized, the to remove the kidney, reperfuse, and start again. Alterna-
transplant artery flushed with at least 50 mL of heparinized tively, the kidney can be left in place and, provided there
saline, and the renal vein clamped. is an adequate length of ureter, it can be provided with a
Extrarenal arterial spasm is a not uncommon finding more circuitous route to the recipient bladder. The latter
and likely the result of undue traction on the renal artery option may increase the risk of subsequent ureteric stenosis
during donor or implantation surgery. The spasm may be because of the longer, more tortuous course of the ureter.
464 Kidney Transplantation: Principles and Practice

POSITIONING THE KIDNEY AND The need for an additional arterial anastomosis is also
WOUND CLOSURE good reason for an early ultrasound in the presence of the
operating surgeon with knowledge of the surgical vascular
The ureteroneocystostomy should be the relaxing part of the anatomy. Patency of an accessory renal artery is difficult to
kidney transplant operation. The kidney is positioned to avoid determine in the early postoperative phase by observation
compression of the vascular pedicle and, all being well, urine of urine output alone. These smaller vessels are more prone
is being produced. A suction drain may be placed, although to thrombosis or kinking, and longer-term consequences
it is not always required and may not have any effect on the include poor graft function and hypertension. An avascu-
risk of wound complications.9 A drain is perhaps most use- lar segment of kidney can occur, at least initially, without
ful in the patient on peritoneal dialysis where delayed func- noticeable effect.
tion is expected, because it will allow the drainage of dialysis Because of the quality of modern ultrasound, indica-
fluid when an unidentified peritoneal breach is present, or tions for formal angiography in the early phase after kidney
in patients receiving sirolimus for primary immunosuppres- transplantation are few. They are perhaps limited to the
sion.10 Many patients on peritoneal dialysis will have high suspicion of proximal iliac artery disease or clamp injury
volume drainage even in the absence of dialysis or a breach, and an obese recipient in whom visualization of the renal
which may lead to prolonged hospital stay.11 artery and iliac vessels is not technically feasible. Computed
During apposition of the abdominal wall muscles, the tomography (CT) angiography is usually easier and faster
potential for kinking of the kidney transplant vasculature to obtain.
increases, particularly in thin patients receiving large kid-
neys, male patients with a narrow deep pelvis, a venous
DRAIN TUBE
anastomosis too close to the inguinal ligament, or an inci-
sion too close to the anterior superior iliac spine. Mobilizing Removal of a suction drain, when placed, should be a
the peritoneum in a medial direction off the undersurface straightforward task. Without suction it is withdrawn
of the anterior abdominal wall may help. Monitoring trans- slowly with a twisting motion to dislodge fatty tissue trapped
plant arterial perfusion during wound closure with ultra- in the small side holes of the drain as a result of the suction.
sound can be reassuring. Small pediatric kidneys have been known to undergo tor-
Failing this, a reliable “surgical escape” is to place the sion of the vascular pedicle on removal of the drain with
kidney into the peritoneal cavity by creating a longitudi- resultant loss of graft function.
nal window in the peritoneum, adjacent to the kidney and The timing of drain tube removal depends on the vol-
more anterior to the vascular anastomoses. The kidney is ume and nature of the drained fluid. It is not unusual to
positioned anterolateral to the cecum on the right side and record 100 to 200 mL of heavily blood-stained drainage
the sigmoid colon on the left side. The greater omentum can in the first few hours of transplantation. Drainage volume
be used to separate the bowel from the kidney. Percutane- can be an unreliable gauge of active bleeding, particularly
ous biopsy subsequently is still feasible after placement of if brisk. Patient discomfort, tachycardia, hypotension, and
local anesthetic agent at the level of the peritoneum. abdominal findings of an enlarging mass around the trans-
plant are indicators of a significant bleed requiring urgent
POSTOPERATIVE RECOVERY surgical exploration. Large-volume drainage of less heav-
ily blood-stained fluid generally indicates residual perito-
An experienced member of the surgical team remains with neal dialysate (if the peritoneum was breached), lymph, or
the recipient in the early recovery phase and until there is urine. Urine is excluded by biochemical analysis or absence
conclusive evidence of satisfactory perfusion of the trans- of glucose on dipstick testing.
planted kidney. The careful positioning of the kidney at time
of surgery can be undone readily by a restless recipient flex-
COMPARTMENT SYNDROME
ing the hips because of pain, urinary catheter intolerance,
and hypoxia, or an unhelpful radiographer who determines All can be well with a transplanted kidney while the recipi-
that the recipient should sit bolt upright for a mobile chest ent is in a supine or near-supine position. Ultrasound is also
x-ray. Transplanted kidneys producing urine at the end of the performed with the recipient in a supine position. However,
surgical procedure are easier to manage, particularly if urine when the patient is placed in a sitting or standing position,
is being produced in volumes that could not be achieved by downward movement of abdominal contents can cause
residual native kidney function. The better the urine volume, external compression of the transplanted kidney or change
the less likely that clots will form in the bladder. its position. Contributing factors include a large native poly-
If no urine has been seen on the operating table or in cystic kidney, heavy fat-laden small-bowel mesentery, and
recovery and the recipient is hemodynamically stable with greater omentum in a patient with truncal obesity. Size of
a central venous pressure of at least 5 cmH2O, ultrasound the transplanted kidney may also play a role.12 Hematoma,
examination is useful before the recipient leaves the oper- urinoma, lymphocele, or paralytic ileus can do likewise,
ating suite complex, particularly if difficulty was encoun- even with the patient in a supine position. The contribution
tered with kidney positioning during wound closure. Out of of the compartment syndrome to initial poor kidney trans-
routine working hours, it helps if the transplant surgeon is plant function should not be underestimated (Fig. 28.9).
adept with the use of an ultrasound machine dedicated to Reversible factors should be resolved without delay. A para-
the transplant unit. An inadequate arterial signal and sig- lytic ileus or pseudoobstruction of the large bowel can be
nificant collections are indications for an immediate return frustrating to manage in the first week after transplanta-
to the operating room. tion. The latter may require a rectal tube to deflate the large
28 • Vascular and Lymphatic Complications after Kidney Transplantation 465

Fig. 28.10 Postmortem preparation of recipient external iliac artery


and two donor renal arteries on an atheromatous donor aortic patch. It
demonstrates a small disruption of the anastomosis (arrow) that led to
catastrophic bleeding 10 days after transplant surgery.

Fig. 28.9 Computed tomography scan with coronal view of abdo-


men 24 hours after kidney transplantation. The perfusion of the kidney
transplant in the right iliac fossa was compromised by gross pseudoob-
struction of the large and small bowel.

bowel under supervision of the colorectal surgery team.


Surgical decompression with insertion of a mesh to relieve
pressure may be required.13

Hematoma
Hematoma formation is a not uncommon finding after
kidney transplantation during the initial inpatient period, Fig. 28.11 Computed tomography scan (without vascular contrast
particularly in anticoagulated recipients or those receiv- material) with coronal view of abdomen showing compression of the
ing antiplatelet agents, thymoglobulin, or plasmapheresis. kidney transplant by an anteriorly placed hematoma.
Most hematomas are small and insignificant ultrasound
findings that resolve spontaneously. Those associated with original wound is reopened and care taken to remove the
discomfort, hypotension, transplant dysfunction, and fall- hematoma, always being alert to the possibility of dislodg-
ing hemoglobin are not. Extreme examples are surgical ing the clot that is providing tenuous hemostasis at the site
emergencies after rupture of the kidney cortex or arterial of bleeding. Surgical exploration in the first day or so after
anastomosis disruption (Fig. 28.10). Others expand pro- transplantation for hematoma evacuation might locate
gressively within the retroperitoneal space with inevitable active bleeding from a hilar vessel, a retroperitoneal vein,
external pressure on the transplant and adverse effect on or divided abdominal wall muscle. Thereafter, a more com-
arterial blood inflow or venous outflow. The extent of the mon finding is a stable hematoma without obvious cause.
hematoma by CT scanning is best shown as a heteroge- Invariably, transplant perfusion and function improve after
neous crescentic peritransplant collection (Fig. 28.11). CT hematoma evacuation. Bruising in dependent subcutane-
findings change with time after the bleeding event, with ous areas lateral to and below the transplant, such as the
evidence of recent bleeding of more concern. Ultrasound labia or the scrotum, is often seen several days later. The
examination is appropriate to assess transplant perfusion risk of hematoma formation is increased by the use of anti-
but because of surrounding bowel gas is unreliable for coagulants, particularly in patients receiving heparin by
assessment of hematoma size. infusion for prophylaxis against vascular thrombosis.14
Percutaneous drainage of the hematoma is unlikely to Careful titration of heparin infusion rate to maintain an
be successful. Indications for surgical exploration of the activated partial thromboplastin time of 60 seconds is not
transplanted kidney include symptoms, progression of easy. The reported risk of need for surgical intervention in
size, ongoing blood loss, and transplant dysfunction. The patients heparinized after transplantation is 30% to 60%.
466 Kidney Transplantation: Principles and Practice

Heparinized patients positive for lupus anticoagulant are


especially difficult to manage with heparin.15 Greater safety
can be achieved with the use of thromboelastography to
direct judicious use of heparin, during and after transplant
surgery in patients at risk.16 Anecdotally at least, hema-
toma formation is more common in the presence of anti-
platelet agents such as aspirin and/or clopidogrel. They are
prescribed increasingly on a long-term basis by cardiologists
and nephrologists in patients with significant cardiovascu-
lar disease or in attempts to improve fistula patency. Their
use is not a contraindication to transplantation. However,
they do reduce the margin for surgical error and dictate the
need for meticulous hemostasis at time of surgery.

Vascular Thrombosis and


Thrombophilia Fig. 28.12 Color duplex ultrasound shows minimal blood flow into kid-
ney transplant as a result of almost complete occlusion by thrombus
Early kidney transplant loss as a result of thrombosis of of the transplant renal artery 5 days after transplantation. The trans-
planted kidney was not viable when explored soon afterward.
artery or vein is a devastating complication, with a 2% inci-
dence (see Fig. 28.1). Compared with other forms of vas-
cular surgery, the incidence of thrombosis is low, perhaps to minimize this risk in selected, highly sensitized recipients
because of the highly vascular nature of the kidney. The low with a negative donor lymphocytotoxicity crossmatch (see
incidence may also support the traditional view that renal Chapter 10). ATN attributable to the reperfusion injury
failure is associated with a bleeding tendency secondary to is also associated with endothelial changes, and, together
platelet and clotting factor dysfunction.17 Arterial throm- with hydronephrosis, is associated with increased intrare-
bosis or infarction of a denervated kidney is often painless nal pressures, making perfusion of the transplanted kidney
and heralded only by loss of graft function. By the time the more difficult. Recipients dependent on peritoneal dialysis
diagnosis is confirmed by imaging, kidney salvage is not a before transplantation are more likely to have thrombotic
practical option (Fig. 28.12). Interruption of the venous complications, likely due to intravascular hypovolemia.
drainage can be spectacular with graft rupture and bleed- The introduction of recombinant human erythropoietin
ing. It has an equally disappointing prospect for kidney sal- (rEPO) has revolutionized the treatment of anemia associ-
vage because of the rapidity of the process after occlusion of ated with end-stage kidney disease, reducing the need for
the renal vein has occurred. Thrombotic complications are routine blood transfusion and improving quality of life and
minimized by identification and management of risk at the patient survival. The rEPO dose is titrated to provide recipi-
time of transplantation. ent hemoglobin in the range of 100 to 120 g/L. With higher
Thrombosis of the kidney vasculature is the end result of hemoglobin values, there is an increased risk of adverse
stasis, endothelial changes, and procoagulant factors and cardiac events. Nevertheless, the current widespread use of
can be multifactorial. Causes of stasis are largely technical rEPO in patients presenting for kidney transplantation has
in nature and readily identifiable at the time of transplant not resulted in an increased risk of vascular thrombosis.18
exploration. They include poorly constructed anastomoses, Erythrocytosis, defined as hematocrit greater than 51%
malpositioning of the transplant, rotation of the kidney, or or hemoglobin greater than 160 g/L, occurs in 10% to 15%
external compression. Recipient hypovolemia and inad- of recipients 6 months to 2 years after kidney transplan-
equate cardiac output, for whatever reason, are contribu- tation.20 About one-quarter regress spontaneously, with
tory but not causal factors. The contribution of intrarenal the remainder persisting for several years and remitting
causes, such as acute vascular rejection and acute tubular as graft function diminishes. Thromboembolic events and
necrosis (ATN), is less quantifiable, but can be diagnosed by symptoms of lethargy, malaise, and headache necessitate
histologic examination provided viable cortical tissue can repeated venipuncture and may be necessary in up to 30%
be obtained. Because this is often not the case, intrarenal of these patients. The problem is more common in male
causes are probably underestimated and underdiagnosed. patients, smokers, and patients with a rejection-free course.
Epidemiologic studies have attempted to identify other Erythropoietin levels are usually in the normal range. By
risk factors, particularly those amenable to preventive strat- chance, patients introduced to small doses of an angioten-
egies.17,18 Those that cannot be modified are recipient and sin-converting enzyme inhibitor for the management of
donor age, recipient and donor vascular pathology, diabe- hypertension were noted to have progressive reduction of
tes mellitus and, at least in the view of some recipients, mor- hematocrit to more normal levels. The suggestion is that
bid obesity. A large registry-based and case-matched study angiotensin II is a growth factor for red blood cells.
has shown that half of all cases of kidney transplant vas-
cular thrombosis occur in repeat transplant recipients.19
THROMBOPHILIC FACTORS
The implication is that transplanted kidneys in the set-
ting of retransplantation are more likely to have endothe- After exclusion of technical causes in a hemodynamically sta-
lial inflammation and development of microthrombi after ble kidney transplant recipient, thrombosis may be explained
exposure to the recipient immune system. Strategies exist by one of the numerous hypercoagulable or thrombophilic
28 • Vascular and Lymphatic Complications after Kidney Transplantation 467

states. Many are inherited, but they are more frequently Prolonged use of heparin can lead to the development
acquired.18,21 These include deficiencies of antithrombin III, of measurable antibodies against platelet factor 4 (PF4) in
protein C, and protein S, each occurring in less than 1% of up to 20% of patients. Heparin-induced thrombocytopenic
the dialysis patients. When a thrombotic event of any kind syndrome (HITS) is an immune-mediated thrombocyto-
occurs in a patient older than 45 years and in the absence of penia together with thrombotic complications occurring
a family history, these deficiencies are unlikely. within 24 hours of a patient’s reexposure to heparin during
Inheritance of factor V Leiden (FVL) or prothrombin the transplant procedure. The subsequent immune com-
G20210A mutations can increase the risk of thrombo- plex activates platelets, predisposes the patient to clotting
sis, usually venous, of the transplant vasculature by at of veins and arteries, and causes the platelets to be con-
least threefold. FVL mutation is present in 2% to 5% of the sumed. The diagnosis of HITS for the first time in a trans-
normal population and is not more common in patients plant recipient is uncommon, but is likely underreported,
with kidney disease. It is found, however, in 15% to 20% particularly in preemptive live donor kidney recipients and
of patients with venous thromboembolism and 60% of patients treated by peritoneal dialysis. A proven past diag-
patients with a family history of thromboembolism. When nosis of HITS (involving measurable PF4 antibody) man-
these mutations are present in kidney transplant recipi- dates absolute avoidance of any form of heparin during
ents, the risk of major thrombotic events, particularly renal the transplant procedure. Anticoagulation can be provided
vein thrombosis (RVT), is 40%.22 The presence of FVL or with direct thrombin inhibitors such as lepirudin, argatro-
prothrombin G20210A mutations is also associated with ban, or bivalirudin.
shorter graft survival, probably as a result of greater micro-
vascular thrombosis in renal vessels affected by rejection or RENAL VEIN THROMBOSIS
intimal thickening. A case could therefore be made for rou-
tine genetic screening for these polymorphisms in patients Occlusion of the renal vein by thrombus soon after trans-
awaiting a renal transplant. It should be mandatory if there plantation is now an unusual event and invariably associ-
is a history of thromboembolism. ated with a technical problem. However, in the era of high
The presence of acquired antiphospholipid antibodies cyclosporine dosing, the incidence of the seemingly spon-
(APAs), including anticardiolipin antibody and lupus anti- taneous RVT was as high as 6% and occurred classically
coagulant, is common in patients awaiting transplantation. toward the end of the first week of transplantation in an oth-
Although present in about 10% of patients, related clinical erwise uncomplicated transplant kidney.24 Witnessing the
events are less common. When associated with a history of dramatic presentation over a couple of hours is an unfor-
thrombotic events, these patients, often with systemic lupus gettable experience. Rapid onset of oliguria and hematuria
erythematosus, are labeled as having APA syndrome. They is accompanied by graft enlargement and rupture associ-
have a universal incidence of graft loss to thrombosis when ated with extreme patient discomfort and life-threatening
prophylaxis is not employed. Equally, anticoagulation after bleeding. RVT can happen during the course of a morning
transplantation offers protection against graft loss.23 The ward round. The Oxford Transplant Unit response almost 2
presence of APAs without a history of thrombosis is seem- decades ago was to introduce daily aspirin from the time of
ingly not a problem. surgery, the effect of which was to decrease the incidence of
RVT from 5.6% to 1.2%.24
CONTRIBUTION OF IMMUNOSUPPRESSIVE The ultrasound findings are of a swollen graft with a cres-
cent of clot along the convex margin of the kidney and cov-
AGENTS
ering a longitudinal rupture of the cortex. In this setting,
The introduction of cyclosporine, usually at doses of 15 mg/ reverse diastolic flow of the arterial waveform is diagnostic
kg or more, was associated with an increased incidence of (Fig. 28.13A). Potentially, the transplant can be saved after
graft thrombosis, particularly RVT, in the first week after early diagnosis if the patient is taken directly to the operat-
transplantation.20 Cyclosporine was subsequently shown ing room by the surgical team (Fig. 28.13B). The operative
to have procoagulant properties, increasing factor VIII and findings will match the ultrasound description along with
causing release of tissue factors from monocytes and von active arterial bleeding from the ruptured cortex. The pre-
Willebrand factor and P-selectin from endothelium. This is sentation, apart from evidence of reverse diastolic flow, is
likely a dose–response effect, for in recent years meticulous similar to the description of graft rupture seen with severe
cyclosporine drug dosing based on drug level monitoring ATN in the era before brain death legislation. Some sur-
has resulted in fewer reports of RVT. Mammalian target of geons of that era performed prophylactic division of the
rapamycin (mTOR) inhibitors are not thought to contribute kidney capsule to allow the kidney to cope better with the
to thromboembolic events after kidney transplantation. inevitable parenchymal swelling associated with tubular
Hemolytic uremic syndrome/thrombotic thrombocytope- necrosis.
nic purpura is an infrequent but well-described complication The technical causes of RVT include an iliac vein valve
of cyclosporine use. The diagnosis, seen soon after transplan- cusp sutured into the anastomosis, kinking of the long left-
tation, is based on deteriorating renal function, decreasing sided donor kidney vein, and compression of the short right
platelet count, and characteristic glomerular thrombi seen donor renal vein.
in core biopsy specimens. Most resolve with discontinua- Surgical management of an acutely occluded renal vein
tion of cyclosporine and conversion to tacrolimus. Reports is difficult. If identified in the early period after transplanta-
also describe the same presentation with tacrolimus, which tion, simple reopening of the wound and making more space
responds with conversion to cyclosporine. The alternative for the transplanted kidney may be associated with a rapid
would be to introduce an mTOR inhibitor. improvement in appearance of the kidney. Placement of
468 Kidney Transplantation: Principles and Practice

A B
Fig. 28.13 Color Doppler ultrasound findings in a patient presenting with acute renal vein thrombosis 24 hours after kidney transplantation.
(A) Immediately before renal vein thrombectomy and demonstrating reverse diastolic flow in the main transplant renal artery. (B) Restoration of normal
flow pattern 2 hours after urgent graft exploration, thrombectomy, and disruption of a valve leaflet caught in the venous anastomosis to the external
iliac vein. MRA, magnetic resonance angiography; MRV, magnetic resonance venography.

the kidney in the peritoneal cavity or use of a mesh to close transplant artery and the presence of worsening hyperten-
may prevent the same from happening again. If thrombus sion and poor graft function are good starting points.
is present in the renal vein of a right-sided donor kidney,
removal of the kidney and reperfusion with preservation SEGMENTAL ARTERIAL THROMBOSIS
solution may be the only practical option. The donor kid-
ney is retransplanted with consideration given to provision About 15% of deceased donor kidneys will have more than
of greater mobilization of the iliac vein and more proximal one renal artery, and the number is likely greater for living
siting of the venous anastomosis. donor kidneys in the era of laparoscopic donor nephrectomy.
RVT is uncommon beyond the early period after trans- The arterial supply to the kidney parenchyma has no col-
plantation, suggesting that transplant renal vein is com- lateral circulation. Hence, failure to anastomose an acces-
paratively resistant to anastomotic stenosis and perhaps sory artery, or thrombosis of an accessory artery or branch,
protected by high renal vein blood flow. It can be seen in will lead inevitably to a wedge-shaped infarct of the kidney.
a subacute situation associated with secondary causes, Inability to transplant a small upper-pole accessory artery or
such as iliofemoral vein thrombosis, de novo membra- branch is a not uncommon event and is usually of little sig-
nous nephropathy, glomerulonephritis, and thrombophilic nificance. It will be obvious at the time of transplantation as
states. Because of the time of presentation many months an ischemic area on the cortex of the kidney and a small per-
after surgery, percutaneous combined mechanical and fusion defect may be apparent with ultrasound examination.
chemical thrombolysis is feasible.25 Larger accessory vessel or branch vessel occlusion will have a
more significant effect on the transplanted kidney.
In the acute phase, evidence of segmental infarction is a lac-
RENAL ARTERY THROMBOSIS
tate dehydrogenase level greater than 500 IU/L.26 Ultrasound
Spontaneous thrombosis of the renal artery is uncommon. will demonstrate an obvious wedge-shaped area of absent
It usually is due to technical reasons such as arterial kinking renal perfusion (Fig. 28.14A). A core biopsy will provide a his-
or torsion of the renal vascular pedicle. Contributing factors tologic diagnosis of necrotic kidney parenchyma. If the necro-
include poor cardiac output, hypotension, intravascular sis is full thickness, from capsule to calyceal system, urine
volume depletion, and thrombophilic states, or increased leakage and formation of a urinoma may occur from about
intrarenal pressure resulting from ATN, hydronephrosis, the fifth day after transplantation. It is not inevitable, and if
or cellular rejection. The renal arteries of kidneys that have not apparent by the end of the second week of transplantation,
failed because of chronic rejection often remain patent for urine leakage is unlikely to occur (see Chapter 29 for manage-
many years. ment). Longer term, the infarcted segment of the kidney trans-
Apart from loss of graft function, the signs and symptoms plant will be replaced by scar tissue (Fig. 28.14B).
are negligible. Kidney graft loss is “silent.” The diagnosis is
made by ultrasound or surgical exploration. Arterial throm- THROMBOSIS PREVENTION STRATEGIES
bosis is a terminal event that can be averted only if arterial
inflow is considered as a cause of poor graft function, and Acknowledging that vascular thrombosis is a multifactorial
immediate intervention undertaken; hence, the importance event, prevention necessitates the need for a combination
of recognition of an arterial problem before thrombosis of general and specific measures that make up Virchow’s
occurs. Routine ultrasound assessment of the extrarenal classic triad of contributing factors. Endothelial injury and
28 • Vascular and Lymphatic Complications after Kidney Transplantation 469

A B
Fig. 28.14 Color Doppler ultrasound demonstrating wedge-shaped avascular area (arrow) of lower pole of a deceased donor kidney transplant with
four renal arteries, one of which was shown to be thrombosed at the time of subsequent transplant exploration. (A) Seven days after transplantation.
(B) Scarred infarcted area 3 months later.

inflammation will be reduced by minimizing factors leading


to ATN at the multiorgan donor procedure, avoidance of
prolonged cold and warm ischemia, and early core biopsy
in a poorly functioning kidney to diagnose and aggressively
manage vascular and antibody-mediated rejection. Stasis
at the time of and after transplant surgery will be minimized
by optimal surgical technique and fluid management.
The recognition of thrombophilic states as a major con-
tributor to vascular thrombosis after kidney transplantation
introduces the possible need for routine screening and, in
turn, directed therapy to reduce the risk of thrombosis. Some
centers do but there is no consensus for either strategy. Rou-
tine screening is expensive, and most thrombophilic states
are rare. The most common are APAs, but, in the absence of a
previous thrombotic event, the risk of allograft thrombosis is
low. It would therefore be reasonable to limit investigation to
potential recipients with previous or family history of throm-
botic events, including deep and superficial vein thromboses,
pulmonary emboli, thrombosed fistulas, multiple occlusions
of central venous dialysis catheters, and problematic clotting
Fig. 28.15 Magnetic resonance imaging (MRI) angiogram demonstrat-
of dialysis lines. To this list could be added patients undergo- ing torsion of a transplant renal artery of a kidney transplant in the right
ing preemptive transplantation with a living donor kidney. iliac fossa. The kidney had been placed in an intraperitoneal position
Management of thrombophilic states is individualized to the when transplanted 3 months earlier in a patient receiving mammalian
patient’s risk factors. For known thrombophilia and a history target of rapamycin (mTOR) inhibitor immunosuppression. The kidney
of clinical events, perioperative heparinization followed by transplant was subsequently lost despite emergency exploration after
MRI angiography.
long-term anticoagulation with warfarin has proven efficacy,
including successful retransplantation.27 The risk of bleeding
seems acceptable in view of the incidence of thrombotic com- abdominal musculature without compromising the transplant
plications. For recipients without known risk factors, a sys- kidney vasculature. It is also performed routinely for diabetic
tematic review exploring the role of low-dose aspirin in kidney patients receiving simultaneous pancreas and kidney trans-
transplant recipients demonstrated a 43% reduction in the plants (SPK) through a midline incision. For these patients, the
risk of allograft failure, 89% reduction in the risk of thrombosis kidney is placed on the left side, lateral to the sigmoid colon,
and 28% reduction in cardiac events or mortality when aspi- perhaps explaining why kidney transplant torsion is rarely
rin was prescribed.28 However, the number of included studies reported after SPK.
was small and data regarding risk of bleeding were limited. Because of its very low incidence, the diagnosis of torsion is
often overlooked and possible warning signs of pain, nausea,
and oliguria are missed. Imaging is likely to show absence or
Torsion very limited perfusion of the kidney transplant. A review of 16
cases described a successful outcome after graft exploration of
Rotation of the kidney about its own vascular pedicle only 25%, and recommends prophylactic nephropexy to prevent
occurs when placed in an intraperitoneal position (Fig. 28.15). torsion of kidneys placed in the peritoneal cavity, particularly
The need for this is usually prompted by difficulty closing the if immunosuppression includes use of a mTOR inhibitor.29
470 Kidney Transplantation: Principles and Practice

A B
Fig. 28.16 Computed tomography angiogram of the left brachial artery in a transplant recipient presenting with clinical evidence of emboli in digital
arteries 10 years after transplantation and 5 years after ligation of left brachiocephalic fistula. (A) Thrombus in two fusiform aneurysms of the brachial
artery measuring 5 and 4 cm in diameter. (B) Arterial reconstruction of the lumen brachial artery.

Vascular Access Thrombosis aneurysmal with tension in the artery wall proportional to
the radius and arterial pressure. Intraluminal mural throm-
The arteriovenous fistula acts as a lifeline for dialysis-depen- bus forms, from which emboli head toward the distal arteries.
dent patients, with effective vascular access surveillance Fig. 28.16 represents an extreme example of this scenario.
being clinical examination along with longitudinal assess-
ment of flow, pump speeds, and pressure readings.30 Care
must be taken when positioning the patient on the operat- Deep Vein Thrombosis
ing table and wrapping the arms to prevent compression
and thrombosis. After transplantation, the fistula provides After any major surgery, a hypercoagulable state persists
reassurance for the recipient with delayed graft function, for 4 weeks. An early retrospective Oxford study based on
and even after successful transplantation, recipients and clinical findings in a kidney transplant population in which
clinicians are reluctant to ligate. However, with less surveil- specific DVT prophylaxis was not used showed an incidence
lance, progressive intimal hyperplasia leads in the majority of 8.3% in 480 patients. However, the peak incidence was
to eventual and “silent” loss of the fistula flow that is not not early after transplantation but in the fourth month and
worthy of resuscitation. In others, the size of the native vein usually associated with an event necessitating bed rest or
fistula continues to grow, with the feeding artery and vein pelvic pathology, such as a lymphocele31 (Fig. 28.17).
continuing to adapt to the low-resistance fistula circuit Risk of thromboembolic events after renal transplantation
by becoming progressively more ectatic and tortuous. In is reported to be eightfold higher than the general popula-
turn, cardiac output increases progressively with 3, 4, and tion.32 Risk is highest during the first year after transplant,
more liters of blood passing through the fistula anastomo- but remains elevated even beyond this time. Risk factors
sis each minute. Tortuosity may lead to progressive kinking include hospitalization, use of sirolimus immunosuppression,
of the fistula vein and eventual stasis, and thrombosis that and anemia, with use of renin-angiotensin system inhibitors
extends proximally. Painful thrombophlebitis intervenes conferring protection. Proximal extension of an iliofemoral
and anticoagulation may be necessary. DVT is an uncommon event, probably because of the volume
In the current era of good long-term transplant graft func- of blood entering the iliac vein from the transplanted kid-
tion, a case can be made for a more proactive approach to ney. Nevertheless, it can happen, presenting with dramatic
ligation of a problematic native vein fistula after transplan- ipsilateral leg swelling followed by loss of graft function. It is
tation, particularly in patients with ectatic feeding arteries associated with a poor outcome if not recognized quickly.33
or concerns about the effect of the fistula circuit on cardiac Suggested management is anticoagulation with heparin,
output. If left, the feeding artery will become progressively placement of a temporary caval filter via the contralateral
28 • Vascular and Lymphatic Complications after Kidney Transplantation 471

Fig. 28.18 Left-sided kidney from an obese deceased donor with an


accessory lower-pole renal artery (cannulated) providing a branch
(arrow) to supply the ureter (held by forceps).

kidneys from obese donors (Fig. 28.18). Failure to preserve


increases the risk of necrosis or stenosis of the distal end of the
ureter. Equally, preservation of the relationship between the
lower-pole accessory artery and the ureter can compromise
the ureter by causing external compression after heterotopic
positioning of the transplant, as demonstrated in Fig. 28.19.
The problem is usually diagnosed when the recipient pres-
ents with a hydronephrosis after removal of the ureteric stent
placed at the time of transplantation. Surgical correction is
challenging and involves either anastomosis of the native
Fig. 28.17 Computed tomography scan (coronal view) with vascular ureter to the transplant renal pelvis or a new transplant
contrast material demonstrates a small lymphocele compressing the
distal right external iliac vein with resultant thrombosis of the more dis-
ureteroneocystostomy to be performed after mobilization of
tal common femoral. DVT, deep vein thrombosis. bladder and viable transplant ureter (see Chapter 29).

femoral vein, followed by thrombolysis through a cannula Mycotic Aneurysm


placed in the ipsilateral popliteal vein with the patient lying
prone on the interventional radiology table. An infected false aneurysm involving the anastomosis of the
The study referenced earlier suggests that routine use donor aortic patch to the iliac artery is an uncommon but poten-
of thromboprophylaxis is warranted, especially during tially lethal vascular complication resulting from bacteremic
periods of hospitalization. Prophylactic measures include shock or massive acute blood loss. It presents in the second or
the fitting of below-knee antithrombosis stockings before third week after transplantation as an acute febrile illness with
surgery and the use of intermittent calf compression dur- possible evidence of distal thromboemboli in the ipsilateral lower
ing surgery. They are considered to be as effective as sub- limb. Alternatively, it can be a ligated transplant renal artery
cutaneous heparin, provided that the stockings are worn stump infected at the time of graft nephrectomy. A site of infec-
throughout the inpatient stay, and early ambulation and tion may be identified elsewhere, such as an infected venous
calf exercises are undertaken. However, caution must be cannula. The organism can be a bacterium or fungus that lodges
exercised in the use of compression stockings in a popula- in a defect in the intima of the aortic patch caused by suturing
tion with high incidence of preexisting peripheral vascu- or ulceration of atheromatous plaque. Subsequent artery wall
lar disease. In these patients, subcutaneous heparin can infection leads to anastomosis rupture. If contained locally, an
be added, with unfractionated preferred to long-acting infected false aneurysm is obvious on ultrasound examination.
fractionated heparin because of its ability to reverse in There is also evidence that some mycotic aneurysms may
situations such as troublesome hematuria or the need for result from organisms of donor origin, particularly in the
a kidney transplant biopsy, and the risk of accumulation presence of candidal infection.34 Routine culture of organ
of low molecular weight heparin in patients with delayed preservation fluid is recommended, with the detection of
graft function. candida resulting in prompt antifungal therapy.35
Management of mycotic aneurysm is individualized with
life-saving measures undertaken first.36,37 Nephrectomy is
Vascular Causes of Ureteric usually undertaken with repair of the arterial defect using
Complications a vein patch and followed by prolonged administration of
appropriate antimicrobial agents. Reports also exist of suc-
The transplant ureter blood supply is totally dependent on cessful local or endovascular repair of small, unruptured
the renal artery, and if present, an accessory lower-pole mycotic aneurysms together with prolonged antimicrobial
artery. The relationship between an accessory vessel and ure- use. Although placement of a covered stent across the iliac
ter is unpredictable and difficult to preserve, particularly for artery in the presence of an acute bleed can be life-saving,
472 Kidney Transplantation: Principles and Practice

A B
Fig. 28.19 External compression of the proximal transplant ureter in a patient receiving a left-sided living donor kidney with two renal arteries sepa-
rately transplanted into the right iliac fossa. (A) The relationship of the transplant ureteric stent (arrow) to the lower-pole accessory renal artery, which
is anastomosed to the distal right external iliac artery. The main renal artery (MRA) is anastomosed end to end to the right internal iliac artery. (B) Retro-
grade pyelogram to investigate hydronephrosis 2 weeks after elective removal of the transplant ureteric stent.

A B
Fig. 28.20 Color Doppler ultrasound in a transplant recipient to investigate anuria after a protocol kidney core biopsy 3 months after transplantation.
(A) Large subcapsular hematoma (arrow) compressing kidney parenchyma. (B) Hilar artery waveforms demonstrating poor arterial blood flow into the
transplant.

there is a risk of colonization of the stent with subsequent not major, complications.38 Hematoma formation is com-
erosion and recurrent bleeding.36 Surveillance is essential, mon, if not inevitable, in the first week or two after trans-
ideally with early surgical repair and removal of the stent. plantation. Clot is seen outside of and adjacent to the kidney
transplant and often extending upwards in the retroperito-
neal plane. CT scanning without vascular contrast provides
Biopsy-Related Vascular a better imaging than ultrasound, which tends to underes-
Complications timate the extent of the hematoma. Biopsy of an established
kidney transplant can be associated with a subcapsular
A needle core biopsy gun with a small spring-loaded hematoma that can quickly compromise transplant func-
18-gauge needle can cause serious injury to a renal trans- tion and viability (Fig. 28.20).
plant even in experienced ultrasound-guided hands. Microscopic hematuria is also an almost universal find-
Informed consent is taken beforehand. Cessation of anti- ing after core biopsy. Macroscopic hematuria is seen after
platelet agents is likely to reduce the incidence of minor, but about 10% of percutaneous biopsies. Problematic bleeding
28 • Vascular and Lymphatic Complications after Kidney Transplantation 473

A B
Fig. 28.21 Incidental finding of an upper-pole biopsy-related false aneurysm at time of computed tomography angiography. (A) Vascular
reconstruction demonstrating 1-cm aneurysm (arrow) and draining arteriovenous fistula. (B) Selective angiography 3 months later to treat the then
2.5-cm partially thrombosed aneurysm and fistula (arrow). Both spontaneously thrombosed during selective angiography.

is usually self-limiting with bed rest, although retrograde


ureteric stenting may be necessary. Small false aneurysms
and arteriovenous fistulas within the transplant kidney are
less common. They are shown by ultrasound examination
and CT angiography, usually as incidental findings (Fig.
28.21). If an acute problem, a regimen of bed rest, inter-
mittent ultrasound with local compression, and temporary
cessation of antiplatelet agents and heparin is usually suc-
cessful in managing a false aneurysm. Occasionally, ultra-
sound will detect an arteriovenous fistula between bigger
vessels within the kidney. They are usually asymptomatic,
although an impressive bruit may be present on ausculta-
tion. In most cases conservative management is successful,
even for large fistulas, as shown in Fig. 28.22. Interven-
tional embolization is a more practical option than open
surgery to control intrarenal bleeding into the urinary col-
lecting system, or to treat significant vascular steal from the
kidney transplant.

Transplant Renal Artery Stenosis


Transplant renal artery stenosis (TRAS) is the most com-
mon vascular complication after kidney transplantation.
Although the etiology is variable, it is a potentially treat-
able cause of hypertension and graft dysfunction. If severe Fig. 28.22 Angiogram of a left-sided kidney transplant shows rapid
and left untreated, it can lead to graft loss or may be fatal. passage of vascular contrast material into the common iliac vein,
consistent with a large intrarenal arteriovenous fistula secondary to a
percutaneous 14-gauge core biopsy of the kidney. The fistula was man-
DEFINITION AND INCIDENCE aged conservatively.

There is no consensus definition of TRAS. At one end of


the spectrum is the classic presentation of a bruit over the reduction in renal artery lumen diameter in a normotensive
transplant, refractory hypertension, deteriorating renal patient in the absence of graft dysfunction. In between is the
function, life-threatening congestive cardiac failure sec- common finding of hypertension, which is multifactorial
ondary to fluid retention, and dramatic reversal by correc- and an independent risk factor for long-term graft survival
tion of the stenosis.39 It presents most commonly 3 months (see Chapter 30). Hence, any measure to improve blood
to 2 years after transplantation with symptoms and signs pressure control may be valid provided the intervention has
caused by activation of the renin–angiotensin system.40 few inherent risks, and even if it may not provide measur-
At the other end of the spectrum is the incidental finding able benefit for the kidney transplant recipient in the short
of a stenosis on ultrasound examination of 50% or greater to medium term, it may in the long term.
474 Kidney Transplantation: Principles and Practice

Fig. 28.23 Computed tomography scan with vascular contrast and


three-dimensional reconstruction showing transplant renal artery ste- Fig. 28.24 Computed tomography angiogram with vascular recon-
nosis (TRAS) (arrow) distal to anastomosis to the internal iliac artery 2 struction of a left-sided deceased donor kidney anastomosed to the
months after living donor kidney transplantation. The stenosis is likely bifurcation of the right common iliac artery. It demonstrates two kinks,
due to intimal fibrosis. one near the anastomosis and a lesser kink closer to the hilum.

The reported incidence of TRAS varies widely from 1% anastomotic stenosis, particularly involving the end of the
to 23% depending on the definition used. The higher figure renal artery, as is the case for living donor kidneys, prob-
dates back to Hamburger’s 14-year experience, published ably represents fibrosis and intimal hyperplasia in response
in 1973.41 His transplant recipients underwent formal to damage to the renal artery at the time of donation or
angiography on a routine basis. Ultrasound examina- implantation surgery, or an intimal flap (Fig. 28.23).
tion is the modern-day equivalent. Its safety and low cost Kinking or twisting of the renal artery of the heterotopic-
make it an indispensable tool in the transplant clinic as the positioned kidney at time of wound closure probably occurs
first-line screening tool to investigate graft dysfunction. more frequently than appreciated by transplant surgeons.
It provides qualitative and quantitative assessment of the The kink occurs either at the apex of the curve of the artery
kidney and has made a major contribution to the continu- or near the anastomosis where the artery is comparatively
ing improvement in graft survival.42 Protocol-driven ultra- fixed in position, or both (Fig. 28.24). A twist in the trans-
sound examination of the transplant renal artery will also plant renal artery is more likely to be seen nearer the hilum
raise awareness of unappreciated arterial pathology con- of the kidney.
tributing to the cause of hypertension. The long and more diffuse stenoses tend to occur later
A reasonable definition of TRAS is one based on a diagno- and have been attributed to immune-mediated endothelial
sis of hypertension requiring increasing antihypertensive injury with progressive intimal proliferation, particularly if
therapy, with or without deterioration in graft function, concentric in nature. Multiple stenoses, often associated with
in the presence of a renal artery stenosis which, when cor- renal artery branching, probably fit into the same category
rected, results in improvement of blood pressure control or (Fig. 28.25). The reported temporal association with vas-
renal function, or both. If such a definition were used, the cular rejection and subsequent stenosis is inconclusive.45 A
incidence of TRAS would be closer to 1% than to 23%. single-center study of 27 patients with TRAS showed a sig-
nificant association by multivariate analysis with cytomega-
PATHOGENESIS lovirus (CMV) infection and delayed graft function.46 CMV
infection is thought to trigger endothelial damage. It has a
The stenosis is usually situated near the anastomosis of the similar role in the development of cardiac allograft vascu-
renal artery to an iliac artery and can be short, diffuse, or at lopathy. An example of stenosis that could be attributed to
multiple sites, and occur at different times, in line with the CMV is demonstrated in Fig. 28.26. The recipient presented
multiple possible causes for TRAS. In a large series of TRAS, with severe hypertension requiring three antihypertensive
Voiculescu et al. reported that most stenoses are identified agents, elevated creatinine, ultrasound findings of elevated
in the first 6 months.43 Fibrosis accounted for 40%, donor peak systolic velocities (PSVs) in both branches, fever, and
artery atherosclerosis for 27%, and renal artery kinking CMV viremia. With reduced immunosuppression and pro-
for 21%. Stenoses at the anastomosis site are more likely longed use of antiviral agents, hypertension resolved and
to be technical and apparent from time of transplantation ultrasound findings returned to normal 3 months later.
and probably stable. End-to-side anastomoses may be more Atherosclerosis, occurring either de novo or already
of a problem than end-to-end anastomoses.44 Progressive present in the donor renal artery (Fig. 28.27), is an
28 • Vascular and Lymphatic Complications after Kidney Transplantation 475

Fig. 28.25 Angiogram demonstrating multiple stenoses of the renal


artery branches (arrow) in a kidney transplanted 2 years previously and
complicated by rejection.

Fig. 28.27 Computed tomography angiogram of kidney transplant


anastomosed to the left external iliac artery (EIA) in a patient with
poor kidney function 2 weeks after transplantation and hypertension.
It demonstrates an intimal flap (arrow) of the proximal EIA consistent
with a vascular clamp injury at time of surgery. It was subsequently
managed with an endovascular stent. A less significant second intimal
injury is noted adjacent to the anastomosis.

uncommon cause of TRAS. Equally, an arterial stenosis


or obstruction anywhere in the arterial tree upstream
from the transplanted kidney can produce the same clin-
ical presentation as TRAS. Many transplant recipients,
particularly smokers and older patients with maturity-
onset diabetes, have progressive diffuse arterial disease
at time of successful transplantation. A stenosis can be
precipitated by a vascular clamp injuring an already dis-
eased iliac artery at the time of transplant surgery (Fig.
28.28). Of clinical relevance in this setting is the com-
bined history of progressive claudication symptoms and
worsening hypertension. An iliofemoral bruit may be
present, together with a weak or absent femoral pulse, as
demonstrated in Fig. 28.29.

PATHOPHYSIOLOGY: “ONE KIDNEY, ONE CLIP”


In 1934 Goldblatt and colleagues published their seminal
study on the hypertensive effect of partial reduction of the
blood flow to a kidney in dogs by applying a silver clip to
one of the two renal arteries.47 They proposed the existence
of a pressor substance released by the ischemic kidney. Over
Fig. 28.26 Computed tomography angiogram with vascular recon-
the next 25 years, others subsequently defined the renin–
struction of a living donor kidney demonstrating stenoses in both main angiotensin system, with renin being the hormone released
renal artery branches 3 months after transplantation. The main renal from the ischemic kidney.48 Elevated renin levels are found
artery is anastomosed to the internal iliac artery. in the venous blood of the ischemic kidney. Its pressor effect
476 Kidney Transplantation: Principles and Practice

is driven by the direct pressor effect of angiotensin II, with


excess salt and water excreted by the nonischemic good kid-
ney, and is treated by inhibitors of the renin–angiotensin
system. This does not apply in the kidney transplant setting.
An analogous situation to the transplanted kidney with
a hemodynamically significant renal artery stenosis was
the effect of applying one clip to the renal artery in a dog
with one kidney (“one kidney, one clip”). Hypertension
also results from the balance between the angiotensin-
dependent system and volume-dependent mechanisms
based on salt and water retention, which otherwise would
be excreted by a normal contralateral kidney. The perfusion
pressure to the single ischemic kidney is maintained by the
high circulating volume and not the direct pressor effect of
angiotensin. Renal vein renin levels are near normal and
sufficient to maintain the elevated circulating volume, and
with it, normal glomerular filtration rate and renal func-
tion. In the presence of renin–angiotensin system inhibitors,
Fig. 28.28 Angiography demonstrating a transplant renal artery ste- however, the drive for salt and water retention is removed,
nosis (arrow) in a deceased donor right-sided kidney about 6 months
after transplantation. At time of transplantation, the atheromatous aor- causing reduction in perfusion to the solitary kidney and
tic patch with a tight orifice stenosis had been excised. Pressure mea- deterioration in kidney transplant function. The diagnosis
surements at time of angiography identified a mean systolic drop of 24 of TRAS is often made by observation of rapid deterioration
mmHg across the stenosis. in function with the introduction of renin–angiotensin sys-
tem inhibitors.
Dogs have been used to determine the minimal degree
of renal arterial stenosis needed to cause hypertension. In
an exceptional publication in 1992, Imanishi et al. demon-
strated clearly the relationship between the degree of ste-
nosis and the ability of the kidney to autoregulate its blood
flow.49 In anesthetized dogs, they constricted the left renal
artery concentrically using a radiolucent device and evalu-
ated the stenosis by cineangiography. With the kidney
either innervated or denervated, systemic blood pressure
began to increase when the stenosis was more than 70% of
the diameter of the renal artery. Renal blood flow decreased
when the stenosis was more than 75% of the diameter. In an
equally impressive canine study using magnetic resonance
imaging (MRI) and an implanted inflatable arterial cuff and
flow probe, Schoenberg et al. demonstrated that stenoses
of 30% to 80% gradually reduced downstream early sys-
tolic peak velocity, but only minimally affected peak mean
flow.50 At 50% stenosis, the mean pressure decrease across
the stenosis was recorded at about 10 mmHg, and at 80%,
28 mmHg. At a stenosis of 90%, mean flow was decreased
by greater than 50%.
An equivalent study in humans is unlikely. However,
extrapolation of Imanishi’s findings into the transplant set-
ting would explain why TRAS of 60% or more can be clinically
significant. Also, in the clinical setting, it should be possible to
learn more about the relationship between the magnitude of
the pressure gradient across a TRAS required to achieve bet-
ter blood pressure control after correction of that stenosis by
Fig. 28.29 Angiogram shows occlusion of an aneurysmal right com- angioplasty. Results could be correlated with spiral CT angi-
mon iliac artery proximal to the kidney transplant. At the time of angi- ography assessment of the cross-sectional area of the stenosis
ography and before proximal arterial bypass surgery, the patient had before angioplasty. Such a study has not been reported.
been receiving hemodialysis for 2 months. After bypass surgery, nor-
mal kidney transplant function returned.
IMAGING
follows the release of angiotensin by enzymatic processes Contrast angiography has long been the gold standard
from the circulating substrate, angiotensinogen, an octa- investigation of TRAS. The 1975 reporting of Hamburger’s
peptide with wide-ranging effects, including vasoconstric- 14-year experience from 1959 of TRAS concluded that
tion, renal sodium retention, aldosterone secretion, and angiography was so valuable that it should be performed
hypertrophy of myocardium and arteries.39 Blood pressure at routine intervals in all transplant recipients.41 Although
28 • Vascular and Lymphatic Complications after Kidney Transplantation 477

Fig. 28.30 Color Doppler ultrasound scanning of a kidney transplant demonstrating peak systolic velocity in the main renal artery of 4.10 m/s, diagnos-
tic of transplant renal artery stenosis.

angiography still might be the gold standard, ultrasound the stenosis. Hence, the preferred approach is to combine
examination has become the imaging modality of choice both, but the request may not be well received in a busy
for the routine assessment of transplant renal arteries. It general hospital ultrasound laboratory at short notice. Nev-
provides an instantaneous assessment of intrarenal vas- ertheless, the routine evaluation for TRAS at designated
culature and a global impression of transplant perfusion. time points after transplantation has merit.
Very few centers perform routine surveillance, preferring Having identified a TRAS by ultrasound examination,
to reserve ultrasound evaluation for symptomatic patients. the next decision is whether to proceed with vascular
There are two ultrasound approaches to the diagnosis contrast studies. The decision is not difficult if graft loss is
of TRAS. The extrarenal approach involves scanning the imminent. The risk of contrast nephropathy in an already
renal artery from the hilum to the anastomosis and beyond compromised kidney can be reduced by adequate hydration
to the proximal iliac artery. The PSV is measured along with normal saline before and after injection of contrast
the whole course of these vessels. A hemodynamically material.52 Multislice helical CT permits accurate assess-
significant stenosis has a PSV of greater than 2.5 m/s.51 ment of the site and degree of TRAS and provides valuable
The degree of stenosis and the site of maximum PSV can imaging for planning subsequent intervention. Advocates
be reported with a high degree of accuracy in the hands claim that helical CT requires less volume of iodinated vas-
of an experienced ultrasonographer (Fig. 28.30). Second- cular contrast medium than formal angiography and less
ary spectral findings of downstream turbulence and spec- toxicity with intravenous infusion of contrast. Protection of
tral broadening increase the confidence of the diagnosis of the transplanted kidney is recommended at all times when
TRAS. A prolonged acceleration time and a ratio of >3.0 vascular contrast medium is injected, regardless of renal
between renal and iliac artery PSV increase the diagnostic function and contrast volume. The alternative is to perform
accuracy. However, the technique is operator-dependent helical CT or MRI with gadolinium, a noniodinated contrast
and time-consuming. The unpredictable course of trans- medium. However, reports of nephrogenic systemic sclero-
plant renal artery makes it difficult to obtain the accurate sis with use of gadolinium are concerning, definition is less
angle of correction necessary for precise spectral quantifica- satisfactory because of its lower density, and therapeutic
tion. Distinguishing a focal stenosis from a tortuous renal intervention is impossible. Nevertheless, good screening
artery can be problematic, and reporting can err on the side images can be achieved, as shown in Fig. 28.15 of a torted
of false-positive findings. A careful diagram and direct com- intraperitoneal kidney. More recent studies using newer
munication with the ultrasonographer can increase the MRI scanners and protocols have demonstrated excellent
value of the report. results with noncontrast enhanced MRI.53
The intrarenal approach has the advantage of being less Conventional angiography remains the gold standard
operator-dependent, more reproducible, and easier to per- investigation because of the quality of definition, the abil-
form. It relies on the intrarenal downstream assessment of ity to measure pressure gradients across the stenosis, and
the effects of a TRAS. The early systolic peak is flattened and the potential to intervene at the same visit to the angiog-
delayed, the so-called parvus-tardus pattern. It is associated raphy suite. The contralateral femoral artery approach
with a low resistive index of 0.5 (Fig. 28.31). The intrarenal is used for kidneys transplanted to the internal iliac
technique is specific but not sensitive and can only diagnose artery by end-to-end technique. Otherwise, an ipsilateral
high-grade stenoses of greater than 75%. It cannot localize approach is used to complete an aortoiliac run using 20
478 Kidney Transplantation: Principles and Practice

Fig. 28.31 Color Doppler ultrasound scanning of a kidney transplant demonstrating arterial waveform of the intrarenal arcuate artery, showing the
features of parvus tardus, also indicative of transplant renal artery stenosis. Note the low resistance index of 0.54.

to 30 mL of iodinated vascular contrast medium, a step longer-term outcome studies are lacking. Intervention has
avoided by helical CT or MRI images. Selective runs are inherent risks, and it can be argued that, unless a signifi-
performed with oblique or other views as necessary using cant pressure drop exists across the TRAS, PTA should not
about 10 mL of contrast medium with each run. False- be undertaken. However, there is not an agreed mean pres-
negative examinations can occur if insufficient views are sure drop beyond which intervention is warranted. The
obtained. To this extent, multislice helical CT angiography canine studies of Schoenberg et al. suggest the figure should
with reconstructions has advantages over conventional be >10 mmHg.50
angiography. In the presence of a satisfactory radiologic result and
no improvement in clinical parameters, other underly-
ing causes of hypertension and graft pathology should
CONSERVATIVE TREATMENT
be sought. To this extent, PTA could be performed as an
If extrarenal Doppler ultrasound demonstrates TRAS in investigation of exclusion if the complication rates were
the order of 60%, kidney transplant function is satisfac- acceptable. As with other forms of interventional angiog-
tory, and the recipient is not hypertensive or has easily raphy, most of the complications relate to puncture site
treated hypertension, continued observation with repeat problems in the groin. Local clinician skill may dictate
ultrasound examination is a practical option. Neverthe- the wisdom of this line of management and the success of
less, there are no reports of the long-term safety of this interventional angiography is likely influenced by coop-
line of management, and the natural history of a 60% erative decision making by the radiologist and transplant
TRAS is unknown. Anecdotal evidence suggests this is surgeon. Increasingly, with newer premounted stents
probably safe for a kinked transplant renal artery, par- deployed by balloons, complications leading to graft
ticularly if there is no deterioration of kidney function.54 loss are unusual.55 Equally, it can be argued that, when
However, based on the previously described canine stud- thrombosis does complicate PTA, an experienced inter-
ies demonstrating reduction in kidney perfusion, one is ventional radiologist using urokinase and further stent-
more nervous about continued observation of a steno- ing usually benefits the recipient much faster and more
sis of >70% on ultrasound examination, irrespective of efficaciously than the surgeon who must find an emer-
acceptable transplant function. Such a stenosis would be gency operating room and rapidly undertake a difficult
more susceptible to occlusion in the presence of periods dissection and vascular reconstruction (Fig. 28.32).
of dehydration or cardiovascular instability, and inter- The restenosis rates are reported to be 10% to 60% and
vention should be considered. are probably influenced by cause of the stenosis, length of
follow-up, and use of stents.57,55,58 Indeed, data on long-
term effects of PTA on kidney allograft survival after PTA
ANGIOPLASTY AND STENTING
are scarce and understandably are based on observational
Percutaneous transluminal angioplasty (PTA) is recognized studies.59 For ethical reasons, a randomized trial might
as the initial treatment of choice for TRAS.55 A systematic only be feasible in patients with stable function and blood
review of 32 studies demonstrated technical success of pressure control, with the measure of success of the pro-
greater than 90%, with clinical success (judged by reduc- cedure based on graft survival. Such a trial is unlikely
tion of blood pressure and improvement in renal function) because it would need to be multicenter and take a decade
of 65.5% to 94% and patency of 42% to 100%.56 However, or more to complete.
28 • Vascular and Lymphatic Complications after Kidney Transplantation 479

A B

C D
Fig. 28.32 (A) Angiogram of a kidney transplant 3 months after transplantation. The mean arterial pressure gradient across the stenosis (arrow) in this
symptomatic patient was 16 mmHg. (B) Appearance of the renal artery 24 hours after percutaneous transluminal angioplasty. (C) Appearance of trans-
plant renal artery after insertion of a self-expanding stent. (D) After additional more distal stent (arrow). The transplant eventually failed 6 years later as
a result of chronic allograft nephropathy.

SURGICAL CORRECTION The surgical approach will depend on the cause. Easiest is
removal of a factor that might be causing the kink, together
Historically, correction of TRAS by surgery is seen as a difficult with release of fibrous tissue surrounding the kink. An
operation, with graft loss rates of 20%.60,41 Surgery is often there- example is demonstrated in Fig. 28.33 of a patient with large
fore viewed as the last resort or rescue therapy for cases unsuit- polycystic kidneys who presented 6 months after transplan-
able for PTA. Sometimes there is no alternative but surgery, as the tation with a hypertensive crisis and rapidly deteriorating
following examples demonstrate. The limiting factors for surgical graft function a day after manually moving several tons of
correction of TRAS are access to the artery, availability of arte- animal feed. The ipsilateral polycystic kidney was excised
rial inflow alternatives, and the resultant warm ischemia time. and the kinked transplant artery fully mobilized. Graft func-
A heparinized kidney allograft might tolerate warm ischemia of tion rapidly returned to baseline, as did antihypertensive
30 minutes or more because of the preexisting diminished blood medication requirement.
flow and the presence of a collateral flow, albeit with increasing Another option is excision of the stenosis with direct
risk of ATN and cortical necrosis as the minutes tick away. anastomosis to adjacent iliac artery, an example of which is
480 Kidney Transplantation: Principles and Practice

A B
Fig. 28.33 (A) Computed tomography scan (coronal view) demonstrating large polycystic kidneys, one of which is directly above a right-sided deceased
donor kidney transplant in the right iliac fossa. The hilum of the transplant is directed inferolaterally. (B) Digital subtraction angiogram of the transplant
renal artery demonstrating marked kinking that was able to be corrected by excision of the right polycystic kidney and mobilization of the artery.

agent to control hypertension. The TRAS was caused by


kinking secondary to distortion caused by the enlarging
aneurysm, off of which came four branches of the renal
artery.

Lymphocele
A lymphocele is a collection of lymph that accumulates in
a nonepithelialized cavity in the postoperative field. After
kidney transplantation, a lymphocele occurs after division
of recipient lymphatics accompanying the iliac vessels. The
incidence and frequency of detection have increased with
the routine surveillance of the kidney transplant by ultra-
sound and the introduction of mTOR inhibitors as part of
maintenance immunosuppression regimens.61 Lympho-
celes are usually innocuous and asymptomatic but can
Fig. 28.34 The transplant renal artery stenosis caused by kinking in Fig. equally cause dramatic presentations as a result of exter-
28.24 has been corrected surgically by ligating the renal artery, excis- nal pressure on the transplant and its adjacent structures
ing the kink, and reanastomosis (arrow) to the more proximal common or when complicated by infection involving the transplant
iliac artery. wound.

used to correct the TRAS illustrated in Fig. 28.24 and dem- INCIDENCE
onstrated in Fig. 28.34. Interposition grafting with long
saphenous vein or recipient internal iliac artery may be Considering the frequency with which iliac vessels are
necessary. Preserved ABO blood group compatible deceased exposed during routine vascular operations and the rarity
donor artery can also be used, with the United Network for of lymphatic complications, it came as a surprise to sur-
Organ Sharing guidelines recommending that deceased geons when the severity of lymphatic leakage after renal
donor artery grafts be used within 7 days of donation. Use of transplantation was first appreciated.62 Early reports after
synthetic graft material is usually avoided because of con- kidney transplantation, based on clinical presentation,
cerns about intimal hyperplasia. estimated the incidence to be around 2%. The advent of
An infrequently used option is autotransplantation of the ultrasound for routine graft surveillance caused the fig-
kidney after back table reconstruction of a complex arte- ure to be revised to about 50%, although accepting that
rial problem. Fig. 28.35 illustrates one such case in which most lymphatic collections remain subclinical and resolve
deceased donor vessels were used to replace an aneurysmal spontaneously.63
transplant renal artery. The original donor kidney came
from an 8-year-old child with brain death resulting from ETIOLOGY
rupture of an intracerebral arterial aneurysm. The recipi-
ent was 14 years old at the time of transplantation and An obvious suspected source of lymphatic leakage after
presented 6 years later with sudden onset of severe graft kidney transplantation would be the graft itself, and
dysfunction after introduction of an angiotensin II blocking occasionally this may be the case. A normal kidney has
28 • Vascular and Lymphatic Complications after Kidney Transplantation 481

A B

C D
Fig. 28.35 (A) Digital subtraction angiogram shows a 4-cm aneurysm of the transplant renal artery of a right-sided donor kidney 6 years after transplan-
tation into a 14-year-old boy. Note the kinking of the artery proximal to the aneurysm. The donor kidney came from an 8-year-old child who sustained
brain death after bleeding from a cerebral artery aneurysm. (B) Complete mobilization of the kidney transplant, before removal for ex situ reconstruc-
tion of the renal artery using cadaver donor vessels. (C) View of the inside of the thin-walled aneurysm showing four branches with takeoff from the
aneurysm of the transplanted renal artery. (D) Computed tomography angiogram with oblique view of arterial reconstruction (arrow) 2 weeks after
replacement of the transplant renal artery and vein with deceased donor iliac vessels and autotransplantation.

well-developed lymphatic drainage that is generally left a role in preventing the normal healing processes from seal-
unligated when transplanted. However, studies of injected ing the lymphatic vessels and is the more likely explanation
radiopaque dyes and radiolabeled substances showed that for the difference in the transplant setting. Macrophage
most lymphoceles originate from iliac vessel lymphatics function is adversely affected by steroids, and there is some
of the recipient (Fig. 28.36). It is estimated that 300 mL evidence that the incidence of lymphoceles has decreased
of lymph per day passes through the external iliac lymph since the introduction of low-dose steroid regimens. The
channels. Why the transplant kidney lymphatics contrib- more recent strong association of mTOR inhibitors with
ute so little, if any, to the presence of a lymphocele remains problematic lymphoceles is attributed to their powerful
unexplained. antifibroblastic activity, particularly in obese patients being
Traditional teaching suggests that meticulous ligation treated for rejection (body mass index > 30 kg/m2).61,68
of even the smallest lymphatic trunk with nonabsorbable Lymphoceles are more common in obese recipients, possi-
or slowly absorbed ligature material during mobilization bly because the lymph channels are more difficult to iden-
of the iliac vessels is crucial in the prevention of lympho- tify during dissection of the iliac vessels. Aggressive use of
celes (Fig. 28.37). However, the utilization of newer energy diuretics also has been implicated, but it could equally be
delivery devices such as the harmonic scalpel and Ligasure, they are more likely to be used in an edematous transplant
and even judicious use of diathermy, may be as effective recipient with greater lower-limb lymph flow.
and less time consuming.64–66 Based on their own experi-
ence, Sansalone et al. proposed that lymphoceles could be PRESENTATION
preventable if the vascular anastomoses were to the com-
mon iliac vessels, where fewer lymphatics and lymph nodes Small lymphoceles containing less than 100 mL of lymph are
are encountered during dissection.67 usually clinically silent and found on routine ultrasonogra-
The only differences between a routine retroperitoneal phy within days of transplantation and often resolve sponta-
vascular procedure on the iliac vessels and kidney trans- neously with time. Larger collections may become apparent
plantation are the physical presence of the kidney, an allo- clinically and usually do so at 1 week to 6 months after
immune response, and immunosuppression. Potentially, transplantation, with a peak incidence after hospital dis-
the kidney can create areas of dead space, particularly near charge toward the end of the first month.63 Most are situated
the upper or lower pole, and into which open lymph chan- adjacent to the lower pole of the kidney and posterolateral to
nels can, and do, drain. Immunosuppression may also have the transplant ureter, as illustrated in Fig. 28.38. Although
482 Kidney Transplantation: Principles and Practice

Fig. 28.38 Computed tomography scan (coronal view) demonstrat-


ing a lymphocele situated inferolateral to the kidney transplant and
stented transplanted ureter.

leg edema is often present. The timing of clinical presentation


of a lymphocele soon after removal of a transplant ureteric
stent 1 to 2 months after transplantation can be anticipated
with clinical concern of deteriorating renal function due to
compression of the ureter.
A lymphocutaneous fistula can develop between a lym-
phocele through an infected transplant wound. A less com-
Fig. 28.36 Lymphangiogram shows leakage of lymph from external mon presentation is as a DVT resulting from compression
iliac lymph channels causing a lymphocutaneous fistula through the of the external iliac vein (see Fig. 28.17). Bladder outlet
transplant wound.
obstruction has also been described.69

DIAGNOSIS
Ultrasound examination is the key to diagnosis. It can dis-
tinguish a lymphocele from hematoma collection on the
basis of characteristic homogeneity and distinctive shape
and position.23 Most lymphoceles are adjacent to, but
clearly separate from, the bladder. They can be multilocular
and multiple in number (Fig. 28.39) and, if in doubt, find-
ings can be confirmed by the passage of a urinary catheter
and repeat ultrasound. The examination also may show
hydronephrosis with obstruction of the ureter with dilated
calyces. Diagnosis can be further confirmed by ultrasound-
or CT-guided drainage, allowing biochemical and cytologic
analysis of the fluid. If emptied, resolution of the hydrone-
phrosis is seen. The use of vascular contrast medium helps
with localization of the ureter in the excretory phase.
Fig. 28.37 Division of external iliac lymphatics after ligation with
absorbable suture material.
TREATMENT
intralymphocele fluid pressure measurements have not been Unnecessary intervention of small and symptom-free collec-
reported, they must be considerable. The most common pre- tions may lead to infective complications. For symptomatic
sentation is sleep disturbance owing to urinary frequency as lymphoceles, a systematic review of retrospective studies
a result of compression of the bladder. They can be associated has suggested an algorithm that begins with ultrasound-
with a sense of fullness in the pelvis and ipsilateral painless or CT-guided drainage both to confirm the diagnosis and
28 • Vascular and Lymphatic Complications after Kidney Transplantation 483

Fig. 28.40 Computed tomography scan with axial view of lymphocele


with percutaneous drain (arrow) below the lower pole of the kidney
transplant. Note displacement to the right and compression of the
bladder.

a lower recurrence rate of 8% and a need for conversion


to open surgery of 12%. The least invasive surgical tech-
nique is a laparoscopic approach.70,73 A planning CT scan
Fig. 28.39 Computed tomography scan (coronal view) demonstrating is obtained to provide information about the position and
three lymphoceles (L) compressing bladder and kidney transplant in a presence of loculi. To facilitate localization, surgery is sched-
patient with polycystic kidneys.
uled when the lymphocele cavity is full, and not the day
after drainage. Alternatively, instillation of methylene blue
to provide initial treatment.70 The possible urgency of the down a correctly placed drain can visualize the lymphocele
situation is resolved by relief of urinary obstruction and res- when viewed laparoscopically.74 The surgeon ensures that
toration of kidney transplant function. About half the time, the recipient has an indwelling catheter and the bladder
simple aspiration will be curative. It may be repeated on is empty. The lymphocele is usually seen bulging into the
several occasions, although the likelihood of spontaneous peritoneal cavity. Localization with intraoperative ultra-
resolution becomes small after three aspirations followed by sound can be of assistance, particularly in obese patients
recurrence.63 Every aspiration brings a small risk of infec- and deeply situated lymphoceles. It is sometimes easy to
tion. Symptoms and signs can recur within days. confuse the swelling made by the extraperitoneal kidney
Prolonged external drainage through a percutaneously with that made by the lymphocele. The role of intraopera-
inserted catheter has been advocated by some authors and tive ultrasound in avoiding confusion is encouraged.70 An
is possible in an outpatient setting (Fig. 28.40). Injection of opening between the lymphocele and the peritoneal cavity
sclerosants has been described. Injection of povidone-iodine is made, taking care to avoid damage to any structures that
in association with external drainage has been claimed to may be running between the wall of the collection and the
be effective, with a low failure rate.71 The drawback of pro- peritoneum, particularly the ureter. The most difficult lym-
longed drainage is that it is up to 30 days before drainage phocele position to treat is one situated deep in the pelvis
ceases, during which time the risk of infection remains. Of and lateral to transplant artery and vein. These are more
further concern is the observation of acute renal failure as a safely treated by an open operative approach.
result of the direct nephrotoxic effect of povidone-iodine.72 To avoid recurrence, various authors have recommended
This treatment option is also poorly tolerated by the trans- maneuvers such as excision of a 5-cm disc of the wall of
plant recipient. the lymphocele, oversewing the edges, and mobilizing the
If simple percutaneous aspiration fails, a recent sys- omentum, which is then stitched down into the cavity.75
tematic review recommends a simple surgical procedure,
namely fenestration of the lymphocele, the principle of
LYMPHOCUTANEOUS FISTULA
which is to drain the potential 300 mL/day of lymph pro-
duction into the peritoneal cavity, where it is absorbed by The most challenging lymphatic complication to man-
the peritoneum.70 This operation of choice has been incor- age is a fistula draining about 300 mL/day from a divided
rectly called “marsupialization”; it is correctly described as external iliac chain lymphatic and a transplant wound
“fenestration.” It can be done either laparoscopically or by infected with a multiresistant organism. They present
open surgery through a lower midline abdominal incision invariably in the first weeks after transplantation, typically
and a transperitoneal approach to the lymphocele. Depend- in obese patients with diabetes and a surgical wound infec-
ing on its relationship to the kidney transplant, the previ- tion. Wound healing is slow and, as a result, skin sutures
ous wound can be reopened to achieve access. The same or staples tend to be left in place for longer than the norm
systematic review identified laparoscopic surgery as having and provide a portal of entry for bacteria. Prolonged use
484 Kidney Transplantation: Principles and Practice

A B C
Fig. 28.41 Left iliac fossa kidney transplant in an obese diabetic recipient. (A) Surgical wound closed with skin staples, 2 weeks after surgery. (B)
Arterial phase of computed tomography angiogram with axial view demonstrating a kidney transplant in the left iliac fossa surrounded by lymphatic
fluid in continuity with an infected open wound. (C) Transplant wound 4 months after transplantation.

of appropriate antibiotics and free drainage are usually References


attempted optimistically, along with all possible measures 1. Hsu THS, Su L-M, Ratner LE, Jarrett TW, Kavoussi LR. Demographics
that might improve wound healing, including reduction in of 353 laparoscopic renal donor and recipient pairs at the Johns Hop-
steroid immunosuppression. kins Medical Institutions. J Endourol 2003;17(6):393–6.
In these situations the transplant wound can be reopened, 2. Vacher-Coponat H, McDonald S, Clayton P, Loundou A, Allen RDM,
Chadban SJ. Inferior early posttransplant outcomes for recipients of
and a large peritoneal fenestration created. Sometimes it is right versus left deceased donor kidneys: an ANZDATA registry analy-
possible to see the offending leaking lymphatic channel at sis. Am J Transplant 2013;13(2):399–405.
the base of the lymphocele cavity, usually anterior to the 3. Özdemir-van Brunschot DMD, van Laarhoven CJHM, van der Jagt
external iliac artery. Suture ligation is invariably success- MFP, Hoitsma AJ, Warlé MC. Is the reluctance for the implantation of
ful. The risk of this procedure is introduction of antibiotic- right donor kidneys justified? World J Surg 2016;40(2):471–8.
4. Troncoso P, Guzman S, Domínguez J, Ortiz AM. Renal vein extension
resistant infection to the peritoneal cavity. The muscle wall using gonadal vein: a useful strategy for right kidney living donor har-
is closed with large absorbable interrupted sutures. If need vested using laparoscopy. Transplant Proc 2009;41(1):82–4.
be, it may be possible to use a vacuum dressing to facilitate 5. Parrott NR, Forsythe JL, Matthews JN, et al. Late perfusion: a simple
closure of subcutaneous tissues. remedy for renal allograft primary nonfunction. Transplantation
1990;49(5):913–5.
There are also lessons for the transplant surgeon from 6. Carter JT, Freise CE, McTaggart RA, et al. Laparoscopic procure-
this almost inevitable scenario, illustrated in Fig. 28.41. ment of kidneys with multiple renal arteries is associated with
Obesity at time of transplantation surgery has become increased ureteral complications in the recipient. Am J Transplant
more commonplace and dictates the need for preventive 2005;5(6):1312–8.
strategies to reduce the risk of lymphatic and coexist- 7. Lafranca JA, Bruggen M van, Kimenai HJAN, et al. Vascular multi-
plicity should not be a contra-indication for live kidney donation and
ing wound complications. Immunosuppression can be transplantation. PLoS ONE 2016;11(4):e0153460.
modified by minimizing steroid and lymphocyte-deplet- 8. Arrazola L, Sutherland DE, Sozen H, et al. May-Thurner syndrome in
ing antibodies and avoiding the use of mTOR inhibitors renal transplantation. Transplantation 2001;71(5):698–702.
for at least 1 month after the operation. The incision 9. Sidebottom RC, Parsikia A, Chang P-N, et al. No benefit when placing
drains after kidney transplant: a complex statistical analysis. Exp Clin
should follow Langer’s lines, but care should be taken not Transplant 2014;12(2):106–12.
to place it under a fold. If possible, the rectus abdominis 10. Derweesh IH, Ismail HR, Goldfarb DA, et al. Intraoperative placing of
muscle is preserved. Lymphatic division should be kept to drains decreases the incidence of lymphocele and deep vein thrombo-
a minimum with either careful ligation or use of appro- sis after renal transplantation. BJU Int 2008;101(11):1415–9.
priate energy devices. Suction drainage can potentially be 11. Kiberd B, Panek R, Clase CM, et al. The morbidity of prolonged
wound drainage after kidney transplantation. J Urol 1999;161(5):
avoided, although can still be of value overnight if placed 1467–9.
in the subcutaneous layer only. Finally, use of staples for 12. Ortiz J, Parsikia A, Horrow MM, Khanmoradi K, Campos S, Zaki R.
wound closure should be avoided, with preference given Risk factors for renal allograft compartment syndrome. Int Surg
to apposition of Scarpa’s fascia to eliminate wound dead 2014;99(6):851–6.
13. Damiano G, Maione C, Maffongelli A, et al. Renal allograft com-
space and use of an absorbable monofilament suture for partment syndrome: is it possible to prevent? Transplant Proc
subcuticular skin closure. 2016;48(2):340–3.
14. Kusyk T, Verran D, Stewart G, et al. Increased risk of hemorrhagic
complications in renal allograft recipients receiving systemic heparin
Conclusions early posttransplantation. Transplant Proc 2005;37(2):1026–8.
15. Mathis AS, Shah NK. Exaggerated response to heparin in a post-
operative renal transplant recipient with lupus anticoagulant under-
Vascular and lymphatic complications are relatively going plasmapheresis. Transplantation 2004;77(6):957–8.
common, but their incidence can be reduced by meticu- 16. Burke GW, Ciancio G, Figueiro J, et al. Hypercoagulable state asso-
lous attention to detail during removal of the kidney(s), ciated with kidney-pancreas transplantation: thromboelastogram-
directed anti-coagulation and implications for future therapy. Clin
back table inspection and preparation, preparation of the Transplant 2004;18(4):423–8.
implantation site in the recipient, and the technique of 17. Casserly LF, Dember LM. Thrombosis in end-stage renal disease.
implantation. Semin Dial 2003;16(3):245–56.
28 • Vascular and Lymphatic Complications after Kidney Transplantation 485

18. Irish A. Hypercoagulability in renal transplant recipients: identifying 43. Voiculescu A, Schmitz M, Hollenbeck M, et al. Management of arterial
patients at risk of renal allograft thrombosis and evaluating strategies stenosis affecting kidney graft perfusion: a single-centre study in 53
for prevention. Am J Cardiovasc Drugs 2004;4(3):139–49. patients. Am J Transplant 2005;5(7):1731–8.
19. Penny MJ, Nankivell BJ, Disney AP, Byth K, Chapman JR. Renal 44. Morris PJ, Yadav RV, Kincaid-Smith P, et al. Renal artery stenosis in
graft thrombosis: a survey of 134 consecutive cases. Transplantation renal transplantation. Med J Aust 1971;1(24):1255–7.
1994;58(5):565–9. 45. Wong W, Fynn SP, Higgins RM, et al. Transplant renal artery stenosis
20. Richardson AJ, Higgins RM, Jaskowski AJ, et al. Spontaneous rupture in 77 patients: does it have an immunological cause? Transplantation
of renal allografts: the importance of renal vein thrombosis in the 1996;61(2):215–9.
cyclosporin era. Br J Surg 1990;77(5):558–60. 46. Audard V, Matignon M, Hemery F, et al. Risk factors and long-term
21. Kujovich JL. Thrombophilia and thrombotic problems in renal trans- outcome of transplant renal artery stenosis in adult recipients after
plant patients. Transplantation 2004;77(7):959–64. treatment by percutaneous transluminal angioplasty. Am J Trans-
22. Wüthrich RP, Cicvara-Muzar S, Booy C, Maly FE. Heterozygosity for plant 2006;6(1):95–9.
the factor V Leiden (G1691A) mutation predisposes renal transplant 47. Goldblatt H, Lynch J, Hanzal RF, Summerville WW. Studies on
recipients to thrombotic complications and graft loss. Transplantation experimental hypertension: I. The production of persistent eleva-
2001;72(3):549–50. tion of systolic blood pressure by means of renal ischemia. J Exp Med
23. Akbar SA, Jafri SZH, Amendola MA, Madrazo BL, Salem R, Bis KG. 1934;59(3):347–79.
Complications of renal transplantation. Radiographics 2005;25(5): 48. Basso N, Terragno NA. History about the discovery of the renin-
1335–56. angiotensin system. Hypertension 2001;38(6):1246–9.
24. Robertson AJ, Nargund V, Gray DW, Morris PJ. Low dose aspirin as 49. Imanishi M, Akabane S, Takamiya M, et al. Critical degree of renal arte-
prophylaxis against renal-vein thrombosis in renal-transplant recipi- rial stenosis that causes hypertension in dogs. Angiology 1992;43(10):
ents. Nephrol Dial Transplant 2000;15(11):1865–8. 833–42.
25. Melamed ML, Kim HS, Jaar BG, Molmenti E, Atta MG, Samaniego MD. 50. Schoenberg SO, Bock M, Kallinowski F, Just A. Correlation of hemo-
Combined percutaneous mechanical and chemical thrombectomy for dynamic impact and morphologic degree of renal artery stenosis in a
renal vein thrombosis in kidney transplant recipients. Am J Trans- canine model. J Am Soc Nephrol 2000;11(12):2190–8.
plant 2005;5(3):621–6. 51. Thalhammer C, Aschwanden M, Mayr M, Staub D, Jaeger KA. Colour-
26. Kanchanabat B, Siddins M, Coates T, et al. Segmental infarction with coded duplex sonography after renal transplantation. Ultraschall Med
graft dysfunction: an emerging syndrome in renal transplantation? 2007;28(1):6–21: quiz 25.
Nephrol Dial Transplant 2002;17(1):123–8. 52. Pannu N, Manns B, Lee H, Tonelli M. Systematic review of the
27. Friedman GS, Meier-Kriesche HU, Kaplan B, et al. Hypercoagu- impact of N-acetylcysteine on contrast nephropathy. Kidney Int
lable states in renal transplant candidates: impact of anticoagula- 2004;65(4):1366–74.
tion upon incidence of renal allograft thrombosis. Transplantation 53. Zhang LJ, Peng J, Wen J, et al. Non-contrast-enhanced magnetic reso-
2001;72(6):1073–8. nance angiography: a reliable clinical tool for evaluating transplant
28. Cheungpasitporn W, Thongprayoon C, Mitema DG, et al. The effect of renal artery stenosis. Eur Radiol 2018;28(10):4195–204.
aspirin on kidney allograft outcomes; a short review to current stud- 54. Buturović-Ponikvar J. Renal transplant artery stenosis. Nephrol Dial
ies. J Nephropathol 2017;6(3):110–7. Transplant 2003;18(Suppl. 5):v74–7.
29. Lucewicz A, Isaacs A, Allen RDM, Lam VWT, Angelides S, Ple- 55. Kobayashi K, Censullo ML, Rossman LL, Kyriakides PN, Kahan BD,
ass HCC. Torsion of intraperitoneal kidney transplant. ANZ J Surg Cohen AM. Interventional radiologic management of renal transplant
2012;82(5):299–302. dysfunction: indications, limitations, and technical considerations.
30. Paulson WD, Moist L, Lok CE. Vascular access surveillance: an ongo- Radiographics 2007;27(4):1109–30.
ing controversy. Kidney Int 2012;81(2):132–42. 56. Ngo AT, Markar SR, De Lijster MS, Duncan N, Taube D, Hamady MS.
31. Allen RD, Michie CA, Murie JA, Morris PJ. Deep venous thrombo- A systematic review of outcomes following percutaneous translu-
sis after renal transplantation. Surg Gynecol Obstet 1987;164(2): minal angioplasty and stenting in the treatment of transplant renal
137–42. artery stenosis. Cardiovasc Intervent Radiol 2015;38(6):1573–88.
32. Verhave JC, Tagalakis V, Suissa S, Madore F, Hébert M-J, Cardinal H. 57. Chen L-X, De Mattos A, Bang H, et al. Angioplasty vs stent in the
The risk of thromboembolic events in kidney transplant patients. Kid- treatment of transplant renal artery stenosis. Clin Transplant
ney Int 2014;85(6):1454–60. 2018;32(4):e13217.
33. Ramirez PJ, Gohh RY, Kestin A, Monaco AP, Morrissey PE. Renal 58. Marini M, Fernandez-Rivera C, Cao I, et al. Treatment of transplant
allograft loss due to proximal extension of ileofemoral deep venous renal artery stenosis by percutaneous transluminal angioplasty and/
thrombosis. Clin Transplant 2002;16(4):310–3. or stenting: study in 63 patients in a single institution. Transplant
34. Albano L, Bretagne S, Mamzer-Bruneel M-F, et al. Evidence that Proc 2011;43(6):2205–7.
graft-site candidiasis after kidney transplantation is acquired dur- 59. Ghazanfar A, Tavakoli A, Augustine T, Pararajasingam R, Riad H,
ing organ recovery: a multicenter study in France. Clin Infect Dis Chalmers N. Management of transplant renal artery stenosis and its
2009;48(2):194–202. impact on long-term allograft survival: a single-centre experience.
35. Mai H, Champion L, Ouali N, et al. Candida albicans arteritis trans- Nephrol Dial Transplant 2011;26(1):336–43.
mitted by conservative liquid after renal transplantation: a report of 60. Bruno S, Remuzzi G, Ruggenenti P. Transplant renal artery stenosis.
four cases and review of the literature. Transplantation 2006;82(9): J Am Soc Nephrol 2004;15(1):134–41.
1163–7. 61. Pengel LHM, Liu LQ, Morris PJ. Do wound complications or lympho-
36. Leonardou P, Gioldasi S, Zavos G, Pappas P. Mycotic pseudoaneu- celes occur more often in solid organ transplant recipients on mTOR
rysms complicating renal transplantation: a case series and review of inhibitors? A systematic review of randomized controlled trials.
literature. J Med Case Rep 2012;6:59. Transpl Int 2011;24(12):1216–30.
37. Ram Reddy C, Ram R, Swarnalatha G, et al. ‘True’ mycotic aneurysm 62. Madura JA, Dunbar JD, Cerilli GJ. Perirenal lymphocele as a complica-
of the anastomotic site of the renal allograft artery. Exp Clin Trans- tion of renal homotransplantation. Surgery 1970;68(2):310–3.
plant 2012;10(4):398–402. 63. Pollak R, Veremis SA, Maddux MS, Mozes MF. The natural history of
38. Mackinnon B, Fraser E, Simpson K, Fox JG, Geddes C. Is it necessary and therapy for perirenal fluid collections following renal transplanta-
to stop antiplatelet agents before a native renal biopsy? Nephrol Dial tion. J Urol 1988;140(4):716–20.
Transplant 2008;23(11):3566–70. 64. Lucan CV, Jurchis I, Suciu M, Selicean SE, Buttice S. Modern lym-
39. Garovic VD, Textor SC. Renovascular hypertension and ischemic phatic dissection techniques for preventing post renal transplant lym-
nephropathy. Circulation 2005;112(9):1362–74. phocele. Clujul Med 2017;90(4):416–9.
40. Hurst FP, Abbott KC, Neff RT, et al. Incidence, predictors and out- 65. Nelson EW, Gross ME, Mone MC, et al. Does ultrasonic energy for sur-
comes of transplant renal artery stenosis after kidney transplantation: gical dissection reduce the incidence of renal transplant lymphocele?
analysis of USRDS. Am J Nephrol 2009;30(5):459–67. Transplant Proc 2011;43(10):3755–9.
41. Lacombe M. Arterial stenosis complicating renal allotransplantation 66. Simforoosh N, Tabibi A, Rad HM, Gholamrezaie HR. Comparison
in man: a study of 38 cases. Ann Surg 1975;181(3):283–8. between bipolar lymphatic vessels cautery and suture ligature in pre-
42. Gao J, Ng A, Shih G, et al. Intrarenal color duplex ultrasonography: a vention of postrenal transplant lymphocele formation: a randomized
window to vascular complications of renal transplants. J Ultrasound controlled trial. Exp Clin Transplant 2019;17(1):26–30. Available
Med 2007;26(10):1403–18. online at: https://doi.org/10.6002/ect.2017.0207.
486 Kidney Transplantation: Principles and Practice

67. Sansalone CV, Aseni P, Minetti E, et al. Is lymphocele in renal trans- 72. Manfro RC, Comerlato L, Berdichevski RH, et al. Nephrotoxic acute
plantation an avoidable complication? Am J Surg 2000;179(3):182– renal failure in a renal transplant patient with recurrent lym-
5. phocele treated with povidone-iodine irrigation. Am J Kidney Dis
68. Vítko S, Margreiter R, Weimar W, et al. Three-year efficacy and safety 2002;40(3):655–7.
results from a study of everolimus versus mycophenolate mofetil in de 73. Smyth GP, Beitz G, Eng MP, Gibbons N, Hickey DP, Little DM. Long-
novo renal transplant patients. Am J Transplant 2005;5(10):2521– term outcome of cadaveric renal transplant after treatment of symp-
30. tomatic lymphocele. J Urol 2006;176(3):1069–72.
69. Hwang EC, Kang TW, Koh YS, et al. Post-transplant lymphocele: an 74. Schilling M, Abendroth D, Kunz R. Treatment of lymphocele in renal
unusual cause of acute urinary retention mimicking urethral injury. transplant recipients by laparoscopic fenestration after transcutane-
Int J Urol 2006;13(4):468–70. ous staining. British J Surg 2005;82(2):246–8.
70. Lucewicz A, Wong G, Lam VWT, et al. Management of primary symp- 75. Bry J, Hull D, Bartus SA, Schweizer RT. Treatment of recurrent lym-
tomatic lymphocele after kidney transplantation: a systematic review. phoceles following renal transplantation: remarsupialization with
Transplantation 2011;92(6):663–73. omentorplasty. Transplantation 1990;49(2):477–80.
71. Rivera M, Marcén R, Burgos J, et al. Treatment of posttransplant lym-
phocele with povidone-iodine sclerosis: long-term follow-up. Nephron
1996;74(2):324–7.
29 Urologic Complications
After Kidney
Transplantation
ALICE CRANE and DANIEL A. SHOSKES

CHAPTER OUTLINE Ureteral Complications Immunosuppression and Donor Selection


Ureteral Leak Erectile Dysfunction and Testosterone
Ureteral Stenosis Replacement Therapy
Use of Prophylactic Ureteral Stents Urologic Malignancies
Urinary Calculi in Transplant Recipients
Urinary Retention

Urologic complications are inevitable in renal transplan- and lower pole of the kidney (the golden triangle) should
tation. Their incidence and effect on graft survival can, be preserved, as demonstrated in Fig. 29.1. When encoun-
however, be minimized. This chapter reviews the types of tered, lower-pole renal artery branches should be preserved
urologic complications that may occur, maneuvers to pre- or repaired, because they are commonly the end artery sup-
vent them, when to suspect and how to diagnose them, and plying the ureter. Ureteral complications are reported at a
treatments to maximize long-term outcome. Retrospective rate of 18% in grafts with duplicated ureters, although the
series quote an incidence of urologic complications of 1% total number of cases reported in the literature is small.5 In
to 15%, which has remained relatively stable.14 The first such cases, small upper-pole arteries should be preserved as
study to examine urologic outcomes after kidney trans- well.
plantation at a population level reported an 11.3% overall
rate of urologic complications in more than 9000 patients.4 URETERAL LEAK
Smaller studies with more restrictive definitions of urologic
complication commonly report rates of 4% to 5%. Ureteral leaks are reported in 1% to 3% of renal trans-
The incidence depends on many factors including dura- plants.6,7 The two most common causes are surgical error
tion of follow-up and how broadly urologic complications and ureteral ischemia with resultant necrosis. Technical
are defined. Some studies include hematuria, urinary tract errors include misplacement of ureteral sutures, unrec-
infection, and urinary retention; others are confined to ure- ognized ureteral transection or renal pelvis laceration,
teric strictures or leaks. This chapter discusses the following and insufficient ureteral length that places tension on the
urologic complications: ureteral leak, ureteral obstruction, anastomosis. Other rare causes of urine leaks include out-
urinary calculi, urinary retention, erectile dysfunction, and flow obstruction (as caused by a blocked Foley catheter or
urologic cancers. urinary retention) with disruption of the ureterovesical
anastomosis, acute ureteral obstruction with perforation
through a renal calyx, and extrusion of a ureteral stent.
Ureteral Complications Leaks resulting from technical errors often occur within the
first 24 hours, whereas leaks resulting from ischemia and
Ureteral leaks and obstructions are the result of technical necrosis usually occur within the first 14 days. However,
errors, ischemia, external compression, or intraluminal kidneys with delayed graft function may not have an evi-
blockage (e.g., ureteral stone). Unlike the native ureter, dent leak until suitable diuresis ensues.
which derives its blood supply from renal and pelvic sources, Preservation of periureteral tissue is essential, especially
the transplant ureter is supplied only by branches of the in living donors procured laparoscopically. Whereas the
anastomosed renal artery; therefore the distal segment of early experience with laparoscopic donor nephrectomy was
the ureter is the most ischemic. To overcome this, the renal associated with high rates of urinary leaks, improvements
allograft is placed into the pelvis, which minimizes the in surgical techniques have led to a decline in the rate to
length of the transplant ureter. During organ procurement, be almost as good as open donors.8 A ureter that appears
care should be taken to preserve the ureteral blood supply compromised at the time of surgery or fails to become pink
by removing the ureter along with a significant margin of and bleed after reperfusion should be cut as proximally as
periureteral tissue. Likewise, during the back table prepara- necessary to reach well-perfused tissue. This may necessi-
tion of the kidney, the perirenal fat bordered by the ureter tate an alternative technique to achieve urinary continuity,
487
488 Kidney Transplantation: Principles and Practice

c
a

A B

Fig. 29.1 (A) Cadaveric donor kidney after back table bench preparation. Note the preservation of the tissue between the lower pole of the kidney and
the ureter (circled), which typically contains the blood supply to the ureter and must be preserved. (B) The golden triangle (as outlined by a, b, and c).
Dissection in this area should be avoided during removal and preparation of the kidney for transplantation.

Urinary Extravasation

A B
Fig. 29.2 (A, B) Fusion single-photon emission computed tomography SPECT/CT images demonstrating a urine leak (arrows) outside confines of the
urinary bladder.

either by anastomosis to the ipsilateral native ureter or by fluid collection should be sent for creatinine measurement,
an extension technique of the bladder, such as a psoas hitch and the value should be compared with serum. Several
or Boari flap (see later). imaging studies may be diagnostic. A 99mTc-MAG-3 reno-
The clinical presentation of ureteral leaks can be obvi- gram may show tracer outside the anatomic confines of the
ous or subtle. The clearest clinical scenario is a patient with urinary tract but can be indeterminate, a cystogram may
excellent recovery of renal function whose urine output show the leak, particularly if it is located at the ureterovesi-
suddenly decreases or stops completely, associated with cal junction, and an ultrasound or computed tomography
drainage of fluid through the wound or increased drain out- (CT) scan may show a fluid collection, but not its source.
put. More often, however, confounding factors, including For more precise diagnosis and localization of a urinary
high urine output produced by the native kidneys, delayed leak, single-photon emission CT (SPECT)/CT fusion imag-
graft function that may limit urine output, and a preexisting ing can be performed (Fig. 29.2).
lymphocele or seroma make the presentation more subtle. Management of a ureteral leak can be endoscopic or
Urine leak should be part of the differential diagnosis in the operative. In a patient with an indwelling ureteral stent and
early posttransplant period whenever there is poor urine no Foley catheter, replacing the catheter often resolves the
output, a new fluid collection, new wound drainage, or leak, unless the entire distal ureter is necrotic. If this is effec-
delayed graft function. Any new fluid drainage or aspirated tive, the catheter should remain in place for at least 2 weeks,
29 • Urologic Complications After Kidney Transplantation 489

TABLE 29.1 Surgical Techniques to Bridge the Gap


between Transplant Ureter and Bladder
Technique Advantages Disadvantages
Direct reanasto- Simple, quick Limited by length
mosis of well-perfused
ureter
Psoas hitch Bladder reconfigured, no Must mobilize
loss of bladder volume bladder, limited
distance for small
bladder
Boari flap Can bridge large distance, Loss of bladder
well vascularized volume
Ureteroureteros- Simple, bladder not Ureter may be
tomy entered, well absent or atretic
vascularized
Pyelovesicostomy No need for donor or May be difficult to
recipient ureter reach, especially
if renal pelvis is
anterior (e.g., left
kidney in right
iliac fossa), free
reflux
Ileal ureter Can bridge large gap, Need for bowel
large lumen in event anastomosis, free
of stone formation reflux

followed by a confirmatory cystogram before catheter


removal. If no ureteral stent was placed, treatment modali- Fig. 29.3 A psoas hitch can provide 5 cm of additional length. The
ties include stenting or immediate surgical exploration. contralateral peritoneal bladder attachments are divided to bring the
Placement of a retrograde stent in a transplant ureter can bladder closer to the ureter. The bladder is incised transversely, and
be technically challenging because of the ectopic position the ureter is reimplanted with a submucosal tunnel superolateral to the
dome of the bladder. The bladder is then tacked down to the fascia of
of the ureteric orifice and lack of periureteral supports. Fur- the ipsilateral psoas muscle. A double-J ureteral stent is placed, and the
thermore, percutaneous nephrostomy with antegrade stent bladder is closed in two layers.
placement can be challenging because of the lack of hydro-
nephrosis with urine leaks. In the case of ureteral necrosis, it in addition to the psoas hitch, a Boari flap of bladder can
is preferable to explore and repair these leaks early. be created to bridge the gap for an anastomosis either to
Multiple surgical approaches exist to repair a ureteral the transplant ureter or to the transplant renal pelvis (Fig.
leak depending on the location and extent of ureteral 29.4).10
necrosis. Regardless of the technique, we prefer to use a Because a Boari flap reduces the total bladder volume,
three-way Foley catheter connected to irrigation that can its use may be inappropriate in the atrophied bladder of a
intermittently fill and empty the bladder to identify the leak previously anuric patient. The preferred technique here is
better. If the ureter is well perfused and a leak at the ure- to anastomose the ipsilateral ureter, if present, to either the
terovesical junction is the result of a technical problem with transplant ureter or the allograft renal pelvis (Fig. 29.5).
the anastomosis, the leak can be repaired by placing addi- Typically, the proximal native ureter can be tied off without
tional interrupted sutures. If the distal portion of the ureter the need for ipsilateral native nephrectomy.11 The advan-
is necrotic, it should be resected back to healthy tissue. If tages of this technique include excellent ureteral blood
the ureteral loss is minor, a simple reimplant of the trans- supply, a large segment of native ureter that can be reposi-
plant ureter is sufficient. Because a urine leak often results tioned without tension, and no compromise of bladder vol-
in local inflammation and tissue edema, all ureteral repairs ume. If native urothelium is unavailable, an ileal ureter can
or reimplantations should be performed over a stent. bridge the bladder and transplant renal pelvis (see Chapters
If a tension-free anastomosis cannot be achieved because 11 and 12).12
of limited ureteral length, several options are available
(Table 29.1). With a psoas hitch, the bladder is brought
closer to the ureter by mobilizing its attachments, in par-
URETERAL STENOSIS
ticular, by severing the contralateral obliterated umbilical Stenosis of the transplant ureter occurs in approximately
artery. The bladder is incised transversely and reconfig- 3% of transplant recipients.3 The obstruction can be
ured by closing the bladder incision in line with the ureter, extraluminal (compression from a lymphocele or spermatic
displacing the bladder toward the transplant ureter (Fig. cord), intrinsic (ureteral ischemia), or intraluminal (renal
29.3).9 The bladder, now elongated in the direction of the stone, fungal ball, sloughed renal papilla, or foreign body).
ureter, is fixed to the ipsilateral psoas muscle to allow a Ureteral stenosis may occur months or years after an oth-
tension-free ureteral reimplant. This technique, however, erwise successful transplant. Risk factors for late ureteral
may not provide sufficient length in a small bladder, as is stenosis include advanced donor age, delayed graft func-
found in long-standing oliguric patients. Alternatively, or tion, and kidneys with more than two arteries.13 Human
490 Kidney Transplantation: Principles and Practice

new-onset hydronephrosis should also be screened for uri-


nary retention by checking a postvoid residual volume.
After discovering hydronephrosis, two further confir-
matory tests include a diuretic 99mTc-MAG-3 renogram
or a percutaneous antegrade nephrostogram (Fig. 29.6).
A diuretic renogram suggests obstruction if the clearance
curve shows pelvicaliceal hold-up, especially after diuretic
administration.6 False-negative results can occur in
patients with poor renal function, and false-positive results
can occur with bladder outlet obstruction or vesicoureteral
reflux. Antegrade pyelography is the preferred test when
obstruction is strongly suspected. A hydronephrotic trans-
plant kidney is easily accessible with a small spinal needle
to inject contrast medium.16 If obstruction is confirmed, the
needle can be converted to a nephrostomy tube over a wire,
and antegrade stenting can be performed immediately or
after the renal function and ureteral edema improve.
Endoscopic management of transplant ureteral strictures
is preferable to surgery, which can be difficult when done
months or years after the original transplant surgery. The
stricture can be accessed in an antegrade or retrograde
fashion. If a stent does not pass easily over a wire, the stric-
ture can be balloon-dilated or an endoureterotomy can
be performed with a laser or a cutting balloon. The endo-
scopic approach is successful in about 50% to 65% of cases;
however, recurrent strictures may result from inadequate
primary therapy or failure because of extensive ischemia.
Although patients with recurrent strictures can be man-
aged with long-term indwelling ureteral stents, recurrent
strictures are best treated with an open approach. When
the site of obstruction is identified and the diseased segment
of the ureter is excised, the operative approach is similar to
that for a ureteral leak (e.g., psoas hitch, Boari flap, uretero-
pyelostomy, pyelocystostomy, or ileal ureter). Successful
treatment of transplant ureteral stenosis is critical, because
long-term graft survival is improved.13
Fig. 29.4 A Boari flap can provide 10 to 15 cm of additional length. The
bladder is mobilized as in the psoas hitch. A full-thickness U-shaped Use of Prophylactic Ureteral
bladder flap is created. The length of the flap varies depending on the
length of the gap that needs to be bridged. To assure adequate blood Stents
supply, the base of the flap should be at least 2 cm greater than the
apex, and the width of the flap should be three to four times the diam- The routine use of ureteral stents (Fig. 29.7) at the time of
eter of the ureter. The ureter is then anastomosed with a submucosal kidney transplantation is controversial. Table 29.2 lists the
tunnel or in an end-to-end fashion with the flap. A double-J ureteral
stent is placed, and the bladder flap is closed in two layers. For a ten- pros and cons. As reported in some series, stents can reduce
sion-free anastomosis, the tip of the flap can be secured to the ipsilat- the incidence of ureteral leaks and early ureteral stenosis,15
eral psoas muscle. while making the early management of leaks easier. Other
reports, including prospective randomized trials, demon-
polyomavirus (BK virus) can also lead to ureteral stenosis strated no utility for prophylactic stenting.17 Mangus and
due to ureteritis.14 Although ureteral stenting at the time of Haag performed a meta-analysis of 49 published studies,
transplant reduces the incidence of early stenosis, it has no including randomized controlled trials (RCTs), and demon-
effect on the rate of late ureteral stenosis.15 strated a significant reduction in ureteric complications with
The clinical presentation of ureteral stenosis can vary stents (from 9% to 1.5%; P < 0.0001).18 A recent Cochrane
according to its location, degree, and speed of onset. Most review that included seven RCTs and quasi-RCTs demon-
commonly, ureteral stenosis is gradual and asymptomatic, strated a decrease in urologic complications with prophylac-
with an unexplained increase in serum creatinine and the tic stenting versus no stenting and no increase in infection
discovery of hydronephrosis on ultrasound. Pain over the risk as long as prophylactic antibiotics were given. However,
allograft is rare, unless the obstruction is sudden and high- the data also supported no advantage to routine stenting
grade. Hydronephrosis is not always synonymous with over selective stenting in experienced hands.19 Although the
obstruction, however. Dilation of the renal pelvis and calices optimal duration of prophylactic stenting has not been deter-
can occur without obstruction in the setting of prior obstruc- mined, for most centers it is typically for 2 to 6 weeks. If a
tion (e.g., long-standing ureteropelvic junction obstruction ureteral stent is placed, the patients chart should be flagged
in the donor), reflux, or loss of renal cortex parenchyma, and the patient should be told to return for stent removal.
such as in chronic allograft nephropathy. Patients with Especially in busy programs, the danger exists that a stent
29 • Urologic Complications After Kidney Transplantation 491

Vein

Anastomosis

Healthy ureter

Ischemic ureter Native ureter


A B
Fig. 29.5 Repair of transplant ureteral necrosis by ureteroureterostomy. (A) Distal ureteral necrosis. Note the distal ureter, proximal ureter, and
accumulation of urine in the wound. (B) After repair. The native ureter was transected and rotated to the proximal transplant ureter. The anastomosis
was made end-to-end over a double-J stent using 5-0 PDS suture. The proximal native ureter was tied off without native nephrectomy.

TABLE 29.2 Advantages and Disadvantages of Routine


Prophylactic Ureteral Stenting in Renal Transplants
Advantages Disadvantages
Reduction in ureteric complica- 95% of patients have unnecessary
tions stent
Urine leak easier to manage Increased risk of urinary tract
infection
Cost effective Risk of stent migration or stone
encrustation
No evidence for patient or graft
survival benefit
Patient discomfort from bladder
spasm

may be forgotten and remain in place for several months or


years until the patient presents with a retained, calcified stent
(Fig. 29.8).

Urinary Calculi in Transplant


Fig. 29.6 Antegrade nephrostogram in a transplanted kidney showing Recipients
an obstructed distal ureter.
The incidence of nephrolithiasis in renal transplant recipi-
ents ranges from 1% to 5%.20,21 In the US only 1 in 1000
transplanted patients had a hospital admission for stones,
with the strongest risk factors being female sex and prior
history of stone disease.22 As more centers transplant kid-
neys from living donors with known asymptomatic renal
stones, this incidence may increase. Other causes of stones
include the use of nonabsorbable sutures in the urinary
tract, foreign bodies (e.g., a retained stent), persistent uri-
nary tract infection, ileal conduit diversion, and incom-
plete bladder emptying. Metabolic evaluation of transplant
recipients who form stones most commonly reveals hypoci-
traturia, hyperparathyroidism, hypophosphatemia, and
hypercalcemia. Hypocitraturia has been linked with the use
Fig. 29.7 Double-J ureteral stent. of calcineurin inhibitors.23
492 Kidney Transplantation: Principles and Practice

urethral stricture, benign prostatic hyperplasia, bladder


calculus, or bladder neck contracture. Anuric men with
benign prostatic hyperplasia should not be offered surgical
treatment before transplantation because transurethral
prostatic surgery in a dry urethra has a high incidence of
stricture formation. After transplant, therapy for men with
significant bladder outlet obstruction from benign pros-
tatic hyperplasia should begin with an alpha-blocker alone
or in combination with a 5-reductase inhibitor. Men in
retention should start intermittent self-catheterization and
delay definitive endoscopic prostatic surgery for at least 3
months posttransplant. Although transurethral resection
of the prostate can be done in the immediate posttransplan-
tation period, significant morbidity24 and mortality3 have
been reported.
Fig. 29.8 Plain radiograph of an encrusted, retained stent. The patient
had stent placement at the time of transplant, but moved to another
country before the stent was removed. The patient presented to our
Immunosuppression and Donor
institution 2 years later with stones in the kidney and bladder. Selection
Because the transplanted kidney is denervated, the clini- Calcineurin inhibitors (CNIs) have been the cornerstone of
cal presentation of transplant nephrolithiasis is varied and maintenance immunosuppression since the 1990s. Com-
includes pain over the graft site, gross hematuria, and bination therapy with tacrolimus, mycophenolate mofetil
reduced or absent urine output. Asymptomatic stones may (MMF), and steroids is most commonly used in the US,
be discovered during routine imaging or as part of a workup and this use has remained relatively stable since 2009.25
of elevated creatinine, and stone number and location are However, with the increasing focus on avoiding the neph-
best delineated by noncontrast CT scan. Urine culture rotoxicity of CNI-based regimens and the introduction of
should be collected from all patients with kidney stones. If costimulation blockade with belatacept, trends in stan-
a patient presents with anuria, emergent intervention with dard immunosuppression may change, which may affect
percutaneous nephrostomy is indicated. Bladder calculi urologic complications. As an example, BK nephropathy
also should be evaluated by cystoscopy to prevent outflow is thought to be increased in tacrolimus-based regimens,
obstruction. which, in turn, can cause ureteral stricture, as discussed
The treatment of stones in a transplanted kidney is simi- earlier. Additionally, there have been case reports of BK
lar to that of native kidneys, with the exception that percu- virus-associated urothelial carcinoma (UCC).26 Outcomes
taneous approaches are easier because of the location of the must be carefully tracked as changes to standard regimens
kidney in the pelvis. Most often, small stones pass spontane- are made to elucidate other unexpected effects on urologic
ously without intervention. Larger obstructing stones can complications.
be treated with extracorporeal shock wave lithotripsy, ante- Over the years there has been an escalation in the use
grade or retrograde ureteroscopic stone extraction (with of deceased donor organs and an increased willingness to
laser lithotripsy if necessary), or, rarely, open surgery.20 use increased risk organs in the face of organ shortage.
When a stone is identified in a living donor, the kidney with Based on Organ Procurement and Transplantation Net-
the stone should be selected for transplantation, and the work (OPTN) data as of September 2017, there has been
stone can be successfully removed by back table ureteros- an increase in deceased versus living donor transplants
copy or ultrasound-guided nephrolithotomy. with deceased donors making up 56% of the donor pool in
2011 and 62% in 2016. The rate of donation after cardiac
death (DCD) donors specifically has risen from approxi-
Urinary Retention mately 14% to 18% over the same time period. Rahnemai-
Azar et al., in a large retrospective single-center series in
After renal transplantation, urinary retention may be New York, found no correlation between DCD donation
caused by bladder outlet obstruction or an acontractile and risk of urologic complication, finding only renal artery
detrusor muscle. In previously anuric patients, these prob- multiplicity as an independent risk factor.2 The European
lems may not be identified until after the Foley catheter is experience has been similar with Szabo-Pap et al., in a
removed. Patients with a flaccid bladder usually have a single-center review of surgical complications in Hun-
prior history of voiding dysfunction or a neurogenic blad- gary, reporting no change in the rate of urologic complica-
der. When bladder pathology is suspected, urodynamics tions before and after 2013, when their expanded criteria
with pressure flow studies and cystoscopy should be per- donors increased as a result of practice pattern changes.27
formed to characterize the bladder and bladder neck, and However, careful attention must be paid, because the lim-
the patient should be instructed to perform intermittent its of donor acceptability are pushed by increasing recipi-
self-catheterization, which is safe and effective in transplant ent demand. This is particularly important with any donor
recipients. parameter that threatens vascularity and perfusion of the
Bladder outlet obstruction after transplantation is graft because this is the most consistent risk factor for the
almost exclusively seen in men and may be a result of development of ureteral leaks and strictures.
29 • Urologic Complications After Kidney Transplantation 493

Erectile Dysfunction and


Testosterone Replacement
Therapy
With an aging transplant population, erectile dysfunction
(ED) is a prevalent and increasingly identified concern,
occurring in up to 53% of male transplant recipients.28
Factors contributing to ED are often the same factors
responsible for renal failure, including diabetes, vasculop-
athy, and hypertension (owing to its medical treatment).
Dialysis patients often have an elevated serum prolactin
level, which decreases testosterone levels, resulting in
low libido and ED. This may partly account for the 20% of
patients whose ED improves after transplant.28 Although
the internal iliac artery is less commonly used for the renal
artery anastomosis, it should be avoided in men receiving a
second transplant, because the risk of developing vasculo-
pathic impotence can be as high as 25% in this situation.29
Given the multifactorial nature of ED in this population,
there is limited value in an extensive workup beyond mea-
suring testosterone and prolactin levels. Fig. 29.9 Plain radiograph of a patient with an artificial urinary sphinc-
For male patients with symptoms of hypogonadism, tes- ter. Note the position of the fluid reservoir containing radiopaque
tosterone replacement therapy should be considered. Low contrast in the lower right pelvis, where it could be damaged during
testosterone has been shown to confer increased mortality transplant recipient dissection. A traditional three-component inflat-
able penile prosthesis would have a similar reservoir but would be
in dialysis and transplant patients in addition to profound filled with saline and be radiolucent. Pretransplant imaging with a non-
effects on patient quality of life. Although the hypogonadal contrast computed tomography (CT) scan can confirm the location and
state of some patients will resolve with transplantation direct the incision to the contralateral side.
and normalization of kidney function, 25% of transplant
patients will have persistent symptomatic hypogonadism the prosthesis, however, is higher in transplant patients
posttransplantation. There has been general hesitation to who receive a traditional three-piece model with a retro-
use testosterone replacement therapy (TRT) in this popula- peritoneal fluid reservoir, owing to the need for multiple
tion in part because of concern over testosterone (T)-medi- retroperitoneal surgeries. A two-piece prosthesis should be
ated stimulation of the renin-angiotensin pathway. This considered in these patients because it lacks a fluid reser-
fear stems from animal studies in which negative effects of voir.36 Patients being evaluated for transplant with a known
blood pressure and kidney function were seen in response penile prosthesis or artificial urinary sphincter should have
to TRT. This has not been documented in humans. In addi- preoperative imaging performed (Fig. 29.9) to confirm the
tion to the lack of evidence of negative consequences on the existence and laterality of a fluid reservoir, and the contra-
cardiovascular health of the human end-stage renal disease lateral side should be chosen for allograft placement.
(ESRD) and transplant population the potential benefits of
TRT on bone health and hematocrit are of particular note
for this fracture- and anemia-prone population. Further- Urologic Malignancies
more, it has been suggested that TRT for men with symp-
tomatic hypogonadism and chronic kidney disease may The most common malignancies postrenal transplant are
experience fewer cardiovascular effects.30 It is unknown skin cancers and posttransplant lymphoproliferative dis-
whether these beneficial cardiovascular effects extend to ease (PTLD). However, the rate of urologic malignancy is
the transplant recipient. Testosterone replacement therapy also increased and can occur as de novo tumors, recur-
has been shown to be safe and effective in this population, rences, or unrecognized transmitted donor malignancies
although studies remain small and underpowered.31,32 Fur- (see Chapter 35). Immunosuppression, infection with onco-
thermore, low serum testosterone is correlated with higher genic viruses, and loss of T-suppressor function are known
mortality in ESRD patients on dialysis and at the time of risk factors for malignant transformation.
transplant.33,34 The largest increase is seen with renal cell carcinoma
Regarding the treatment of ED specifically, transplant (RCC), which more commonly arises in the native kidneys,
patients tolerate phosphodiesterase-5 inhibitor therapy in part because of the increased risk conferred by acquired
well, with sildenafil demonstrating good efficacy and no renal cystic disease on prolonged dialysis. Radical nephrec-
effect on calcineurin levels.35 For patients who fail oral tomy is performed for tumors of the native kidneys, and
therapy, intracorporeal injection of agents such as prosta- nephron-sparing surgery should be attempted for masses
glandin E1 or papaverine is effective. Alternatively, inflat- in the allograft. Although the risk of RCC is increased most
able penile prostheses are safe and effective in transplant significantly, with up to 15-fold risk over non-ESRD/trans-
patients, and the risk of prosthesis infection and erosion is planted individuals, prostate cancer remains the most com-
no higher despite the immunocompromised state and poor mon malignancy, although studies conflict on whether
tissue healing. Risk of device malfunction and damage to transplant patients have a higher incidence of prostate
494 Kidney Transplantation: Principles and Practice

cancer than the general population.37,38 Age-appropriate malignancies, specifically RCC and bladder cancer, remains
screening with a digital rectal examination and prostate- a significant unknown in transplant patients. It is mecha-
specific antigen should be performed annually. As with nistically possible that immune checkpoint inhibitors may
other cancers in the immunosuppressed patient, the data place the graft at increased risk of rejection. There have
suggest that prostate cancer tends to be more aggressive and been scattered reports in the skin cancer literature of rapid
has poorer outcomes in transplant patients, although this is graft failure with programmed cell death protein and ligand
less clearly defined than in other genitourinary malignan- 1 (PD-1 and PD-L1) inhibitors.47 However, the use of ipi-
cies.39 Although reported prostate cancer series are small limumab, a cytotoxic T-lymphocyte-associated protein 4
in the transplanted population, this may be expected to rise (CTLA-4) inhibitor did not have a dramatic effect on graft
with increases in recipient longevity and transplantation of function. This is logical as belatacept itself is essentially
older individuals. a CTLA-4 inhibitor, although belatacept does confer an
In addition to the increased prevalence of malignancy increased risk of acute rejection in some patients. As our
after transplant, there is added complexity to the cancer knowledge evolves, any immunotherapy must be used with
treatment strategy in the setting of a prior kidney trans- utmost care and proper patient counseling, including the
plant. In the case of prostate cancer active surveillance is possibility of graft failure, before initiation of therapy.
avoided, with fewer than five patients reported in the litera-
ture as on watchful waiting or active surveillance.37 Most References
patients undergo definitive therapy with radiation or sur- 1. Gil-Sousa D, Oliveira-Reis D, Teves F, et al. Ureteral stenosis after renal
gery despite a lack of evidence that active surveillance is transplantation: a single-center 10-year experience. Transplant Proc
2017;49:777–82.
harmful in these patients. External beam radiation in this 2. Rahnemai-Azar AA, Gilchrist BF, Kayler LK, et al. Independent risk
population is difficult given the graft is within the radiation factors for early urologic complications after kidney transplantation.
field, in addition to ureteral complications and increased Clin Transplant 2015;29:403–8.
gastrointestinal side effects.40 This has been recently chal- 3. Shoskes DA, Hanbury D, Cranston D, et al. Urological complications
lenged but on the experience of only one patient with a in 1000 consecutive renal transplant recipients. J Urol 1995;153:
18–21.
functioning graft.41 Caution should be applied to renal 4. Sui W, Lipsky MJ, Matulay JT, et al. Timing and predictors of early
transplant recipients in applying external beam radiation. urologic and infectious complications after renal transplant: an
If radiation is desired, brachytherapy appears to be an effec- analysis of a New York statewide database. Exp Clin Transplant
tive choice without the side effects of external radiation 2018;16(6):665–70. Available online at: https://doi.org/10.6002/
ect.2016.0357.
although experience is limited.42 5. Alberts VP, Minnee KAMI, van Donselaar-van der Pant FJ, et al.
Surgical considerations in transplanted oncology Duplicated ureters and renal transplantation: a case-control study
patients include prior surgery, comorbidities, immunosup- and review of the literature. Transplant Proc 2013;45:3239–44.
pressed state, and anatomic considerations. The lymph 6. Nankivell BJ, Cohn DA, Spicer ST, et al. Diagnosis of kidney trans-
node dissection on the graft side is often omitted because of plant obstruction using Mag3 diuretic renography. Clin Transplant
2001;15:11–8.
fibrosis. The largest series to date in transplanted patients 7. Streeter EH, Little DM, Cranston DW, et al. The urological compli-
(20/30 were kidney recipients) undergoing open radical cations of renal transplantation: a series of 1535 patients. BJU Int
prostatectomy demonstrated no increase in final pathologic 2002;90:627–34.
staging or immediate postoperative complications although 8. Philosophe B, Kuo PC, Schweitzer EJ, et al. Laparoscopic versus open
donor nephrectomy: comparing ureteral complications in the recipi-
the incontinence rate was increased.43 With robotic sur- ents and improving the laparoscopic technique. Transplantation
gery, adjustments to port placement should be considered 1999;68:497–502.
in addition to early posterior approach and judicious mobi- 9. Matthews R, Marshall FF. Versatility of the adult psoas hitch ureteral
lization of the bladder. reimplantation. J Urol 1997;158:2078–82.
Just as prior transplant can alter the approach of onco- 10. del Pizzo JJ, Jacobs SC, Bartlett ST, et al. The use of bladder for total
transplant ureteral reconstruction. J Urol 1998;159:750–2; discus-
logic surgery, so too can prior oncologic surgery alter the sion 752-3.
kidney transplant, particularly in the case of urinary diver- 11. Gallentine ML, Wright Jr FH. Ligation of the native ureter in renal
sion. Renal transplantation into patients with a prior con- transplantation. J Urol 2002;167:29–30.
duit is associated with a high morbidity and incidence of 12. Shokeir AA, Shamaa MA, Bakr MA, et al. Salvage of difficult trans-
plant urinary fistulae by ileal substitution of the ureter. Scand J Urol
urosepsis and lithiasis.44 Orientation of the kidney may Nephrol 1993;27:537–40.
need to be altered to avoid ureteral kinking. Although this 13. Karam G, Hetet JF, Maillet F, et al. Late ureteral stenosis following renal
situation is more commonly seen in patients with congeni- transplantation: risk factors and impact on patient and graft survival.
tal abnormalities, the incidence of oncology patients with Am J Transplant 2006;6:352–6.
urinary diversion who need a subsequent transplant is 14. Coleman DV, Mackenzie EF, Gardner SD, et al. Human polyomavirus
(BK) infection and ureteric stenosis in renal allograft recipients. J Clin
likely to increase. Pathol 1978;31:338–47.
Regarding the use of nephrotoxic platinum-based che- 15. Sansalone CV, Maione G, Aseni P, et al. Advantages of short-time
motherapy, it appears safe in this population if they have ureteric stenting for prevention of urological complications in kidney
the renal function to tolerate it, although long-term data transplantation: an 18-year experience. Transplant Proc 2005;37:
2511–5.
are lacking.45 The use of Bacillus Calmette-Guerin (BCG) in 16. Bach D, Grutzner G, Kniemeyer HW, et al. Diagnostic value of ante-
immunosuppressed patients for lower tract UCC, although grade pyelography in renal transplants: a comparison of imaging
contraindicated per package insert, has also been safely modalities. Transplant Proc 1993;25:2619.
done in a small series; this will require stronger studies 17. Dominguez J, Clase CM, Mahalati K, et al. Is routine ureteric stenting
and longer follow-up.46 In addition to traditional therapy, needed in kidney transplantation? A randomized trial. Transplanta-
tion 2000;70:597–601.
the increased prevalence of immunotherapy for urologic
29 • Urologic Complications After Kidney Transplantation 495

18. Mangus RS, Haag BW. Stented versus nonstented extravesical ure- 34. Shoskes DA, Kerr H, Askar M, et al. Low testosterone at time of trans-
teroneocystostomy in renal transplantation: a metaanalysis. Am J plantation is independently associated with poor patient and graft
Transplant 2004;4:1889–96. survival in male renal transplant recipients. J Urol 2014;192(4):
19. Wilson CH, Rix DA, Manas DM. Routine intraoperative ureteric stent- 1168–71.
ing for kidney transplant recipients. Cochrane Database Syst Rev 35. Zhang Y, Guan DL, Ou TW, et al. Sildenafil citrate treatment for
2013;17(6):CD004925. erectile dysfunction after kidney transplantation. Transplant Proc
20. Challacombe B, Dasgupta P, Tiptaft R, et al. Multimodal management 2005;37:2100–3.
of urolithiasis in renal transplantation. BJU Int 2005;96:385–9. 36. Cuellar DC, Sklar GN. Penile prosthesis in the organ transplant recipi-
21. Khositseth S, Gillingham KJ, Cook ME, et al. Urolithiasis after kidney ent. Urology 2001;57:138–41.
transplantation in pediatric recipients: a single center report. Trans- 37. Marra G, Dalmasso E, Angello M, et al. Prostate cancer treat-
plantation 2004;78:1319–23. ment in renal transplant recipients: a systematic review. BJU Int
22. Abbott KC, Schenkman N, Swanson SJ, et al. Hospitalized nephrolithi- 2018;121(3):327–44. Available online at: https://doi.org/10.1111/
asis after renal transplantation in the United States. Am J Transplant bju.14018.
2003;3:465–70. 38. Melchior S, Franzaring L, Shardan A, et al. Urological de novo malig-
23. Stapenhorst L, Sassen R, Beck B, et al. Hypocitraturia as a risk fac- nancy after kidney transplantation: a case for the urologist. J Urol
tor for nephrocalcinosis after kidney transplantation. Pediatr Nephrol 2011;185(2):428–32.
2005;20:652–6. 39. Miao Y, Everly JJ, Gross TG, et al. De novo cancers arising in organ
24. Reinberg Y, Manivel JC, Sidi AA, et al. Transurethral resection of transplant recipients are associated with adverse outcomes compared
prostate immediately after renal transplantation. Urology 1992;39: with the general population. Transplantation 2009;87:1347–59.
319–21. 40. Mouzin M, Bachaud JM, Kamar N, et al. Three-dimensional conformal
25. Lim MA, Kohli J, Bloom RD. Immunosuppression for kidney trans- radiotherapy for localized prostate cancer in kidney transplant recipi-
plantation: where are we now and where are we going? Transplant ents. Transplantation 2004;78:1496–500.
Rev (Orlando) 2017;31(1):10–7. 41. Lizuka J, Hashimoto Y, Hashimoto Y, et al. Efficacy and feasibility of
26. Oikawa M, Hatakeyama S, Fukita T, et al. BK virus-associated urothe- intensity-modulated radiation therapy for prostate cancer in renal
lial carcinoma of a ureter graft in a renal transplant recipient: a case transplant recipients. Transplant Proc 2016;48(3):914–7.
report. Transplant Proc 2014;46(2):616–9. 42. Beydound N, Bucci J, Malouf D. Iodine-125 prostate seed brachy-
27. Szabo-Pap M, Zadori G, Fedor L, et al. Surgical complications follow- therapy in renal transplant recipients: an analysis of oncological
ing kidney transplantations: a single-center study in hungary. Trans- outcomes and toxicity profile. J Contemp Brachytherapy 2014;6(1):
plant Proc 2016;28:2548–51. 15–20.
28. Russo D, Musone D, Alteri V, et al. Erectile dysfunction in kidney trans- 43. Beyer B, Mandel P, Michl U, et al. Oncological, functional and periop-
planted patients: efficacy of sildenafil. J Nephrol 2004;17:291–5. erative outcomes in transplant patients after radical prostatectomy.
29. Taylor RM. Impotence and the use of the internal iliac artery in renal Worl J Urol 2016;34:1101–5.
transplantation: a survey of surgeons attitudes in the United Kingdom 44. McLoughin LC, Davis NF, Dowling CM, et al. Outcome of deceased
and Ireland. Transplantation 1998;65:745–6. donor renal transplantation in patients with an ileal conduit. Clin
30. Haring R, Nauck M, Vlzke H, et al. Low serum testosterone is associate Transplant 2014;28:307–13.
with increased mortality in men with stage 3 or greater nephropathy. 45. Wang ZP, Wang WY, Zhu YC, et al. Adjuvant chemotherapy with
Am J Nephrol 2011;33(3):209–17. gemcitabine plus cisplatin for kidney transplant patients with
31. Chatterjee R, Wood S, McGarrigle HH, et al. A novel therapy with tes- locally advanced transitional cell carcinoma. Transplant Proc
tosterone and sildenafil for erectile dysfunction in patients on renal 2016;48:2076–9.
dialysis or after renal transplantation. J Fam Plan Reprod Health Care 46. Tiliou X, Doerfler A. Urological tumors in renal transplantation.
2004;30(2):88–90. Minerva Urol Nefrol 2014;66(1):57–67.
32. Mazjoub A, Shoskes DA. A case series of the safety and efficacy of tes- 47. Kwatra V, Karanth NV, Priyadarshana K, et al. Pembrolizumab for
tosterone replacement therapy in renal failure and kidney transplant metastatic melanoma in a renal allograft recipient with subsequent
recipients. Transl Androl Urol 2016;5(6):814–8. graft rejection and treatment response failure: a case report. J Med
33. Carrero JJ, Qureshi AR, Parini P, et al. Low serum testosterone Case Rep 2017;11(1):73.
increases mortality risk among male dialysis patients. J Am Soc
Nephrol 2009;20(3):613–20.
30 Cardiovascular Disease in
Renal Transplantation
JENNIFER S. LEES and ALAN G. JARDINE

CHAPTER OUTLINE Introduction Renal Function


Background: CVD in CKD Smoking
Epidemiology of Posttransplant CVD New-Onset Diabetes after Transplantation
Novel and Transplant-Specific Risk Factors Obesity and the Metabolic Syndrome
Specific Risk Factors and Management Other Risk Factors and Interventions
Hypertension and Uremic Screening
Cardiomyopathy Intervention and Secondary Prevention
Phosphate and the FGF23-Klotho Axis Other Cardiovascular Conditions: Valvular
Vascular Stiffness and Calcification Disease, Arrhythmia, and Stroke
Guidelines and Observed Patterns of Predicting Cardiovascular Risk
Usage Trial Endpoints
Dyslipidemia Conclusion

Introduction to RTRs.1,3 Finally, we are limited in our ability to provide


guidelines for the management of CVD in this population
A successful renal transplant is the most effective way of because of the lack of epidemiologic and clinical trial data in
reducing the incidence of cardiovascular disease (CVD) this patient population.
and cardiovascular (CV) mortality in patients with end-
stage renal disease (ESRD). Although the risk of CVD in
renal transplant recipients (RTR) is approximately three Background: CVD in CKD
times that of the general population, this is much less than
the 10- to 20-fold increase in CV risk in patients receiving The recognition that CKD is associated with increased CV
maintenance hemodialysis.1–3 Premature CVD is also a sub- risk has resulted in the adoption of estimated glomeru-
stantial cause of graft failure, due to “death with a function- lar filtration rate (eGFR) as a CV risk equivalent.6 As GFR
ing graft.”4,5 These observations are well established and declines in patients with CKD, the risk of CVD increases
illustrated in Fig. 30.1. progressively, with the highest risk being in ESRD. In early
There are specific problems in managing CVD in RTRs CKD, the pattern of CVD is probably similar to the general
compared with the general population. First, RTRs are not population, with an increased risk of lipid-dependent coro-
a homogeneous group; there is variability in duration and nary artery disease (CAD). In more advanced CKD, there is a
underlying cause of chronic kidney disease (CKD) and dialy- disproportionate increase in deaths due to heart failure and
sis history, which determine the individual burden of accu- sudden, presumed arrhythmic deaths. The latter pattern is
mulated CV risk. Second, the transplant population carries similar to that seen in advanced heart failure, where CHD is
specific risk associated with transplantation surgery and the uncommon, regardless of the primary etiology of heart fail-
need for immunosuppressive therapy with agents that have ure.3 In both ESRD and advanced heart failure, total serum
direct and indirect effects on CV risk factors. Third, unlike cholesterol levels are low, markers of inflammation are
the general population, in patients with ESRD including elevated, and the conventional relationship between lipids
RTR, coronary heart disease (CHD) is not the dominant and total CV events (CVE) is lost (or even reversed). More-
pathophysiologic process. Structural and functional abnor- over, treatment strategies proven in the general population,
malities in the heart—uremic cardiomyopathy—contrib- specifically statins, have little or no effects in these patient
ute to an increased risk of heart failure and sudden death; groups,7–11 presumably reflecting the low proportion of the
and vascular changes including calcification, together with overall CV burden which is due to cholesterol-dependent
conventional risk factors, contribute to an increased risk coronary disease.
of stroke and peripheral vascular disease. Thus traditional The determinants of excess CVD in advanced CKD
risk factor relationships and therapeutic strategies derived include vascular calcification, elevated phosphate (and
from the general population may not be directly applicable fibroblast growth factor-23 [FGF23]), hypertension,
496
30 • Cardiovascular Disease in Renal Transplantation 497

GP 3 Infection Other

Deaths/100 pt yrs w/functioning graft


100
Tx Malignancy Unknown
HD CVD
10
2
% mortality p.a.

1
0.1

0.01 0
4 12 20 28 36 44 52 60 68 76 84 92
Years
0.001 Fig. 30.2 Deaths per 100 patient years, in a renal transplant popula-
25-34 35-44 45-54 55-64 65-74 75-84 84+ tion, showing cause of death with time after transplantation surgery.
Age group (years) The data include first-time kidney-only transplant recipients, age 18
and older, and transplanted between 1997 and 2006, who died with a
Fig. 30.1 Percentage cardiovascular mortality per annum (p.a.) by age functioning graft (n = 14,169). CVD, cardiovascular disease. (Data from
group in the general population (GP), renal transplant population (Tx), the USRDS annual report; 2008. www.usrds.org.)
and hemodialysis population (HD). The figure demonstrates the vastly
increased cardiovascular risk in patients on hemodialysis; this risk is
improved in patients who are transplanted, but not back to baseline. rates appears to be stable or even falling in RTR, reflect-
(Derived from the USRDS. Foley RN, Parfrey PS, Sarnak MJ. Clinical epidemi-
ology of cardiovascular disease in chronic renal disease. Am J Kidney Dis ing improved management of transplant recipients and
1998;32:S112–9.) improved graft outcomes.4,5,16,17

inflammation, malnutrition–bone mineral disease, and


Epidemiology of Posttransplant
intravascular volume overload.1,3 Vascular stiffness and CVD
hypertension, and elevated levels of the phosphaturic
hormone FGF23, contribute to the development of ure- Registry and cohort studies, and a small number of clinical
mic cardiomyopathy,12 the most common form of which trials, have examined the natural history and determinants
is extreme left ventricular hypertrophy (LVH) with fibro- of CVD in RTR. In looking at these data it is important to
sis which, in turn, promotes the development of systolic consider background improvements in immunosuppressive
dysfunction and sudden arrhythmic death.13 Overall, the therapy, the increasing age of transplant recipients, and
pattern of CVD in ESRD, its pathogenesis, and the implica- the use of kidneys from extended criteria donors (where the
tions for treatment are markedly different from the gen- expectations for graft function are less good). Each of these
eral population; although the risk of CHD is increased, the factors is likely to influence the pattern of CVD in RTR in
disproportionate increase is in sudden death and death the future. It is also important to remember that endpoints
due to heart failure. recorded in registries may be inaccurate, and investigators
Potential RTR recipients carry this burden of accumu- in clinical trials often pool CV endpoints that are believed to
lated CV risk—conventional and nonconventional—from share common pathophysiologic mechanisms, which may
time spent with CKD and on maintenance hemodialysis to not be true in atypical populations, including RTR.1,3,18 A
the posttransplant period. After transplantation surgery, final issue that is often ignored is that of “competing risk,”
there is an abrupt increase in overall (and CV) events and where an individual risk factor may contribute to more
mortality, particularly in the early postoperative period than one adverse outcome. In transplant recipients, for
(Fig. 30.2). This falls progressively thereafter, and survivors example, smoking may increase the risk of infection, malig-
beyond the first few months have a mortality rate approxi- nancy, and CVD, thus diminishing the apparent effect on
mately half that of patients receiving maintenance dialy- any single outcome.19
sis,14 and a comparable reduction in nonfatal CV events.3 The early studies of Kasiske and colleagues, who followed
After transplantation, several specific CV risk factors are up more than 1000 RTRs in a single US center,20,21 dem-
more prevalent. Dyslipidemia and hypertension are both onstrated a high incidence of CVEs and CV mortality, and a
very common, affecting nearly all patients. Lipid levels rise high prevalence of preexisting CVD in RTR. They confirmed
in the weeks after transplantation,15 reflecting improved that conventional CV risk factors, including age, sex, smok-
well-being, diet, and immunosuppressive agents. Immu- ing status, and the presence of diabetes mellitus (either
nosuppressive agents also contribute to hypertension and preexisting or developing after transplantation), were asso-
to the development of diabetes (new-onset diabetes after ciated with the development of CVEs (which they termed
transplantation [NODAT]). These risk factors, together “coronary heart disease”). For each year of life, the risk of
with the level of posttransplant renal function (the relation- a CVE was increased by 3% to 5%; males and patients with
ship between eGFR and CVD being similar to that seen in diabetes had a twofold increase in risk of a CVE. However,
primary CKD), preexisting CVD, and the increasing age of the strongest risk factors were preexisting CHD, peripheral
RTR, contribute to the overall level of CV risk after trans- vascular disease, or cerebral vascular disease, reflecting
plantation.1,3 However, in spite of this, CVD and mortality the importance of the burden of CV disease that individual
498 Kidney Transplantation: Principles and Practice

patients carry at the time of transplantation. Most of the TABLE 30.1 Data on Risk Factors for Myocardial
risk factors they identified, such as preexisting disease, age, Infarction and Sudden Cardiac Death From the FAVORIT
and sex, are irremediable, and it has proved more difficult Study140
to identify any relationship between modifiable risk factors
Confidence
and CVEs.1,3,20,21 RR Interval P
Kasiske’s initial analysis revealed no association between
posttransplant levels of triglyceride, total or low-density Age 1.13 (1.08, 1.19) <0.0001
Diabetes 2.30 (1.90, 2.80) <0.0001
lipoprotein (LDL) cholesterol, and CVE in RTR.21 However, Smoking (current) 1.38 (1.05, 1.82) 0.07
a subsequent larger analysis20 did show an association of Cardiovascular disease 2.06 (1.71, 2.48) <0.0001
risk with hyperlipidemia, albeit only with very high levels Low-density lipoprotein 1.01 (0.98, 1.04) 0.41
of total cholesterol and increased risk of long-term CVE. Systolic blood pressure 1.17 (1.11, 1.23) <0.0001
Unlike the general population there is not a clear, progres- Diastolic blood pressure 0.89 (0.81, 0.98) 0.02
Body mass index 0.91 (0.84, 0.98) 0.02
sive relationship between lipid levels and CVEs. This obser- Lymphoproliferative 0.84 (0.70, 1.01) 0.07
vation, in keeping with the pattern seen in patients with disease
ESRD receiving maintenance dialysis,22 provides support
for the notion that CVD in RTR differs from the traditional RR, relative risk of event with 95% confidence intervals for age in years,
diabetes (presence or absence of), cigarette smoking, preexisting car-
atherosclerotic model.1,3 Similar observations have been diovascular disease, blood pressure, and body mass index with levels of
reported in single-center studies from Europe.23 Long-term significance.
follow-up of clinical trials provides additional data, with
the benefit that endpoints are externally validated and
more accurate than registry data.24–28 The ALERT (Assess- 23,575 adult RTRs for a median of 4.5 years. CVD disease
ment of LEscol in Renal Transplantation)25–27 and, more was defined as a composite of proven myocardial infarc-
recently, the FAVORIT (Folic Acid for Vascular Outcome tion, coronary intervention, and cardiac death. The over-
Reduction in Transplantation)24,28 studies have been used all cumulative incidence was 3.1%, 5.2%, and 7.6% at 1,
to provide data on CVEs collected during follow-up of poten- 3, and 5 years after transplantation, respectively. In the
tial interventions in large populations of RTR. In the ALERT first year the distribution of events was nonfatal myocar-
study26 2100 stable RTRs were randomized to receive pla- dial infarction (49%), coronary intervention (38%), and
cebo or fluvastatin (40–80 mg/day) and followed for up cardiac death (13%); beyond 1 year the corresponding
to 8 years.25,26 Compared with studies of statin therapy values were 39%, 38%, and 23%. Conventional modifi-
in nontransplant populations at comparable high risk of able CV risk factors were very poor predictors of cardiac
CVEs, there are clear differences. Nontransplant patients events, and varied with time after transplantation. Early
with dyslipidemia and a history of CAD are likely to have events were predicted by age, male sex, history of cancer or
further coronary events, and the risk of cardiac death is diabetes, obesity, preexisting CV disease (CHD, peripheral
approximately one-third that of a nonfatal event.7,9,10 In vascular disease, or cerebrovascular disease), deceased
contrast, patients with ESRD receiving maintenance dialy- donor transplant, and time on dialysis before transplan-
sis8,11 are much more likely to suffer cardiac death than a tation. Conventional risk factors such as smoking, hyper-
nonfatal coronary event. RTRs occupy a position interme- cholesterolemia, and hypertension were not significant,
diate between these populations, with the increase in CVEs although they did correlate with a past history of CVD.
associated with an equal risk of cardiac death and nonfatal Later events were dependent on poor graft function (low
coronary events.25,26 It is likely that this alteration in pro- eGFR; and factors that adversely influence graft function
portions reflects an increase in the risk of death due to pri- such as acute rejection, delayed graft function, and post-
mary arrhythmia or heart failure, a pattern similar to that transplant lymphoproliferative disease), the development
seen in patients with congestive heart failure.29 Of note, of NODAT, and race.
around 10% of otherwise stable RTRs experienced a cardiac Some of the differences in the analyses just discussed may
event during the first 5 years of follow-up—an event rate be explained by pooling endpoints. The ALERT study also
(2% per annum) comparable to the annual mortality rate allows us to examine the relationship between risk factors
of stable RTR and the annual graft failure rate after the first and individual CVE (e.g., acute myocardial infarction [aMI]
year.26,28,30 or cardiac death; Fig. 30.3), and to examine relationships
In the FAVORIT24,28 study 4110 stable RTRs were ran- masked by pooling of CVE with different determinants.27,33
domized to high-dose folic acid, the primary endpoint being In a multivariate analysis, the leading potentially remedi-
a vascular composite of myocardial infarction, CV death, able determinants of aMI (in addition to age, gender, and
resuscitated CVD, revascularization procedures (coronary preexisting diabetes) were lipid levels. As in the general
and noncoronary), and stroke. Given these analyses and population, all major serum lipid subfractions were associ-
the potentially disparate nature of the pooled endpoints, it ated with aMI: total and LDL cholesterol, and triglyceride
is perhaps not surprising that there was no benefit of the with an increased risk; HDL cholesterol with reduced risk.27
intervention, that LDL cholesterol had no relationship with In contrast, no lipid subfraction was significantly associated
the composite outcome, and that the main determinants with cardiac death, the main determinants of which were
were age, preexisting CVD, diabetes, systolic blood pres- low eGFR and LVH, particularly when associated with sub-
sure, and low eGFR.24,28 Table 30.1 shows the relationship endocardial ischemia (LVH with “strain”) and pulse pres-
between risk factors and CVEs in this population. sure.27 These observations strongly support the established
A prospective multinational study—the PORT (Patient literature on “uremic cardiomyopathy” and suggest that
Outcomes in Renal Transplantation) study31,32—followed severe LVH, driven by renal dysfunction, hypertension, and
30 • Cardiovascular Disease in Renal Transplantation 499

the presence of LVH at the time of transplantation, may lead


to increased risk of death due to heart failure or arrhyth- Novel and Transplant-Specific
mia—with or without coexistent coronary disease. Risk Factors
The key message from these observations is that RTRs do
suffer from CAD (fatal and nonfatal MI), the determinants of In the general population, the limited predictive ability
which are similar to the general population. However, car- of conventional CV risk factors has led to the search for
diac death is perhaps a greater problem, the determinants of novel risk factors and potential therapeutic targets. The
which are LVH, vascular stiffness, and hypertension. Stud- presence of inflammation has become a central mecha-
ies by other investigators, including Abbott34 and Rigatto35 nism, with the recognition that inflammatory cells are
support these findings and underscore the observation that involved in atherosclerosis and that circulating markers
noncoronary events such as heart failure are common. In of inflammation, such as C-reactive protein, can iden-
addition, they demonstrated that specific transplant risk tify patients at increased risk of atherosclerotic vascular
factors including graft dysfunction (specifically graft failure) disease who may benefit from established treatments.
were associated with an approximately threefold increase In RTRs there are similar initiatives. Markers of inflam-
in CVEs, including heart failure. Anemia proved to be a risk mation and circulating inhibitors of endothelial func-
factor for the development of heart failure, although with tion38,39 have been studied in transplantation and are
improved anemia management a relationship with hemo- associated with an increased risk of CVD. Patients with
globin is now difficult to confirm.3,35–37 simple features of inflammation, such as low albumin,
are at higher risk.23,38 There are also transplant-specific
risk factors, including those factors that contribute to
poor graft function. These include the occurrence and
20 severity of acute rejection episodes, delayed graft func-
tion, chronic rejection, cytomegalovirus infection, and
Placebo other factors.3,36,37
Cumulative incidence (%)

15 Fluvastatin

Specific Risk Factors and


10 Management
Cardiac death or definite
non-fatal MI P = 0.014 In this section we will cover individual CV risk factors,
5 their role, and their management. As noted previously, it
is important to realize that transplantation is one phase
0 in the course of progressive renal disease. Patients bring
with them to transplantation accumulated risk, much of
Patients at risk 0 1 2 3 4 5 6 7
Fluvastatin 1050 1031 1006 973 938 891 811 746 which is irremediable. For example, vascular stiffness and
Placebo 1052 1030 1002 971 935 911 820 761 calcification, which develop in advanced CKD, contribute
Time (years) to hypertension after transplantation. Moreover, nearly
Fig. 30.3 Kaplan–Meier curves of time to cardiac death and nonfatal
all of the immunosuppressive drugs that have revolution-
acute myocardial infarction (MI) in the ALERT study (with extended ized the management of transplant recipients have effects
follow-up) of fluvastatin 40 to 80 mg/day versus placebo in 2100 renal on CV risk factors—some good, such as higher GFR; oth-
transplant recipients. Statin therapy was associated with a reduction in ers potentially bad, such as hypertension and dyslipid-
cardiac death and nonfatal MI during 8 years of follow-up (P = 0.014, emia. The pattern of effects of immunosuppressive agents
log rank test). (Data from Holdaas H, Fellstrom B, Cole E, et al. Long-term
cardiac outcomes in renal transplant recipients receiving fluvastatin: the is shown in Table 30.2,1,3 and discussed in more detail
ALERT extension study. Am J Transplant 2005;5:2929–36.) later.

TABLE 30.2 Effects of Immunosuppressive Agents in Conventional Cardiovascular Risk Factors, Including Hypertension, Left
Ventricular Hypertrophy, Total Cholesterol, Low-Density Lipoprotein Cholesterol, Triglycerides, Diabetes Mellitus (New-Onset
Diabetes After Transplantation), and Renal Function
Azathioprine/
Cardiovascular Risk Factors Steroids MMF Belatacept Cyclosporine Tacrolimus mTOR Inhibitors
Hypertension 1.1 ↑ 1.2 ↔ 1.3 ↔ 1.4 ↑ 1.5 ↑ 1.6 ↔
Left ventricular hypertrophy 1.7 ↑ 1.8 ↔ 1.9 ↔ 1.10 ↑ 1.11 ↑ 1.12 ↔
Total cholesterol 1.13 ↑ 1.14 ↔ 1.15 ↔ 1.16 ↑ 1.17 ↑ 1.18 ↑
Low-density lipoprotein 1.19 ↑ 1.20 ↔ 1.21 ↔ 1.22 ↑ 1.23 ↑ 1.24 ↑
Triglycerides 1.25 ↑ 1.26 ↔ 1.27 ↔ 1.28 ↑ 1.29 ↑ 1.30 ↑
Diabetes mellitus 1.31 ↑ 1.32 ↔ 1.33 ↔ 1.34 ↑ 1.35 ↑ 1.36 ↑
Renal function 1.37 ↔ 1.38 ↔ 1.39 ↔ 1.40 ↓ 1.41 ↓ 1.42 ↔

The arrows indicate an adverse or beneficial effect, or the absence of any significant effect, for the individual classes of agents, including corticosteroids, azathio-
prine/mycophenolate mofetil (MMF), cyclosporine, tacrolimus, mammalian target of rapamycin (mTOR) inhibitors, and newer biologic agents.
500 Kidney Transplantation: Principles and Practice

Recipient age 20–49 years


Hypertension and Uremic 12
Cardiomyopathy
10
Hypertension is an almost invariable accompaniment
of renal transplantation—a consequence of preexisting

% Cardiovascular death
hypertension at the time of transplantation and the effects 8
1 yr 3 yr
of immunosuppressive agents. Both increased vascular
resistance and increased intravascular volume contribute >140 >140 n = 2358
to the development of hypertension. With declining GFR 6
as CKD progresses, salt and water excretion are impaired,
>140 140 n = 2358
and volume-dependent mechanisms assume greater impor- 4 140 >140 n = 2414
tance; after transplantation the contribution of volume-
dependent mechanisms will similarly depend on the level of
graft function,3,40,41 Details on the role of vasoconstrictor 4 140 140 n = 8106
mechanisms and the relevance for treatment are described
later.
Hypertension in RTRs is known to be associated with 0
0 2 4 6 8 10
poorer patient and graft outcomes. There is no trial evi-
dence for specific blood pressure targets in the RTR popula- A Years
tion, but data from the European Registry support the need
to manage hypertension and inform treatment targets. In Recipient age 50 years
a series of RTRs with a functioning graft 1 year after trans- 12
plantation, Opelz and colleagues42,43 demonstrated that 1y 3y
blood pressure—recorded at outpatient clinics—is a major 10 >140 >140 n = 2420
determinant of long-term patient and graft survival, albeit >140 140 n = 1760
not independently from graft function. Current guidelines
% Cardiovascular death

140 >140 n = 1644


recommend a blood pressure target <130/80 mmHg, irre- 8 140 140 n = 3344
spective of the level of proteinuria,44 but in fact, the data
suggest that graft outcomes start to deteriorate when sys-
tolic blood pressure is above 120 mmHg.42,43 Similarly, 6
there appears to be an important relationship between
blood pressure across the range from “normal” to hyper- 4
tensive and the development of posttransplant CV disease
(Fig. 30.4).42,43 Epidemiologic studies, which include the
placebo arms of interventional trials in transplant recipi- 4
ents, have confirmed that hypertension is associated with
CVEs,27,28,45,46 specifically stroke, cardiac death, and heart
0
failure, rather than nonfatal coronary events. Hyperten-
0 2 4 6 8 10
sion was the strongest determinant of cardiac death in the
ALERT study,27 the most significant blood pressure param- B Years
eters being systolic blood pressure and pulse pressure, both Fig. 30.4 (A, B) Kaplan–Meier curves of cumulative incidence of cardio-
markers associated with vascular stiffness, rather than dia- vascular events by systolic blood pressure (mmHg) at 1 and 3 years of
stolic blood pressure. follow-up, in two age groups from the Collaborative Transplant Study.
In both age groups, having a systolic blood pressure <140 mmHg, com-
Most clinics use “office-based” blood pressure mea- pared with above 140 mmHg, at both 1 and 3 years of follow-up was
surements, using a standard sphygmomanometer. Blood associated with a reduced incidence of cardiovascular events. (Data
pressure should preferably be assessed using repeated mea- from Opelz G, Dohler B. Collaborative Transplant Study. Improved long-
surements, with the patient seated after a period of rest, or term outcomes after renal transplantation associated with blood pressure
measuring ambulatory47,48 or home blood pressures47,49 control. Am J Transplant 2005;5:2725–31.)
as a more informative measure. In patients with essential
hypertension these methods are recommended for patients RTRs, and LVH is strongly associated with poor outcome27
with resistant hypertension, or where “white coat” syn- in RTRs. The pathophysiology of LVH in CKD (uremic car-
drome is suspected. In transplant recipients ambulatory diomyopathy) is marked by the presence of subendocardial
recordings are associated with prognosis, and loss of diur- ischemia and myocardial fibrosis.13,54 Fibrosis is believed
nal profile, or loss of the “nocturnal dip,” confers additional to promote aberrant conduction and is associated with
prognostic information.50 These methods should be used in markers of arrhythmogenicity, such as prolonged QT inter-
patients with poor blood pressure control and may provide val and abnormal T-wave alternans,55 which provide the
additional information in clinical trials. likely link to fatal arrhythmias and sudden cardiac death.3
Hypertension exhibits unfavorable effects indirectly Arrhythmias may be spontaneous or complicate other-
through development of end-organ damage in RTRs— wise minor ischemic episodes. These observations identify
specifically proteinuria and LVH.51,52 Hypertension is the hypertension, LVH, and electrocardiographic abnormali-
major contributor to LVH in patients with ESRD,53 including ties as markers of adverse outcome in RTR, and as potential
30 • Cardiovascular Disease in Renal Transplantation 501

targets for intervention.3 The less common manifestation of because of concerns about the possible adverse effects in
uremic dilated cardiomyopathy (with systolic dysfunction) patients with undiagnosed, functional stenosis of the single
may be a sequel of LVH or may be associated with (often transplant renal artery.67 Caution should also be taken with
silent) CHD.3,13,56 Uremic cardiomyopathy develops, pri- regard to hyperkalemia in the context of ACE inhibition and
marily, during the time patients spend with advanced CKD, CNI.68 More radical approaches to the treatment of hyper-
and on maintenance dialysis programs, and is therefore tension, such as embolization or laparoscopic removal of the
common in new transplant recipients.53 Although there native kidneys, have been employed and may be effective.
are studies that suggest that the manifestations of uremic Whereas patients with bilateral native nephrectomy before
cardiomyopathy may improve after transplantation,57 with transplantation (including pediatric patients) may have good
apparent regression of LVH and improved systolic func- blood pressure control, the benefits are less clear in patients
tion, these studies may not reflect the true situation. Echo- with established hypertension after transplantation.41,68
cardiographic analyses are highly dependent on chamber Patient and graft survival is associated with prescription
diameters (e.g., in the estimation of left ventricular [LV] of ACEI/ARB in retrospective analyses (Fig. 30.5). There
mass)13,52 which are, in turn, dependent on hydration sta- are few prospective trials of antihypertensive treatment in
tus. Although patients with advanced CKD and treated by RTRs.69–72 In recent years, two trials have assessed the pre-
dialysis have a tendency to volume overload, this improves scription of ACE inhibitors on “hard” CV endpoints, with
after successful transplantation, correcting artifactual inconclusive results. Paoletti et al.72 report that treatment
overestimation of LV mass and systolic dysfunction. Hence, with lisinopril (5 mg once daily then titrated to response) is
studies that use volume-independent technology (spe- associated with a reduction in major CVEs and a composite
cifically cardiac magnetic resonance imaging) have not endpoint of death, major CVE, renal graft loss, or creatinine
shown similar improvement.45 Long-term risks associated doubling, compared with placebo. Knoll et al.70 report no
with the various manifestations of uremic cardiomyopa- effect of ramipril 5 mg once daily (OD) versus placebo on
thy in patients receiving maintenance dialysis are carried doubling of creatinine, graft loss, or all-cause mortality.
forward in patients who undergo transplantation.12,45,58 Most RTRs with hypertension require treatment with more
After transplantation, there are limited data on uremic than one antihypertensive agent.40,41,66,73 In the absence
cardiomyopathy, restricted to LVH, suggesting that effec- of specific evidence of CV risk reduction with any particu-
tive blood pressure control and the avoidance of calcineu- lar class of drug, the choice of antihypertensive agent, as in
rin inhibitors (CNIs) may reduce LVH.59,60 A series of small other populations, should consider the need to treat other
short-term studies suggests that the use of dihydropyridine comorbidities, for example, beta-blockers for patients with
calcium antagonists59; mammalian target of rapamycin symptomatic angina, and blockers of the renin–angiotensin
(mTOR) inhibitors,60 sirolimus or everolimus, in place of system for patients with proteinuria.66,68
CNI; or CNI withdrawal61 is associated with improvement
in blood pressure and regression—or lack of progression—
of LVH in RTRs.
Phosphate and the FGF23-Klotho
There is evidence of increased or inappropriate activa- Axis
tion of vasoconstrictor mechanisms in RTRs, including
the sympathetic nervous system, the renin–angiotensin FGF23, a phosphaturic hormone, works in conjunction with
system, and endothelin both in humans and experimental vitamin D and parathyroid hormone (PTH) to maintain cor-
animals.40,41,62,63 These, coupled with evidence of impaired rect serum levels of phosphorus. Phosphate handling and
endothelium-dependent (nitric oxide-mediated) vascular metabolism is impaired early in the course of CKD,74 and
relaxation,39–41,62,63 shift the balance toward vasoconstric- FGF23 has been established as a more sensitive biomarker
tion. The mechanisms underlying these phenomena are for disordered phosphate metabolism in CKD than serum
less well understood. Corticosteroids are associated with phosphate or PTH. FGF23 becomes elevated early in the
hypertension in other clinical conditions and have two disease course, whereas phosphate and PTH may become
principal actions—to promote retention of salt and water abnormal at GFR <30 (CKD stage 4), FGF23 levels are ele-
due to actions of corticosteroids on the kidney64 and to vated even in CKD stage 2, with around one-third of patients
enhance sympathetic activity, leading to increased vascu- with GFR 60 to 69 with elevated FGF23 levels.74
lar tone.64 CNIs cause hypertension through direct renal FGF23 appears to be responsible for persistent hypophos-
sodium retention and increased vasoconstrictor tone as phatemia seen in the early posttransplant period,75 but
well as indirectly via renal impairment, as a consequence of FGF23 levels usually will fall around 3 months after trans-
the nephrotoxic effects of CNI.65 plantation, then can reach a state of normal or near-normal
Several short-term studies have demonstrated that the from 1 year after transplant.75,76 Higher levels of FGF23 are
most commonly used antihypertensive drugs, such as angio- again seen as transplant function declines. In a cross-sec-
tensin receptor blockers, angiotensin-converting enzyme tional, observational study of 279 stable, prevalent RTRs,77
(ACE) inhibitors, and calcium channel blockers, have effects fewer than 50% of those with CKD stage 1 or 2 had nor-
on blood pressure comparable to those seen in other popu- mal FGF23 and PTH, and this reduced to 26.3% in those
lations.40,41,66 Dihydropyridine calcium channel antago- with more advanced transplant CKD. FGF23 is associated
nists, such as nifedipine and amlodipine, may attenuate the with LVH, inducing hypertrophy of isolated cardiac myo-
nephrotoxic effects of CNI40,41,64 and have been favored in cytes, and in in vivo mouse models,78 and this effect can be
the early phases after transplantation. Blockers of the renin– ameliorated by administration of FGF23 antagonists, inde-
angiotensin system have been favored in patients with pro- pendent of blood pressure.78,79 FGF23 is associated with
teinuria40,41,44 and LVH, although the uptake has been slow cardiovascular and all-cause mortality, and allograft loss
502 Kidney Transplantation: Principles and Practice

1 1
Log-rank P < 0.002 Log-rank P = 0.002

0.8 0.8

Proportion functioning
Proportion alive

0.6 0.6

0.4 0.4

ACEI/ARB ACEI/ARB
0.2 No ACEI/ARB 0.2 No ACEI/ARB

0 0
0 2 4 6 8 10 12 0 2 4 6 8 10 12
Patient survival time (years) Graft survival time (years)
Patients at risk, ACEI/ARB: Patients at risk, ACEI/ARB:
1250 1020 774 559 396 228 103 1190 925 671 456 300 153 67

Patients at risk, no ACEI/ARB: Patients at risk, no ACEI/ARB:


781 511 390 276 180 107 51 841 489 355 240 148 83 42
Fig. 30.5 A retrospective analysis of patient and graft survival in patients taking and not taking angiotensin-converting enzyme inhibitors (ACEI) or
angiotensin receptor blockers (ARB), adjusted for covariates. Patient and graft survival was better in patients prescribed ACEI/ARB. (Data from Heinze
G, Mitterbauer C, Regele H, et al. Angiotensin-converting enzyme inhibitor or angiotensin II type 1 receptor antagonist therapy is associated with prolonged
patient and graft survival after renal transplantation. J Am Soc Nephrol 2006;17:889–99.)

after transplantation,80,81 after adjustment for known car- Moreover, they are also linked to adverse CV outcomes in
diovascular risk factors and markers of mineral handling. RTR, with coronary artery calcification at time of trans-
It is likely that LVH induced by elevation of FGF23 explains plantation also being predictive of cardiac events,88 even
some of the excess CV morbidity and mortality in RTRs. in those asymptomatic patients with no prior history of
Independent of FGF23, phosphate is a risk factor for vascu- CVD.89 These measures provide potential short-term sur-
lar disease in patients with CKD and renal transplantation. rogate endpoints for trials of CV interventions in this
By contrast to FGF23, this is likely mediated through direct patient group.90
effects on blood vessels, and by promoting vascular calcifi- There are no treatments proven to improve vascular
cation (discussed later). In 1501 patients from the Chronic calcification or stiffness in RTRs. Vitamin K deficiency is a
Renal Insufficiency Cohort (CRIC), coronary and thoracic potentially remediable risk factor of vascular stiffness and
aorta calcification were measured by computed tomogra- calcification. Vitamin K is essential to facilitate carboxyl-
phy scan. Serum phosphate, but not FGF23, was found to ation (activation) of various Gla proteins, including Matrix
be associated with presence and severity of arterial calcifi- Gla protein—an enzyme that opposes vascular calcifica-
cation, through a direct effect on inducing calcification of tion in its active form. Vitamin K deficiency is common in
vascular smooth muscle cells.82 Furthermore, in vitro stud- RTRs91,92 and is associated with mortality.92 Vitamin K
ies support a direct effect of high phosphate on endothelial supplementation appears to improve vascular/valvular cal-
function (via interference with the nitric oxide pathway),83 cification and vascular stiffness in some populations.93–98
and high dietary phosphate load effects on vascular func- Studies of vitamin K supplementation in CKD and dialysis
tion,83 with impairment of vessel relaxation, independent of are underway.
serum phosphate. Thus although FGF23 and phosphate are
intrinsically linked in maintaining phosphate homeostasis,
they appear to contribute to cardiovascular risk in RTRs
Guidelines and Observed Patterns of
independently and via distinct mechanisms. Usage
There are numerous guidelines on the management of
Vascular Stiffness and hypertension after transplantation.68 Although these are
Calcification based on the evidence discussed in this chapter, they also
draw on the practical expertise of the guideline committees.
Vascular calcification (particularly of the medial arte- In the absence of studies that have addressed appropriate tar-
rial/arteriolar layer) and stiffness (secondary to calcifica- gets for blood pressure control in RTR, they have endorsed
tion or vascular hypertrophy) are common in the setting targets derived from observational studies, such as that of
of progressive CKD, ESRD, and in RTRs,7,52,55 Calcifica- Opelz and colleagues42,43 and targets taken from other popu-
tion persists and appears to continue to progress after lations, specifically those with CKD. Thus the targets are arbi-
renal transplantation,84 although vascular stiffness may trary and are not aimed, for example, at regression of LVH or
be partially reversed after kidney transplantation.85 Stiff, specific reversal of proteinuria. The recently published Kid-
calcified vessels increasing systolic hypertension, increase ney Disease Improving Global Outcomes (KDIGO) guidelines
afterload, and contribute to the development of LVH.86,87 suggest a target of 130/80 mmHg in patients with diabetes
30 • Cardiovascular Disease in Renal Transplantation 503

16

Odds ratio for medication


use within 4 months
4

0
1990–94

1995–99

2000–06

1990–94

1995–99

2000–06

1990–94

1995–99

2000–06

1990–94

1995–99

2000–06

1990–94

1995–99

2000–06

1990–94

1995–99

2000–06

1990–94

1995–99

2000–06

1990–94

1995–99

2000–06
Beta- ACE/ARB Anti- Diuretic CCB Other anti- Statin Other lipid-
blocker platelet hypertensive lowering
A agent

Beta-blocker Statin
Antiplatelet Other antihypertensive
CCB ACE/ARB
Diuretic Other lipid-lowering agent

70

60

50
% Cardiovascular death

40

30

20

10

0
1990 1992 1994 1996 1998 2000 2002 2004 2006
B Year of transplant
Fig. 30.6 Graph demonstrating the use of cardioprotective medications in the PORT study, documenting changes in prescription over time posttrans-
plant. (A) Adjusted odds ratios for CVD medication use in the first 4 months posttransplant in more recent eras. (B) Model adjusted for age, sex, race,
and primary cause of renal failure. ACE/ARB, angiotensin-converting enzyme inhibitors/angiotensin II receptor blockers; CCB, calcium channel blockers.
(Data from Pilmore HL, Skeans MA, Snyder JJ, et al. Cardiovascular disease medications after renal transplantation: results from the Patient Outcomes in Renal
Transplantation study. Transplantation 2011;91:542–51.)

or proteinuria, the use of lifestyle modifications including salt in usage with time in recent years. The observation that
restriction, and the use of blockers of the renin–angiotensin 50% of patients receive a blocker of the renin–angiotensin
system as first-line therapy.68 Moreover, they endorse the use system suggests that reluctance to use these agents is less
of specific targets and agents determined by comorbidity. The than it was (Fig. 30.6).32,67 Data on achieved blood pres-
European and American Transplant Society guidelines are sure and the use of individual agents are difficult to obtain.
broadly concordant.99 In our own center, a proportion of patients remain uncon-
The implementation of guidelines and the adoption of trolled despite therapy; the majority of those controlled
new practices is dependent on the behavior of clinicians require multiple agents when we considered a historical
as much as the available evidence. Two large-scale studies target of 140/90 mmHg.73 Whether or not blood pressure
have examined the use of CV drugs and risk management targets are appropriate and whether one agent has ben-
in RTR in North America and in Australasia. The results efits over another require to be assessed in a prospective
are encouraging in that they show a progressive increase clinical trial.
504 Kidney Transplantation: Principles and Practice

Dyslipidemia Fluvastatin is not metabolized by CYP3A4, an enzyme


inhibited by CNIs. In patients receiving CNI, use of statins
Dyslipidemia is almost an invariable accompaniment of metabolized by CYP3A4 (specifically simvastatin, lovas-
renal transplantation.1,3 The pattern typically comprises tatin, and, to a lesser extent, atorvastatin) is associated
elevated total and LDL cholesterol, triglycerides, and HDL with increased statin bioavailability and with increased
cholesterol. There are also increased concentrations of efficacy and side effects.105 An important, underappreci-
intermediate—highly atherogenic—lipoproteins, includ- ated message is that, with the exception of fluvastatin and
ing small, dense LDL.100,101 The mechanisms behind this pravastatin, statins should be started at very low dose and
dyslipidemia include impaired renal function and the monitored cautiously in CNI-treated RTR. A study of flu-
influences of immunosuppressive agents. The mecha- vastatin at the time of transplantation (SOLAR),15 which
nisms that lead to dyslipidemia in CKD will contribute proved that the pleiotropic effects of statins on lymphocyte
to a varying degree in RTR, depending on the achieved function do not reduce the risk of acute rejection in RTR,
level of renal function, and explain the varying patterns showed no increase in adverse effect. Thus of the available
of dyslipidemia observed. In CKD the typical abnormali- statins, there is most evidence for the safety of fluvastatin,
ties are high triglycerides, low HDL cholesterol, elevated particularly in the perioperative period.15,25,26
intermediate-density lipoprotein (IDL) cholesterol, and a Recently, the SHARP study examined the use of simvas-
neutral effect on LDL and total plasma cholesterol,101 the tatin plus ezetimibe on dyslipidemia and outcomes in 9000
main mechanisms being reduced activity of lipoprotein patients with CKD, including 3000 on maintenance dialy-
lipase, and hepatic lipase and lipoprotein receptor dys- sis. Although medication was not continued in patients
function.101 To the effects of impaired renal function are who received transplants, there was an overall benefit in
added the effects of individual antirejection agents that the incidence of atherosclerotic CVEs that lends support to
have specific, often synergistic effects on serum lipid levels. the use of statin-based therapy for patients with progressive
Corticosteroids cause an increase in total and LDL choles- CKD, including those who will ultimately receive trans-
terol, triglycerides, and HDL cholesterol; CNIs—cyclospo- plants.106 This study adds to the ALERT data set, and is con-
rine and, to a lesser extent, tacrolimus—at commonly sistent with registry data and retrospective uncontrolled
used doses increase total and LDL cholesterol; and mTOR studies.107
inhibitors—sirolimus and everolimus—increase total The use of statins has been adopted by guidelines for min-
cholesterol, LDL cholesterol, HDL cholesterol, and triglyc- imizing CV risk in RTR.34,108 These guidelines have tended
eride in a dose-dependent manner.100,102,103 Typically, in to adopt lipid targets from the general population (LDL
the first 6 weeks after transplantation, immunosuppres- cholesterol for adult patients of 2.6 mmol/L), as there are
sion, normalization of renal function, and increased appe- inadequate data on targets specific for the transplant popu-
tite are associated with an increase in total cholesterol of lation. Of interest, however, is the fact that the most recent
around 1.0 to 1.5 mmol/L, an increase in LDL cholesterol KDIGO guidelines (www.kdigo.org)109 on lipid lowering in
of around 1 mmol/L, and increased triglyceride and HDL CKD have proposed that it is more important to establish
cholesterol.15 patients on statin therapy than it is to achieve a target, and
Statin therapy is one of the few interventions to be they have not recommended a target-driven approach in
tested in a large CV outcome study in RTR.25,26 The main this patient population (including RTR). Although there is
ALERT trial studied 2100 stable, cyclosporine-treated a strong rationale for statin use, one report suggests much
RTRs, followed for up to 6 years, and randomized initially lower uptake of therapy in RTR compared with other high-
to fluvastatin 40 to 80 mg daily or placebo. The primary risk populations.67 However, there is a pattern of increasing
endpoint was a composite of myocardial infarction, car- use that suggests that transplant clinicians may simply be
diac death, stroke, and coronary intervention. A 2-year cautiously following the trend in other patient groups (see
extension, where all patients were offered fluvastatin 80 Fig. 30.6).32
mg/day, prolonged follow-up to 8 years.25,26 With hind- One reason for the slow adoption of statin therapy and
sight, the core study was underpowered for the chosen CV risk management in RTR is the inherent relation-
composite primary endpoint, although statin therapy ship between immunosuppression and dyslipidemia; the
was associated with a 35% reduction in MI. With pro- expectation is that posttransplant reduction of immu-
longed follow-up, there was a significant reduction in the nosuppression will correct dyslipidemia. Many studies
primary endpoint in those patients randomized to statin have investigated the short-term effect of modification
therapy and to a variety of individual cardiac endpoints. of immunosuppressive therapy alone on hyperlipidemia
The main effect was on lipid-dependent endpoints such as in transplant recipients, including steroid withdrawal or
MI (see Fig. 30.3).26 Fluvastatin reduced LDL by 1 mmol/L avoidance110,111 or CNI withdrawal.112 The only study to
for the duration of the study and was well tolerated, with directly compare modification of immunosuppression with
placebo-like side effects. Post hoc analyses of this study the initiation of lipid-lowering therapy is the study of Wiss-
revealed that early introduction after transplantation was ing and colleagues.113 In this study patients were switched
associated with additional benefit,104 Overall, the ALERT from cyclosporine to tacrolimus-based therapy, and this
study can be summarized as showing that fluvastatin has was compared with the addition to atorvastatin. Although
beneficial effects on the secondary dyslipidemia associ- tacrolimus-based therapy was associated with a reduction
ated with renal transplantation, that this translates into in total LDL cholesterol and triglycerides, patients on cyclo-
reduced incidence of MI (with a lesser reduction in other sporine and atorvastatin had lipid levels comparable to
CV events), and that to maximize benefits, early initiation those on tacrolimus and atorvastatin combined. Thus mod-
of therapy is important. ification of the CNI provided no additional benefit to statin
30 • Cardiovascular Disease in Renal Transplantation 505

1.0 1.0
Adjusted probability without a CVD event

Adjusted probability without death


0.9 0.9

eGFR categories eGFR categories


1 <30 1 <30
0.8 2 30–45 0.8 2 30–45
3 45–60 3 45–60
4 60–75 4 60–75
5 75+ 5 75+

0.7 0.7
0 500 1000 1500 2000 2500 0 500 1000 1500 2000 2500
A Time to event (days) B Time to event (days)

Fig. 30.7 Effect of renal function on long-term cardiovascular event (A) and patient death risk (B) from the FAVORIT trial. Patients were stratified into
five groups based on estimated glomerular filtration rate (eGFR), demonstrating that patients with poorer renal function are at increased risk of adverse
events. CVD, cardiovascular disease. (Data from Bostom AG, Carpenter MA, Kusek JW, et al. Homocysteine-lowering and cardiovascular disease outcomes in
kidney transplant recipients: primary results from the Folic Acid for Vascular Outcome Reduction in Transplantation trial. Circulation 2011;123:1763–70; Weiner
DE, Carpenter MA, Levey AS, et al. Kidney function and risk of cardiovascular disease and mortality in kidney transplant recipients: the FAVORIT trial. Am J
Transplant 2012;12:2437–45.)

therapy. Additionally, the fact that some components of the outcomes in trials of transplantation is important, specifi-
dyslipidemia (e.g., hypertriglyceridemia) are insensitive to cally, to permit long-term graft and patient outcomes (see
statin therapy, and that both atherogenic and potentially later).117
protective lipid subfractions (HDL cholesterol) are increased Nonetheless, graft function has emerged as a strong
with immunosuppression has heightened reluctance to use determinant of graft and patient survival, and of CV risk
statins. Most clinicians and patients are reluctant to change in transplant recipients. Post hoc analyses of the two larg-
immunosuppression primarily on the basis of dyslipidemia, est CV outcome trials in RTR, FAVORIT and ALERT,27,28
without data to support long-term outcomes with this have shown renal function to predict the risk of graft loss
strategy. and of patient outcomes. Fig. 30.7 shows the relation-
Finally, there is little trial evidence, and considerable neg- ship between GFR and outcomes in the FAVORIT study.28
ative information, on the use of alternative lipid-lowering These data show the importance of achieving good graft
agents, specifically fibrates and nicotinic acid derivatives, function in RTR. The available data support the use of low-
in transplantation. At present, their use is not encouraged dose tacrolimus in combination with mycophenolic acid/
by guidelines, and any use, particularly as add-on therapy, mycophenolate and corticosteroids (with antiinterleukin-2
should be carefully monitored.108 receptor antibodies) as the most effective primary immuno-
suppressive strategy to produce optimal graft function. The
SYMPHONY study116 compared cyclosporine and tacroli-
Renal Function mus-based immunosuppressive regimens with sirolimus as
an adjunct agent. Low-dose tacrolimus (with a target blood
In the general population, renal dysfunction (CKD) is an level of 4–7 ng/mL) in combination with mycophenolate
established risk factor for CV disease. Although statisti- mofetil and corticosteroids was the best tolerated and most
cally an independent risk factor, CKD is associated with effective regimen, lending strong support to the established
dyslipidemia and hypertension. The “independent” effect of pattern of use in the clinical community. Moreover, the
renal dysfunction may reflect the presence of, as yet poorly achieved level of renal function was best in this group, with
defined “uremic toxins” or factors known to be associated a mean eGFR of 65.4 mL/min compared with 56.7 to 59.4
with renal impairment, such as hyperphosphatemia or ele- mL/min in the other groups.116 These outcomes persisted 3
vated FGF23.114,115 These same factors are likely to play a years after transplantation,118 albeit with lesser differences.
role in the pathophysiology of CVD in RTR with suboptimal CNI minimization, for example, by switching to an mTOR
renal function. Until recently, renal function had not been inhibitor (sirolimus or everolimus) has been examined in a
reported routinely as an outcome of trials of immunosup- variety of trials, incorporating a switch from CNI-based to
pression in renal transplantation, where the primary out- mTOR inhibitor-based therapy at various time points from
come is limited to the incidence of acute rejection and graft 7 weeks to years after transplantation.119–123 Although this
and patient survival. Recent major trials have reported strategy has been associated with an increased risk of acute
mean (or other summary measures of) serum creatinine rejection and the side effects limit the general applicability,
levels (or eGFR). This neglects the fact that mean levels are renal function is generally improved after early protocol-
of relevance to populations and not to individuals,116 and driven switching.
does not include proteinuria or other renal factors associ- More recently, the trials of belatacept, a costimu-
ated with poor graft outcomes. Improved reporting of renal lation blocker, in place of CNI in standard recipients
506 Kidney Transplantation: Principles and Practice

(BENEFIT)124,125 or recipients at risk of suboptimal graft development of NODAT. NODAT is reported in 3% to 50%
function (BENEFIT-EXT)124,125 have shown a similar pat- of RTRs. It is more common in older patients, in those who
tern of increased risk of acute rejection, but better graft are overweight or who gain weight,135 those of African
function, associated with reduced incidence of dyslipidemia or Asian origin, or who have a history of stress-induced
and hypertension. In the post hoc analyses of the ALERT diabetes (associated with surgery, use of corticosteroids,
study, renal function was one of the main determinants of or pregnancy).136,137 NODAT occurs against the back-
CV risk, regardless of whether an arbitrary cut-off was used ground development of age-related type 2 diabetes in the
(serum creatinine 200 μmol/L) or renal function was used population of patients with ESRD awaiting transplanta-
as a continuous variable.36,37 Patients whose grafts failed tion, and may be viewed as accelerating that process by
were at highest risk of CVEs. When risk of specific CVEs was increasing insulin resistance and, to a lesser extent, reduc-
analyzed, renal dysfunction was a stronger risk factor for ing insulin secretion.30,138 NODAT is most likely to occur
cardiac death than were nonfatal atheromatous coronary within the first 3 months of transplantation, occurring de
events,27,36,37 the latter being more dependent on lipid novo or as a manifestation of worsened preexisting disease
levels. (Fig. 30.8).136,139
Several other analyses lend support to the effect of graft The main contributory factor is thought to be the use
function on CVEs after transplantation.28,37,126 These of corticosteroids, which cause insulin resistance and
highlight the underlying importance of rejection episodes, are associated with the development of diabetes in other
cytomegalovirus infection, BK virus nephropathy, and patient groups.136,137,140 Observational data suggest that
drug-induced nephrotoxicity, which contribute to graft minimization of steroid dose reduces the rate of NODAT and
dysfunction and may show association with CVD in uni- may reverse diabetes and restore insulin sensitivity,46,141
variate analyses.20,21,27 Strategies to limit the effect of graft although randomized trials of early corticosteroid with-
dysfunction on CV risk will necessarily address the underly- drawal do not support this assumption. Woodle et al.110
ing factors, and are thus the strategies we employ to maxi- described no change in the rate of NODAT in RTRs with
mize graft survival and to minimize rejection. early corticosteroid withdrawal compared with chronic
corticosteroid treatment, but did see a trend toward reduc-
tion in insulin dose in those who developed NODAT. Pirsch
Smoking et al.142 conducted a randomized, double-blind study of
early corticosteroid withdrawal versus chronic corticoste-
In the general population cigarette smoking is highly asso- roid treatment (prednisolone 5 mg OD). Over 5 years of
ciated with the development of CVD. Studies in RTR have follow-up, there was no significant improvement in insulin
shown that smoking is associated with all-cause mortality, requirement or HbA1c.142 Such trials are difficult to inter-
with CVEs, with graft loss, and more rapid progression of pret due to “drop-in” therapy after randomization, and at
chronic transplant glomerulopathy.27,127–133 A system- present, minimization of corticosteroid use seems the best
atic review and meta-analysis investigated correlations strategy.
and outcomes associated with smoking in 73 studies (43 CNI can also contribute to the development of diabetes;
in kidney) and found significantly higher risk of cardio- tacrolimus is more diabetogenic than cyclosporine. This
vascular disease after transplant in those who continued reflects the role of tacrolimus-specific, intracellular FK-bind-
or resumed smoking after transplantation (odds ratio [OR] ing proteins on insulin secretion,136,143,144 and is probably
1.41; confidence interval (CI) 1.02–1.95).134 There can be the common mechanism through which mTOR inhibitors
substantial benefits from stopping smoking in advance of promote the development of NODAT.145 The DIRECT study
renal transplantation, and the risk of CV disease is reduced compared cyclosporine- and tacrolimus-based regimens
by around one-third in RTRs who stop smoking compared in de novo transplantation and showed conclusively, in a
with those who continue to smoke.19 Active smoking in study with NODAT as the primary endpoint, that tacroli-
live kidney donors is also associated with reduced recipi- mus-based therapy was associated with a higher incidence
ent survival,129 which has implications for pretransplant of NODAT, and of NODAT that required pharmacologic
counseling. Smoking increases risk of CV events by a haz- management. Moreover, the study confirmed that tacro-
ard ratio of 1.75.127 Smoking status is a good example limus use, compared with cyclosporine, was associated
of a risk factor that tends to be inaccurately reported by with reduced insulin secretion.146 Posthoc analysis of the
patients27 and, because it is a risk factor for “competing FAVORIT trial confirmed an association of tacrolimus
outcomes,” such as respiratory infection, malignancy, and therapy with greater risk of posttransplant diabetes.147
non-CV death,19,127 conventional survival analyses may Further studies support switch from tacrolimus to ciclospo-
underestimate the true effect of smoking on CV disease.18 rin in NODAT to improve fasting sugar levels and insulin
As yet, there are no studies of smoking cessation, although requirements, without increase in rejection episodes, but
this strategy is strongly endorsed in published guidelines.68 with weight gain in the ciclosporin group.148 There is some
evidence to support switching from CNI to sirolimus, which
may yield improvements in metabolic parameters without
New-Onset Diabetes After sacrificing patient or graft survival.149 Switching from CNI
Transplantation to everolimus cannot be similarly recommended because of
an increased risk of acute rejection.150
Diabetes in transplant recipients is increasingly preva- Despite these measures, however, many patients require
lent, relating to an increase in ESRD caused by type 2 dia- insulin or oral hypoglycemic agents. An alternative strat-
betes, which is now the leading cause of ESRD, and the egy is to avoid or minimize tacrolimus and/or steroids in
30 • Cardiovascular Disease in Renal Transplantation 507

35%

30%
Cyclo
FK506 25%

20%
Cumulative incidence

15%

10%

5%

–730 –365 0 365 730


–5%

–10%

–15%
Days before and after transplant

Days before transplant Days after transplant


Sample size
–730 –365 –1 1 365 730

Type of Cyclosporine 1776 3954 5867 6014 5521 2551


calcineurin
inhibitor Tacrolimus 471 911 1260 929 835 302

Note: The incremental incidence of diabetes for cyclosporine was 9.4% at 1 year and 8.4% at 2 years. The
incremental incidence of diabetes for tacrolimus was 15.4% at 1 year and 17.7% at 2 years.
Fig. 30.8 Data on the cumulative risk of diabetes in the 2 years before transplantation, and 2 years after transplantation, in renal transplant recipients
taking cyclosporine (Cyclo)- or tacrolimus (FK506)-based immunosuppression. Patients prescribed tacrolimus are at increased risk. (Data from Wood-
ward RS, Schnitzler MA, Baty J, et al. Incidence and cost of new onset diabetes mellitus among U.S. wait-listed and transplanted renal allograft recipients. Am
J Transplant 2003;3:590–8.)

patients at high risk for the development of posttransplant Guidelines have endorsed regular screening,157 with the
diabetes mellitus. This strategy may be of particular rel- use of oral glucose tolerance tests (if practical) and regular
evance in older patients where rejection is less of an issue fasting glucose measurements in transplant follow-up to
than in younger patients.140 However, such an approach identify patients with NODAT, and its precursor, impaired
has few advocates, due to the availability of management glucose tolerance. As described previously, recent trial data
strategies for diabetes and the failure to recognize the long- do not support early corticosteroid withdrawal alone, but
term consequences of NODAT. the combination of minimizing steroid and CNI dose, or
Data describing the long-term implications of NODAT switching from tacrolimus to cyclosporine or sirolimus is
for patients are contradictory. NODAT has been associated likely to be helpful.146,148,149
with LVH,151 and a two- to threefold increase in all-cause The management of NODAT is similar to that of type 2
mortality and CVEs.127,137,152,153 Indeed, data from our diabetes in the general population. The available guide-
own unit suggest that outcomes of patients with NODAT lines concur on the importance of monitoring for long-term
beyond 7 years after transplantation are comparable complications of diabetes and early involvement of dia-
between patients with ESRD due to diabetic nephropa- betic services.68,152 A proportion of patients who develop
thy (Fig. 30.9).137 Risk of CV-related death is reported to severe hyperglycemia, particularly early after transplan-
increase threefold in patients developing NODAT.30,131 tation, require insulin as primary therapy (although it
Studies to highlight the importance of NODAT have also may not be required in the long term, if immunosuppres-
shown that the risk to the patients far outweighs the risk sion is tailored appropriately), whereas those with milder
associated with acute rejection in the first year after trans- hyperglycemia may be managed with lifestyle interven-
plantation.152 Analyses conducted more recently appear tions and oral agents.152,158 Choice of oral agent may be
to show no significant effect of NODAT on CV events or different in NODAT than in type 2 diabetes. Metformin is
death.154–156 These contradictory findings may relate to generally not recommended as a first-line agent in treat-
greater awareness and monitoring of NODAT in the cur- ment of diabetes in RTRs158 because of concerns about the
rent era,154 thus strategies to limit the incidence and effect gastrointestinal side effects, particularly when considered
of NODAT remain a major target in the prevention of CVD. in combination with particular immunosuppressive agents
508 Kidney Transplantation: Principles and Practice

1.0 fracture risk, but in small groups of RTRs with NODAT


or impaired glucose tolerance it appears to be safe.162–164
Glinides appear to be safe in short-term studies in RTRs,
although they have no demonstrable effect on long-term
0.8
CV risk.158

0.6
Obesity and the Metabolic
Syndrome
The metabolic syndrome is defined as the presence of three
0.4 of the following: hyperglycemia, hypertriglyceridemia, low
HDL cholesterol, hypertension, and high body mass index.
Diabetes
As one would predict, given the high prevalence of diabetes
0.2 Diabetic nephropathy and the individual components, the metabolic syndrome is
No DM common in RTR.31,165–168 The presence of metabolic syn-
PTDM drome is associated with the degree of suppression of the
hypothalamic-pituitary axis, which is, in turn, associated
0 with chronic prednisolone treatment after renal transplan-
0 10 20 tation.169 Data from the PORT study31 report a prevalence
Years of 40% in the second year after transplantation and 35% in
Fig. 30.9 Long-term survival of renal transplant recipients without the fourth year after transplantation. Studies have demon-
diabetes ( ), with diabetes at the time of transplantation (diabetic strated an increased risk of developing diabetes in patients
nephropathy, ), and those who developed posttransplant with the metabolic syndrome without diabetes, and an
diabetes mellitus (PTDM, ), demonstrating an increased car- increased risk of CVD, acute rejection, and graft loss, inde-
diovascular risk, matching those with preexisting diabetes, in patients
with PTDM. (Data from Revanur VK, Jardine AG, Kingsmore DB, et al. Influ- pendent of NODAT.31,165–167,170–172
ence of diabetes mellitus on patient and graft survival in recipients of kid- Obesity is increasingly common, reflecting the trends
ney transplantation. Clin Transplant 2001;15:89–94.) in the wider population.173 Morbid obesity is associated
with a higher incidence of complications around the time
of transplant surgery.167,174 However, the relationship
(e.g., mycophenolate mofetil). Sulfonylureas are effec- with long-term outcomes is more complex. There is a non-
tive in reducing HbA1c in the first 12 months after renal linear relationship between weight and all-cause mortal-
transplant, but at the expense of weight gain, reduction in ity,170 reflecting the effect of comorbidity, whereas obese
B cell function and increased risk of hypoglycemia.158 Tri- patients exhibit a progressive increase in CV risk. Some
als of SGLT2 inhibitors, which promote glucose excretion centers have employed weight reduction surgery175,176 as
via renal tubules and encourage weight loss over the lon- part of the workup for inclusion onto the waiting list for
ger term, have yielded promising results in type 2 diabetes. transplantation, although there is no current evidence
The EMPA-REG OUTCOME trial assessed the effect of empa- that pretransplant weight loss is associated with survival
gliflozin (10 or 25 mg daily) versus placebo on cardiovas- advantage.177
cular events and mortality in patients with type 2 diabetes
and GFR >30 mL/min.159 This trial showed a significant
reduction in CV events and all-cause mortality in patients
Other Risk Factors and
treated with empagliflozin compared with placebo. It is not Interventions
clear whether these effects will translate to have the same
benefit on RTRs, and SGLT2 inhibitors have not yet been There are other strategies for the prevention of CV risk in
specifically tested in RTRs, but the glucose-lowering effect the general population. Exercise and weight loss are rec-
of SGLT2 inhibitors appears less pronounced in those with ommended in guidelines regarding posttransplant man-
GFR 30.60 mL/min compared with patients with normal agement.68,99 Patients with ESRD have very poor exercise
renal function,158 and there is an increased propensity to capacity, which should improve after transplantation.
urinary and genital fungal infections in recipients of empa- However, exercise and improvement in objective measures
gliflozin,159 which may have significance in immunosup- of exercise capacity (such as Vo2max) may require active
pressed individuals. encouragement in transplant recipients who often find
Gliptins are safe and effective in the treatment of NODAT barriers to engagement in exercise, both physical and psy-
in RTRs.160–162 The VINODAT study161 was a randomized chological, which are related to protracted disability and ill
double-blind placebo-controlled trial of vildagliptin (50 mg health. Several studies have shown the association of poor
once daily) versus placebo in stable RTRs 6 months or more exercise capacity with adverse CV outcomes after transplan-
from transplant. There was a dramatic improvement in tation,178–180 although a systematic review of the effects of
2-hour glucose and HbA1c in those patients treated with exercise on outcome after kidney transplantation showed
vildagliptin, with limited side effects and no deleterious minimal or contradictory effects of exercise on transplant
effects on immunosuppression or transplant function.161 function or the metabolic syndrome.181
Pioglitazone comes with the increased risk of weight A number of other factors have been associated with CVD
gain, fluid retention, bladder cancer, and increased in the general population, although they have not been
30 • Cardiovascular Disease in Renal Transplantation 509

assessed fully in RTRs. These include C-reactive protein, at screening.196 Coronary angiography, with or without
inflammation, oxidative stress, hyperhomocysteinemia, intervention, is typically reserved for those with positive
and elevated serum phosphate. However, recent studies noninvasive tests or those who are at particularly high
have shown an association of a wide variety of biomarkers risk.190–192
with CV and related outcomes. These include circulating
inhibitors of nitric oxide,38,39 elevated phosphate, FGF23,
osteoprotegerin,81,114,115,182,183 oxidative stress,184,185
Intervention and Secondary
endothelin,186 paraoxonase,187 and hyperuricemia.188 Prevention
Whether these are independent markers and therapeutic
targets remains to be resolved. Functional factors, such Just as the likelihood of intervention after screening is low,
as depression, have also been associated with graft loss there is concern that RTRs are less likely to receive interven-
and mortality after transplantation and are potentially tion for an MI (thrombolysis or primary revascularization)
remediable.189 or secondary preventions (high-dose statin therapy, anti-
platelet agents, plus blockade of the renin–angiotensin sys-
tem) proven in the general population. This is particularly
Screening true in patients with advanced CKD. However, beyond the
early postoperative period, the general consensus, endorsed
Clearly, with CVD in RTR being a common cause of in guidelines, is that RTRs should receive the same inter-
morbidity, mortality, and graft loss posttransplant, and vention and management as the general population.195
with considerations relating to equitable distribution Data in the population with CKD do suggest that there is
of organs, interest has focused on screening patients for increasing use of primary revascularization in this popula-
CVD before transplantation. The aims of this approach tion, albeit lagging behind the general population. It may
are to identify patients for whom the risk of transplan- be prudent to delay the introduction of new treatments in
tation outweighs the risks associated with maintenance high-risk populations, such as RTRs at higher risk of com-
dialysis.190–192 In reality, there are very few patients for plications, and thus the reported pattern of introduction
whom successful transplantation would not offer a sur- probably reflects cautious practice. The data in Fig. 30.10,
vival advantage,14 and the decision on whether to list collated by Henry and Herzog197 in the US, show that RTRs
someone for transplantation is largely subjective.190 CV have a substantial risk of mortality after coronary interven-
screening in some form is performed in most units with tions, but it is a risk that is considerably better than that of
the aim of identifying CAD that can be corrected before patients receiving maintenance dialysis. The PORT study,32
transplantation.190–192 Although this seems like a laud- which examined the use of CV risk medications, suggests
able aim and one which may benefit patients regardless of that the use of risk factor medications (see Fig. 30.6), includ-
whether they are ultimately transplanted, in the UK car- ing those used for secondary prevention, has increased year
diologists have become reluctant to recommend coronary by year, with the pattern of adoption and use following that
revascularization in asymptomatic patients in light of in the general population. A similar pattern of increasing
the COURAGE trial, which showed no benefit over medi- use has been reported by Lentine and colleagues198 for sec-
cal therapy.193 Moreover, the high prevalence of calcified ondary prevention after myocardial infarction and in other
lesions makes coronary revascularization challenging.194 populations.67
In our own center, the delay to waitlisting inherent in a Overall, the message for RTR is that patients should not
screening program did not benefit patients and those with be denied revascularization and secondary prevention for
coronary disease were less likely to be listed and unlikely CAD. Although there does appear to be an increase in mor-
to undergo coronary intervention. One consistent obser- tality associated with revascularization procedures (spe-
vation in screening programs is that the intervention rate cifically bypass surgery and vein grafting) compared with
is low—around 5% of those who are screened ultimately nontransplant recipients, the mortality risk is much less
undergo revascularization.190 than that associated with patients with ESRD receiving
Thus there is little to support routine screening of asymp- dialysis.
tomatic patients based on the available literature, and a
general view that a trial of screening is required but unlikely
to be performed.195 Other Cardiovascular Conditions:
Screening typically involves echocardiography or
some assessment of left ventricular structure and func-
Valvular Disease, Arrhythmia, and
tion, together with a “stress test” to look for reversible Stroke
ischemia.191,192 Conventional treadmill exercise tests are
difficult to interpret due to a high prevalence of electro- In this chapter, we have focused on CHD and the com-
cardiographic abnormalities in patients receiving mainte- mon patterns of CVD and risk that affect RTRs. However,
nance dialysis and poor exercise capacity. Pharmacologic RTRs are also at increased risk of the common valvular
stress tests are commonly performed, with echocardiog- conditions and arrhythmias. The risk of atrial fibrillation,
raphy or nuclear imaging, to look for regions of revers- a common cause of stroke in the general population, is
ible ischemia.191,192 Even mild reversible abnormalities increased in CKD and RTRs, and associated with signifi-
on perfusion imaging are predictive of mortality in RTRs; cantly elevated risk of stroke, graft loss, and mortality post
those with normal perfusion imaging are more likely to transplant.199,200 There are no clinical trials of anticoag-
receive a renal transplant than those with any abnormality ulation for atrial fibrillation in RTRs, and observational
510 Kidney Transplantation: Principles and Practice

Survival of ESRD patients with cardiovascular diagnosis and procedures

CHF CVA/TIA PAD Cardiac arrest


1.0

0.8

0.6

0.4
Hemodialysis
Peritoneal dialysis
Probability of survival

0.2
Transplant

0.0
Coronary revascularization: Coronary revascularization:
AMI PCI surgical ICD/CRT-D
1.0

0.8

0.6

0.4

0.2

0.0
0 3 6 9 12 0 3 6 9 12 0 3 6 9 12 0 3 6 9 12
Months after diagnosis or intervention
Fig. 30.10 Mortality for individual cardiac interventions for patients with end-stage renal disease (ESRD), subdivided by modality. In all cases, patients
with a transplant have better outcomes. AMI, acute myocardial infarction; CHF, congestive heart failure; CVA/TIA, cerebrovascular accident/transient
ischemic attack; ICD/CRT-D, implantable cardiodefibrillator/cardiac resynchronization device; PAD, peripheral arterial disease; PCI, percutaneous coro-
nary intervention. (Data from Henry TD, Herzog CA. Bad kidneys are bad for the heart. Catheter Cardiovasc Interv 2012;80:358–60.)

data of warfarin treatment in atrial fibrillation do not progresses more rapidly in parallel with the development
describe significant reduction in stroke, graft failure, or of vascular calcification.56,203 The incidence of infective
death.200,201 Direct oral anticoagulant (DOAC) agents are endocarditis is increased, reflecting the higher prevalence
of interest in the general population because of the lack of of valvular abnormalities and concomitant immunosup-
requirement for monitoring, similar efficacy, and favorable pression.204 The management of valvular disease and endo-
side effect profile compared with warfarin. In the acute carditis in RTRs is the same as in the general population.
posttransplant setting, RTRs are prone to fluctuations in Again, the risk associated with valve replacement is higher
renal function; acute illness, including infection, and rejec- in RTRs but better than that of patients with ESRD receiving
tion requiring investigation, including renal biopsy. Under maintenance dialysis.56,203
these circumstances, the inability to monitor and reverse
DOAC action may be disadvantageous. There are no data
describing outcomes of RTRs who were prescribed DOAC Predicting Cardiovascular Risk
for nonvalvular atrial fibrillation, nor are there direct con-
traindications, but use of these medications would be bet- In the general population we predict CV risk based on estab-
ter postponed until stable graft function and maintenance lished factors and calculations. The most commonly used
immunosuppression has been achieved. Rivaroxaban or is the Framingham risk factor calculator, or its deriva-
apixaban are the least likely to interact with cyclosporine tives. This has not proved appropriate for patients with
or tacrolimus,202 and thus are likely to be the preferred CKD, including transplant recipients,20,21,205 reflecting the
choices in RTRs. Until prospective confirmation can be atypical nature of CVD. Using data from the ALERT study,
obtained, it is prudent to offer RTRs the same treatment we have derived a CV risk factor calculator from data in
proven to reduce stroke and mortality seen in other pop- RTR.117,206 There are limitations, notably, that all patients
ulations, though anticoagulation should be approached were treated with cyclosporine, and it has yet to be inde-
with caution in those RTRs with poor graft function, as for pendently verified. However, it performed well on internal
those with CKD stage 5 or receiving other forms of renal validation. The factors included in the calculator are: age,
replacement therapy. diabetes, LDL cholesterol, serum creatinine, smoking sta-
Data from our own center demonstrate an increased risk tus, preexisting CHD, and the number of transplants. The
of stroke in RTRs and a greater risk of mortality from stroke details of the calculator are shown in Fig. 30.11. Prospec-
compared with the general population,200 despite similar tive collection of risk factor data and validation within exist-
risk factors, case mix, and management of stroke.200 Cal- ing data sets should allow the derivation of more accurate
cific aortic stenosis is common in patients with ESRD and calculators for transplant recipients in the future.
30 • Cardiovascular Disease in Renal Transplantation 511

7-year risk for 7-year risk for


CVD death rates/100 patient years according
Calculator type MACE death to time from transplant

Diabetes mellitus yes no includes PTDM 4 1980–84 1985–89 1990–94


RR 0.42
LDL-cholesterol mmol/L 1995–99 2000–04 2005–06
3

Death rate
Current smoker yes no
RR 0.35
RR 0.32
Previous smoker yes no 2
Coronary heart disease yes no
1
Number of transplants

Creatinine µmol/L mg/dL 0


Age years 0-1 1-4 5-9 10-14
7-year risk for MACE Time posttransplant (years)
MACE - major adverse cardiac event Fig. 30.12 Changing pattern of cardiovascular disease (CVD) survival
PTDM - post transplant diabetes mellitus in the ANZDATA registry by era. Cardiovascular survival can be seen
The use of this calculator requires clinical judgement and depends on context to have improved over the eras studied. RR, relative risk. (Data from
Pilmore H, Dent H, Chang S, et al. Reduction in cardiovascular death after
Fig. 30.11 Cardiovascular risk calculator for renal transplant recipients.
kidney transplantation. Transplantation 2010;89:851–7.)
(Data from Soveri I, Holme I, Holdaas H, et al. A cardiovascular risk calcu-
lator for renal transplant recipients. Transplantation 2011;94:57–62. Avail-
able online at: http://www.anst.uu.se/insov254/calculator/.)
prevention of other aspects of CV disease in this popula-
tion, and much still needs to be done. Blood pressure targets
Trial Endpoints and improvements in guidance on RTR antihypertensive
therapy are still required. There have been major changes
Clinical trials of immunosuppression in transplantation in immunosuppression over the past 20 years, and we now
have, for the most part, concentrated on graft-specific have a spectrum of agents with differing immunosuppres-
endpoints and the current favored US Food and Drug sive actions and side-effect profiles. This may permit tailor-
Administration endpoint of acute rejection, graft loss, ing of immunosuppression to minimize the risk of long-term
death, or loss to follow-up, and to short timescales of up complications, including CV disease risk; but at present, the
to 12 months. Although these are dictated by the need clinical community continues to focus on rejection and
to develop drugs in a timely manner, they fail to address immunologic risk when selecting immunosuppressive strat-
long-term patient-specific issues, and ignore observa- egies. A future change in the design and conduct of clinical
tions that, for example, the development of NODAT has a trials in transplantation to recognize the importance of the
greater negative effect on patient survival than an acute major “endpoints” for patients—CVD, malignancy, and
rejection episode.117,152 infection, as well as rejection and graft survival—might
One way to address the importance of CV disease in trans- focus attention on the importance of immunosuppressive
plantation would be to include CVEs in trial design, perhaps choice.
as a part of a patient-specific composite endpoint. Such a Regardless, and despite the increasing age of transplant
strategy would probably require larger trials, with longer recipients, their increasing comorbidity, and other changes
follow-up, that might limit the applicability of this design. to the transplant population that may have been predicted
An alternative approach is to model the expected CV effects to worsen outcome, there has been a progressive reduc-
of a particular immunosuppressive strategy, based on data tion in CVD in the transplant population (Fig. 30.12).208
available at earlier time points. For example, two recent This follows the pattern of CVD across the developed world,
studies have attempted to predict long-term outcomes reflecting improved understanding of CVD and lifestyle
based on short-term data.117,207 Although the validity of improvements, and may reflect the improved management
this approach remains to be established, reporting of accu- of RTR in general, together with the progressive uptake of
rate risk factor data: blood pressure, lipid levels, renal func- cardioprotective mechanisms. It is an encouraging obser-
tion, and NODAT, should be a minimum requirement of vation and provides a positive point on which to finish.
trials in transplantation, and the inclusion of informative, However, the absolute rate of CVD remains high and must
novel biomarkers should be considered. remain a priority for those involved in renal transplanta-
tion. Given the accrued burden of risk that many patients
have at the time of transplantation, the most important
Conclusion message is the need to consider the prevention of posttrans-
plant CVD when one first encounters a patient with pro-
In writing this chapter we reviewed previous editions of this gressive primary renal disease at the renal clinic.
textbook, specifically the chapter by professor Tony Raine
in the fourth edition, which is now more than 20 years References
old. His review focused on atherosclerotic CV disease and 1. Baigent C, Burbury K, Wheeler D. Premature cardiovascular disease
highlighted the need for trials of lipid-lowering therapy, in chronic renal failure. Lancet 2000;356:147–52.
that have since been performed. These trials have shown 2. Foley RN, Parfrey PS, Sarnak MJ. Clinical epidemiology of car-
diovascular disease in chronic renal disease. Am J Kidney Dis
that statins are effective in the prevention of atherosclerotic 1998;32:S112–9.
events but that atherosclerotic coronary disease is only 3. Jardine AG, Gaston RS, Fellstrom BC, Holdaas H. Prevention of car-
one component of the complex accelerated CV disease in diovascular disease in adult recipients of kidney transplants. Lancet
this population. We have made much less progress on the 2011;378:1419–27.
512 Kidney Transplantation: Principles and Practice

4. National Institutes of Health. 2016 USRDS annual data report: epi- 30. Hjelmesaeth J, et al. The impact of early-diagnosed new-onset post-
demiology of kidney disease in the United States. United States Renal transplantation diabetes mellitus on survival and major cardiac
Data System 2016. events. Kidney Int 2006;69:588–95.
5. UK Renal Registry. 18th Annual report of the renal association. 31. Israni AK, et al. Predicting coronary heart disease after kidney trans-
Nephron 2016:132. plantation: Patient Outcomes in Renal Transplantation (PORT)
6. Sarnak MJ, Levey AS, Schoolwerth AC, et al. Kidney disease as a risk study. Am J Transplant 2010;10:338–53.
factor for development of cardiovascular disease: a statement from 32. Pilmore HL, Skeans MA, Snyder JJ, Israni AK, Kasiske BL. Cardiovas-
the american heart association councils on kidney in cardiovascular cular disease medications after renal transplantation: results from
disease, high blood pressure research, clinical cardiology, and epide- the patient outcomes in renal transplantation study. Transplanta-
miology and prevention. Circulation 2003;108:2154–69. tion 2011;91:542–51.
7. Chonchol M, Kjekshus J, Pedersen TR, Lindenfeld J. Simvastatin for 33. Soveri I, et al. Renal transplant dysfunction: importance quantified
secondary prevention of all-cause mortality and major coronary in comparison with traditional risk factors for cardiovascular disease
events in patients with mild chronic renal insufficiency. Am J Kidney and mortality. Nephrol Dial Transplant 2006;21:2282–9.
Dis 2007;49:373. 34. Abbott KC, et al. Hospitalized congestive heart failure after renal
8. Fellstrom BC, Jardine AG, Schmieder, et al. Rosuvastatin and cardio- transplantation in the united states. Ann Epidemiol 2002;12:
vascular events in patients undergoing hemodialysis. N Engl J Med 115–22.
2009;360:1395–407. 35. Rigatto C, et al. Congestive heart failure in renal transplant recipi-
9. Sacks FM, Pfeffer MA, Moye LA, et al. The effect of pravastatin on ents: risk factors, outcomes, and relationship with ischemic heart
coronary events after myocardial infarction in patients with average disease. J Am Soc Nephrol 2002;13:1084–90.
cholesterol levels. N Engl J Med 1996;335:1001–9. 36. Fellström B, et al. Risk factors for reaching renal endpoints in the
10. Scandinavian Simvastatin Survival Group. Randomised trial of assessment of Lescol in renal transplantation (ALERT) trial. Trans-
cholesterol lowering in 4444 patients with coronary heart disease. plantation 2005;79:205–12.
Lancet 1994;344:1383–9. 37. Fellström B, et al. Renal dysfunction is a strong and independent risk
11. Wanner C, Krane V, Marz W, et al. Atorvastatin in patients with factor for mortality and cardiovascular complications in renal trans-
type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med plantation. Am J Transplant 2005;5:1986–91.
2005;353:238–47. 38. Abedini S, et al. Inflammation in renal transplantation. Clin J Am
12. Foley RN, et al. The prognostic importance of left ventricular geome- Soc Nephrol 2009;4:1246–54.
try in uremic cardiomyopathy. J Am Soc Nephrol 1995;5:2024–31. 39. Abedini S, et al. Asymmetrical dimethylarginine is associated with
13. Mark PB, et al. Redefinition of uremic cardiomyopathy by con- renal and cardiovascular outcomes and all-cause mortality in renal
trast-enhanced cardiac magnetic resonance imaging. Kidney Int transplant recipients. Kidney Int 2010;77:44–50.
2006;69:1839–45. 40. Mangray M, Vella JP. Hypertension after kidney transplant. Am J
14. Wolfe RA, Ashby VB, Milford EL, et al. Comparison of mortality Kidney Dis 2011;57:331–41.
in all patients on dialysis, patients on dialysis awaiting transplan- 41. Ponticelli C, Cucchiari D, Graziani G. Hypertension in kidney trans-
tation, and recipients of a first cadaveric transplant. N Engl J Med plant recipients. Transpl Int 2011;24:523–33.
1999;341:1725–30. 42. Opelz G, Dohler B. Improved long-term outcomes after renal trans-
15. Holdaas H, et al. Effect of fluvastatin on acute renal allograft rejec- plantation associated with blood pressure control. Am J Transpl
tion: a randomized multicenter trial. Kidney Int 2001;60:1990–7. 2005;5:2725–31.
16. Lam NN, et al. The risk of cardiovascular disease is not increasing 43. Opelz G, Wujciak TRE. Association of chronic kidney graft failure
over time despite aging and higher comorbidity burden of kidney with recipient blood pressure. Kidney Int 1998;53:217–22.
transplant recipients. Transplantation 2016;0:1–9. 44. Becker G, et al. KDIGO clinical practice guideline for the man-
17. Australia and New Zealand Dialysis and Transplant Registry. agement of blood pressure in chronic kidney disease. Kidney Int
AccNZDATA Registry 39th Report. Chapter 8. 2017. 2012;2(Suppl):337–414.
18. Holme I, Fellström BC, Jardine AG, Hartmann A, Holdaas H. Model 45. Ramesh Prasad GV. Ambulatory blood pressure monitoring in solid
comparisons of competing risk and recurrent events for graft failure organ transplantation. Clin Transplant 2012;26:185–91.
in renal transplant recipients. Clin J Am Soc Nephrol 2013;8:241–7. 46. Haydar AA, et al. Insights from ambulatory blood pressure monitor-
19. Opelz G, Dohler B. Influence of current and previous smoking on ing: diagnosis of hypertension and diurnal blood pressure in renal
cancer and mortality after kidney transplantation. Transplantation transplant recipients. Transplantation 2004;77:849–53.
2016;100:227–32. 47. Agena F, et al. Home blood pressure (bp) monitoring in kidney
20. Kasiske BL, Chakkera HA, Roel J. Explained and unexplained isch- transplant recipients is more adequate to monitor bp than office bp.
emic heart disease risk after renal transplantation. J Am Soc Nephrol Nephrol Dial Transplant 2011;26:3745–9.
2000;11:1735–43. 48. Ibernon M, et al. Reverse dipper pattern of blood pressure at
21. Kasiske BL, et al. Cardiovascular disease after renal transplantation. 3 months is associated with inflammation and outcome after
J Am Soc Nephrol 1996;7:158–65. renal transplantation. Nephrol Dial Transplant 2012;27:
22. Liu Y, et al. Association between cholesterol level and mortality 2089–95.
in dialysis patients: role of inflammation and malnutrition. JAMA 49. Pascual J, Perez-Saez MJ, Mir M, Crespo M. Chronic renal allograft
2004;291:451–9. injury: early detection, accurate diagnosis and management. Trans-
23. Vanrenterghem YE. Risk factors for cardiovascular events after suc- plant Rev (Orlando) 2012;26:280–90.
cessful renal transplantation. Transplantation 2008;85:209–16. 50. Patel RK, et al. Determinants of left ventricular mass and hyper-
24. Bostom AG, et al. Homocysteine-lowering and cardiovascular dis- trophy in hemodialysis patients assessed by cardiac magnetic reso-
ease outcomes in kidney transplant recipients: primary results from nance imaging. Clin J Am Soc Nephrol 2009;4:1477–83.
the folic acid for vascular outcome reduction in transplantation trial. 51. Shamseddin MK, Knoll GA. Posttransplantation proteinuria: an
Circulation 2011;123:1763–70. approach to diagnosis and management. Clin J Am Soc Nephrol
25. Holdaas H, et al. Long-term cardiac outcomes in renal transplant 2011;6:1786–93.
recipients receiving fluvastatin: the ALERT extension study. Am J 52. Stewart GA, et al. Electrocardiographic abnormalities and uremic
Transplant 2005;5:2929–36. cardiomyopathy. Kidney Int 2005;67:217–26.
26. Holdaas H, et al. Effect of fluvastatin on cardiac outcomes in renal 53. Patel RK, et al. Microvolt T-wave alternans in end-stage renal dis-
transplant recipients: a multicentre, randomised, placebo-controlled ease patients: associations with uremic cardiomyopathy. Clin J Am
trial. Lancet 2003;361:2024–31. Soc Nephrol 2011;6:519–27.
27. Jardine AG, et al. Cardiovascular risk and renal transplantation: post 54. Bakri K, Goldsmith DJA. Accelerated progression of calcific aortic
hoc analyses of the Assessment of Lescol in Renal Transplantation stenosis in dialysis patients: what we still need to learn. Nephron Clin
(ALERT) study. Am J Kidney Dis 2005;46:529–36. Pract 2003;94:c27–8.
28. Weiner DE, et al. Kidney function and risk of cardiovascular disease 55. Vaidya OU, et al. Effect of renal transplantation for chronic renal dis-
and mortality in kidney transplant recipients: the favorit trial. Am J ease on left ventricular mass. Am J Cardiol 2012. Available online at:
Transplant 2012;12:l2437–45. https://doi.org/10.1016/j.amjcard.2012.02.067.
29. Investigators Gissi-HF, et al. Effect of rosuvastatin in patients with 56. Patel RK, et al. Renal transplantation is not associated with regres-
chronic heart failure (the GISSI-HF trial): a randomised, double- sion of left ventricular hypertrophy: a magnetic resonance study.
blind, placebo-controlled trial. Lancet 2008;372:1231–9. Clin J Am Soc Nephrol 2008;3:1807–11.
30 • Cardiovascular Disease in Renal Transplantation 513

57. McGregor E, Jardine A, Dargie H, et al. Echocardiographic abnor- 82. Stevens KK, et al. Deleterious effects of phosphate on vascular and
malities and survival following renal transplantation. Nephrol Dial endothelial function via disruption to the nitric oxide pathway.
Transplant 1998;13:1499–505. Nephrol Dial Transplant 2016:1–11. Available online at: https://
58. Midtvedt K, et al. Reduction of left ventricular mass by lisinopril doi.org/10.1093/ndt/gfw252.
and nifedipine in hypertensive renal transplant recipients: a pro- 83. Seyahi N, et al. Progression of coronary artery calcification in renal
spective randomized double-blind study. Transplantation 2001;72: transplant recipients. Nephrol Dial Transplant 2012;27:2101–7.
107–11. 84. Karras A, et al. Reversal of arterial stiffness and maladaptative arte-
59. Paoletti E, Cannella G. Regression of left ventricular hypertrophy in rial remodeling after kidney transplantation. J Am Heart Assoc
kidney transplant recipients: the potential role for inhibition of mam- 2017;6:e006078.
malian target of rapamycin. Transpl Proc 2010;42:S41–3. 85. Poulikakos D, et al. Left ventricular hypertrophy and endothelial
60. Bachelet-Rousseau C, et al. Evolution of arterial stiffness after kidney dysfunction in chronic kidney disease. Eur Heart J Cardiovasc Imag-
transplantation. Nephrol Dial Transplant 2011;26:3386–91. ing 2014;15:56–61.
61. Morris S, McMurray J, Rodger R, et al. Endothelial dysfunction in 86. Cho I, et al. Aortic calcification is associated with arterial stiffening,
renal transplant recipients. Kidney Int 2000;57:1100–6. left ventricular hypertrophy, and diastolic dysfunction in elderly
62. Recio-Mayoral A, Banerjee D, Streather C, Kaski JC. Endothelial male patients with hypertension. J Hypertens 2015;33:1633–41.
dysfunction, inflammation and atherosclerosis in chronic kidney 87. Imanishi K, et al. Post-transplant renal function and cardiovascu-
disease: a cross-sectional study of predialysis, dialysis and kidney- lar events are closely associated with the aortic calcification index in
transplantation patients. Atherosclerosis 2011;216:446–51. renal transplant recipients. Transplant Proc 2014;46:484–8.
63. Walker BR. Glucocorticoids and cardiovascular disease. Eur J Endo- 88. DeLoach SS, Joffe MM, Mai X, Goral S, Rosas SE. Aortic calcifica-
crinol 2007;157:545–59. tion predicts cardiovascular events and all-cause mortality in renal
64. Calò LA, et al. Pathophysiology of post transplant hypertension in transplantation. Nephrol Dial Transplant 2009;24:1314–9.
kidney transplant: focus on calcineurin inhibitors induced oxidative 89. Yazbek DC, De Carvalho AB, Barros CS, Pestana JOM, Canziani MEF.
stress and renal sodium retention and implications with RhoA/Rho Effect of statins on the progression of coronary calcification in kidney
kinase pathway. Kidney Blood Press Res 2017:676–85. Available transplant recipients. PLoS ONE 2016;11:1–15.
online at: https://doi.org/10.1159/000483023. 90. Boxma PY, et al. Vitamin K intake and plasma desphospho-uncar-
65. Cross NB, Webster AC, Masson P, O’Connell PJ, Craig JC. Antihy- boxylated matrix gla-protein levels in kidney transplant recipients.
pertensives for kidney transplant recipients: systematic review and PLoS ONE 2012;7:e47991.
meta-analysis of randomized controlled trials. Transplantation 91. Keyzer CA, et al. Vitamin K status and mortality after kidney trans-
2009;88:7–18. plantation: a cohort study. Am J Kidney Dis 2015;65:474–83.
66. Gaston RS, et al. Use of cardioprotective medications in kidney trans- 92. Fulton RL, et al. Effect of vitamin K on vascular health and physical
plant recipients. Am J Transplant 2009;9:1811–5. function in older people with vascular disease: a randomised con-
67. Kidney Disease: Improving Global Outcomes (KDIGO) Transplant trolled trial. J Nutr Health Aging 2016;20:325–33.
Working Group. KDIGO clinical practice guideline for the care of 93. Braam L, et al. Beneficial effects of vitamins D and K on the elastic
kidney transplant recipients. Am J Transplant 2009;3(Suppl. 9): properties of the vessel wall in postmenopausal women: a follow-up
S1–155. study. Thromb Haemost 2004;91:373–80.
68. Philipp T, et al. Candesartan improves blood pressure control and 94. Kurnatowska I, et al. Effect of vitamin K2 on progression of ath-
reduces proteinuria in renal transplant recipients: results from erosclerosis and vascular calcification in nondialyzed patients
secret. Nephrol Dial Transplant 2010;25:967–76. with chronic kidney disease stages 3-5. Pol Arch Med Wewn
69. Knoll GA, et al. The Canadian ace-inhibitor trial to improve renal 2015;125:631–40.
outcomes and patient survival in kidney transplantation - study 95. Brandenburg VM, et al. Slower progress of aortic valve calcification
design. Nephrol Dial Transplant 2008;23:354–8. with vitamin K supplementation: results from a prospective inter-
70. Knoll GA, et al. Ramipril versus placebo in kidney transplant ventional proof-of-concept study. Circulation 2017;135:2081–3.
patients with proteinuria: a multicentre, double-blind, randomised 96. Knapen M, et al. Menaquinone-7 supplementation improves arte-
controlled trial. Lancet Diabetes Endocrinol 2016;4:318–26. rial stiffness in healthy postmenopausal women: a double-blind ran-
71. Paoletti E, et al. Effects of ace inhibitors on long-term outcome of domised clinical trial. Thromb Haemost 2015;113:1135–44.
renal transplant recipients: a randomized controlled trial. Trans- 97. Shea MK, et al. Vitamin K supplementation and progression of
plantation 2013;95:889–95. coronary artery calcium in older men and women. Am J Clin Nutr
72. Tutone VK, et al. Hypertension, antihypertensive agents and out- 2009;89:1799–807.
comes following renal transplantation. Clin Transpl 2005;19: 98. Heemann U, Abramowicz D, Spasovski G, Vanholder R. Endorse-
181–92. ment of the Kidney Disease Improving Global Outcomes (KDIGO)
73. Isakova T, et al. FGF23, PTH and phosphorus metabolism in the guidelines on kidney transplantation: a European Renal Best
chronic renal insufficiency cohort. Kidney Int 2011;79:1370–8. Practice (ERBP) position statement. Nephrol Dial Transplant
74. Prasad N, et al. FGF23 is associated with early post-transplant hypo- 2011;26:2099–106.
phosphataemia and normalizes faster than iPTH in living donor 99. Holdaas H, Kobashigawa JBF, et al. In: Ballantyne C, editor. Clinical
renal transplant recipients: a longitudinal follow-up study. Clin Kid- lipidology. Amsterdam: Elsevier; 2008. p. 486–99.
ney J 2016;9:669–76. 100. Vaziri ND. Dyslipidemia of chronic renal failure: the nature, mech-
75. Cianciolo G, Cozzolino M. FGF 23 in kidney transplant: the strange anisms and potential consequences. Am J Physiol Ren Physiol
case of doctor jekyll and mister hyde. Clin Kidney J 2016;9:665–8. 2006;290:F262–72.
76. Sánchez Fructuoso AI, et al. Serum level of fibroblast growth factor 101. Miller LW. Cardiovascular toxicities of immunosuppressive agents.
23 in maintenance renal transplant patients. Nephrol Dial Trans- Am J Transplant 2002;2:807–18.
plant 2012;27:4227–35. 102. Kasiske BL, et al. Mammalian target of rapamycin inhibitor dyslip-
77. Faul C, et al. FGF23 induces leftt ventricular hypertrophy. J Clin idemia in kidney transplant recipients. Am J Transplant 2008;8:
Invest 2011;121:4393–408. 1384–92.
78. Di Marco GS, et al. Treatment of established left ventricular hyper- 103. Holdaas H, et al. Beneficial effect of early initiation of lipid-lowering
trophy with fibroblast growth factor receptor blockade in an animal therapy following renal transplantation. Nephrol Dial Transplant
model of CKD. Nephrol Dial Transplant 2014;29:2028–35. 2005;20:974–80.
79. Baia LC, et al. Fibroblast growth factor 23 and cardiovascular 104. Jardine A, Holdaas H. Fluvastatin in combination with cyclosporin
mortality after kidney transplantation. Clin J Am Soc Nephrol in renal transplant recipients: a review of clinical and safety experi-
2013;8:1968–78. ence. J Clin Pharm Ther 1999;24:397–408.
80. Wolf M, et al. Elevated fibroblast growth factor 23 is a risk fac- 105. Baigent C, et al. The effects of lowering LDL cholesterol with simvas-
tor for kidney transplant loss and mortality. J Am Soc Nephrol tatin plus ezetimibe in patients with chronic kidney disease (Study of
2011;22:956–66. Heart and Renal Protection): a randomised placebo-controlled trial.
81. Scialla J, et al. Fibroblast growth factor 23 is not associated with Lancet 2011;377:2181–92.
and does not induce arterial calcification. Kidney Int 2013;83: 106. Cosio FG, et al. Patient survival after renal transplantation III: the
1159–68. effects of statins. Survival (Lond) 2002;40:638–43.
514 Kidney Transplantation: Principles and Practice

107. Kidney Disease Outcomes Quality Initiative Group. K/DOQI clinical 132. Khalil MAM, et al. Cigarette smoking and its hazards in kidney trans-
practice guidelines for managing dyslipidemias in chronic kidney plantation. Adv Med 2017:1–11.
disease. Am J Kidney Dis 2003;41. 133. Duerinckx N, et al. Correlates and outcomes of posttransplant
108. KDIGO. Clinical. Practice guideline for lipid management in chronic smoking in solid organ transplant recipients: a systematic lit-
kidney disease. Kidney Int 2013;3:182–9. erature review and meta-analysis. Transplantation 2016;100:
109. Woodle ES, et al. A prospective, randomized, double-blind, placebo- 2252–63.
controlled multicenter trial comparing early (7 day) corticosteroid 134. Kim Y, et al. Patients with persistent new-onset diabetes after trans-
cessation versus long-term, low-dose corticosteroid therapy. Ann plantation have greater weight gain after kidney transplantation.
Surg 2008;248:564–77. J Korean Med Sci 2013;28:1431–4.
110. Knight SR, Morris PJ. Steroid avoidance or withdrawal after renal 135. Woodward RS, et al. Incidence and cost of new onset diabetes mel-
transplantation increases the risk of acute rejection but decreases litus among U.S. wait-listed and transplanted renal allograft recipi-
cardiovascular risk: a meta-analysis. Transplantation 2010;89: ents. Am J Transplant 2003;3:590–8.
1–14. 136. Revanur VK, et al. Influence of diabetes mellitus on patient and graft
111. Rostaing L, et al. Conversion to tacrolimus once-daily from ciclo- survival in recipients of kidney transplantation. Clin Transplant
sporin in stable kidney transplant recipients: a multicenter study. 2001;15:89–94.
Transpl Int 2012;25:391–400. 137. Wauters RP, et al. Cardiovascular consequences of new-onset
112. Wissing KM, et al. Effect of atorvastatin therapy and conversion to hyperglycemia after kidney transplantation. Transplantation
tacrolimus on hypercholesterolemia and endothelial dysfunction 2012;94:377–82.
after renal transplantation. Transplantation 2006;82:771–8. 138. Cullen TJ, McCarthy MP, Lasarev MR, Barry JM, Stadler DD. Body
113. Mazzaferro S, et al. Distinct impact of vitamin D insufficiency on cal- mass index and the development of new-onset diabetes mellitus or
citriol levels in chronic renal failure and renal transplant patients: a the worsening of pre-existing diabetes mellitus in adult kidney trans-
role for FGF23. J Nephrol 2012;25(6):1108–18. Available online at: plant patients. J Ren Nutr 2014;24:116–22.
https://doi.org/10.5301/jn.5000102. 139. Joss NS, Staatz CE, Thomson AH, Jardine AG. Predictors of
114. Stevens KK, et al. Serum phosphate and outcome at one year new onset diabetes after renal transplantation. Clin Transplant
after deceased donor renal transplantation. Clin Transplant 2007;21(1):136–43.
2011;25:E199–204. 140. Serrano OK, et al. Rapid discontinuation of prednisone in kidney trans-
115. Ekberg H, et al. Reduced exposure to calcineurin inhibitors in renal plant recipients. Transplantation 2017;101(10):2590–8. Available
transplantation. N Engl J Med 2007;357:2562–75. online at: https://doi.org/10.1097/TP.0000000000001756.
116. Soveri I, et al. The external validation of the cardiovascular risk 141. Midtvedt K, et al. Insulin resistance after renal transplantation: the
equation for renal transplant recipients: applications to Benefit and effect of steroid dose reduction and withdrawal. J Am Soc Nephrol
Benefit-ext trials. Transplantation 2013;95:142–7. 2004;15:3233–9.
117. Ekberg H, et al. Calcineurin inhibitor minimization in the symphony 142. Pirsch JD, et al. New-onset diabetes after transplantation: results
study: observational results 3 years after transplantation. Am J from a double-blind early corticosteroid withdrawal trial. Am J
Transplant 2009;9:1876–85. Transplant 2015;15:1982–90.
118. Johnson RW, et al. Sirolimus allows early cyclosporine withdrawal 143. Hjelmesæth J, et al. The impact of short-term ciclosporin A treat-
in renal transplantation resulting in improved renal function and ment on insulin secretion and insulin sensitivity in man. Nephrol
lower blood pressure. Transplantation 2001;72:777–86. Dial Transplant 2007;22:1743–9.
119. Mjörnstedt L, et al. Improved renal function after early conversion 144. van Hooff JP, Christiaans MH, van Duijnhoven EM. Tacrolimus and
from a calcineurin inhibitor to everolimus: a randomized trial in kid- posttransplant diabetes mellitus in renal transplantation. Trans-
ney transplantation. Am J Transplant 2012;12:2744–53. plantation 2005;79:1465–9.
120. Holdaas H, et al. Conversion of long-term kidney transplant recipi- 145. Johnston O, Rose CL, Webster AC, Gill JS. Sirolimus is associated
ents from calcineurin inhibitor therapy to everolimus: a randomized, with new-onset diabetes in kidney transplant recipients. J Am Soc
multicenter, 24-month study. Transplantation 2011;92:410–8. Nephrol 2008;19:1411–8.
121. Budde K, et al. Everolimus-based, calcineurin-inhibitor-free regi- 146. Vincenti F, et al. Results of an international, randomized trial com-
men in recipients of de-novo kidney transplants: an open-label, ran- paring glucose metabolism disorders and outcome with cyclosporine
domised, controlled trial. Lancet 2011;377:837–47. versus tacrolimus. Am J Transpl 2007;7:1506–14.
122. Campistol JM, et al. Everolimus and long-term outcomes in renal 147. Weinrauch LA, et al. Does diabetes impact therapeutic immuno-
transplantation. Transplantation 2011;92:S3–26. modulation therapy decisions for kidney transplant recipients?
123. Larsen CP, et al. Belatacept-based regimens versus a cyclosporine Data from the Folic Acid for Vascular Outcome Reduction in
A-based regimen in kidney transplant recipients: 2-year results from Transplant (FAVORIT) trial. Int J Nephrol Renovasc Dis 2017;10:
the Benefit and benefit-ext studies. Transplantation 2010;90:1528– 233–42.
35. 148. Rathi M, et al. Conversion from tacrolimus to cyclosporine in
124. Vanrenterghem Y, et al. Belatacept-based regimens are associated patients with new-onset diabetes after renal transplant: an open-
with improved cardiovascular and metabolic risk factors compared label randomized prospective pilot study. Transplant Proc 2015;47:
with cyclosporine in kidney transplant recipients (BENEFIT and 1158–61.
BENEFIT-EXT studies). Transplantation 2011;91:976–83. 149. Veroux M, et al. Conversion to sirolimus therapy in kidney trans-
125. Meier-Kriesche HU, Baliga R, Kaplan B. Decreased renal function is plant recipients with new onset diabetes mellitus after transplanta-
a strong risk factor for cardiovascular death after renal transplanta- tion. Clin Dev Immunol 2013;2013:24–8.
tion. Transplantation 2003;75:1291–5. 150. Kälble F, et al. Switch to an everolimus-facilitated cyclosporine A
126. Seoane-Pillado MT, et al. Incidence of cardiovascular events and sparing immunosuppression improves glycemic control in selected
associated risk factors in kidney transplant patients: a competing kidney transplant recipients. Clin Transplant 2017:e13024. Avail-
risks survival analysis. BMC Cardiovasc Disord 2017;17:72. able online at: https://doi.org/10.1111/ctr.13024.
127. Van Laecke S, Nagler EV, Peeters P, Verbeke F, Van Biesen W. Former 151. Sezer S, et al. New-onset diabetes and glucose regulation are signifi-
smoking and early and long-term graft outcome in renal transplant cant determinants of left ventricular hypertrophy in renal transplant
recipients: a retrospective cohort study. Transpl Int 2017;30:187– recipients. J Diabetes Res 2015;2015.
95. 152. Wilkinson A, et al. Guidelines for the treatment and manage-
128. Underwood PW, et al. Cigarette smoking in living kidney donors: ment of new-onset diabetes after transplantation. Clin Transplant
donor and recipient outcomes. Clin Transplant 2014;28:419–22. 2008;19:291–8.
129. Hurst FP, et al. Effect of smoking on kidney transplant outcomes: 153. Cole EH, Johnston O, Rose CL, Gill JS. Impact of acute rejection and
analysis of the United States Renal Data System. Transplantation new-onset diabetes on long-term transplant graft and patient sur-
2011;92:1101–7. vival. Clin J Am Soc Nephrol 2008;3:814–21.
130. Kasiske BL, Klinger D. Cigarette smoking in renal transplant recipi- 154. Gaynor JJ, et al. Single-centre study of 628 adult, primary kid-
ents. J Am Soc Nephrol 2000;11:753–9. ney transplant recipients showing no unfavourable effect of
131. Agarwal PK, et al. Smoking is a risk factor for graft failure and mor- new-onset diabetes after transplant. Diabetologia 2015;58:
tality after renal transplantation. Am J Nephrol 2011;34:26–31. 334–45.
30 • Cardiovascular Disease in Renal Transplantation 515

155. Kuo HT, Sampaio MS, Vincenti F, Bunnapradist S. Associations of 181. Macdonald JH, Kirkman D, Jibani M. Kidney transplantation: a
pretransplant diabetes mellitus, new-onset diabetes after trans- systematic review of interventional and observational studies of
plant, and acute rejection with transplant outcomes: an analy- physical activity on intermediate outcomes. Adv Chronic Kidney Dis
sis of the organ procurement and transplant network/united 2009;16:482–500.
network for organ sharing (OPTN/UNOS) database. Am J Kidney Dis 182. Gungor O, et al. The relationships between serum stweak, FGF-23
2010;56:1127–39. levels, and carotid atherosclerosis in renal transplant patients. Ren
156. Cotovio P, et al. New-onset diabetes after transplantation: assess- Fail 2013;35:77–81.
ment of risk factors and clinical outcomes. Transpl Proc 2013;45: 183. Svensson M, et al. Osteoprotegerin as a predictor of renal and car-
1079–83. diovascular outcomes in renal transplant recipients: follow-up data
157. Bayer ND, et al. Association of metabolic syndrome with develop- from the alert study. Nephrol Dial Transplant 2012;27:2571–5.
ment of new-onset diabetes after transplantation. Transplantation 184. Turkmen K, et al. The relationship between oxidative stress, inflam-
2010;90:861–6. mation, and atherosclerosis in renal transplant and end-stage renal
158. Jenssen T, Hartmann A. Emerging treatments for post-transplanta- disease patients. Ren Fail 2012;34.
tion diabetes mellitus. Nat Rev Nephrol 2015;11:465–77. 185. Dahle DO, et al. Inflammation-associated graft loss in renal trans-
159. Zinman B, et al. Empagliflozin, cardiovascular outcomes, and mor- plant recipients. Nephrol Dial Transplant 2011;26:3756–61.
tality in type 2 diabetes. N Engl J Med 2015;373:2117–28. 186. Raina A, Horn ET, Benza RL. The pathophysiology of endothelin in
160. Boerner BP, Miles CD, Shivaswamy V. Efficacy and safety of sita- complications after solid organ transplantation: a potential novel
gliptin for the treatment of new-onset diabetes after renal transplan- therapeutic role for endothelin receptor antagonists. Transplanta-
tation. Int J Endocrinol 2014;2014:617638. Available online at: tion 2012;94:885–93.
https://doi.org/10.1155/2014/617638. 187. Sztanek F, et al. Decreased paraoxonase 1 (PON1) lactonase activity
161. Haidinger M, et al. Efficacy and safety of vildagliptin in new-onset in hemodialyzed and renal transplanted patients: a novel cardiovas-
diabetes after kidney transplantation: a randomized, double-blind, cular biomarker in end-stage renal disease. Nephrol Dial Transplant
placebo-controlled trial. Am J Transplant 2014;14:115–23. 2012;27:2866–72.
162. Werzowa J, et al. Vildagliptin and pioglitazone in patients with 188. Chung BH, et al. Clinical significance of early-onset hyperuricemia
impaired glucose tolerance after kidney transplantation: a placebo- in renal transplant recipients. Nephron Clin Pract 2011;117.
controlled clinical trial. Transplantation 2013;95:456–62. 189. Dew MA, Rosenberger EM, Myaskovsky L, et al. Depression and
163. Luther P, Baldwin D. Pioglitazone in the management of diabetes anxiety as risk factors for morbidity and mortality after organ trans-
mellitus after transplantation. Am J Transplant 2004;4:2135–8. plantation: a systematic review and meta-analysis. Transplantation
164. Voytovich MH, et al. Short-term treatment with rosiglitazone 2016;100:988–1003.
improves glucose tolerance, insulin sensitivity and endothelial 190. Patel RK, et al. Prognostic value of cardiovascular screening in
function in renal transplant recipients. Nephrol Dial Transplant potential renal transplant recipients: a single-center prospective
2005;20:413–8. observational study. Am J Transplant 2008;8:1673–83.
165. Nicoletto BB, et al. Effects of obesity on kidney transplantation out- 191. Webster A, et al. Cardiac testing for coronary artery disease in
comes: a systematic review and meta-analysis. Transplantation potential kidney transplant recipients. Cochrane Database Syst
2014;98:167–76. Rev 2011. Available online at: https://doi.org/10.1111/j.1440-
166. Hill CJ, et al. Recipient obesity and outcomes after kidney transplan- 1797.2012.01624.x.
tation: a systematic review and meta-analysis. Nephrol Dial Transpl 192. De Lima JJ, Wolff Gowdak LH, De Paula FJ, Franchini Ramires JA,
2015;30:1403–11. Bortolotto LA. The role of myocardial scintigraphy in the assessment
167. Lafranca JA, IJermans JN, Betjes MG, Dor FJ. Body mass index and of cardiovascular risk in patients with end-stage chronic kidney
outcome in renal transplant recipients: a systematic review and disease on the waiting list for renal transplantation. Nephrol Dial
meta-analysis. BMC Med 2015;13:111. Transplant 2012;27:2979–84.
168. Sood A, Hakim DN, Hakim NS. Consequences of recipient obesity on 193. Boden WE, et al. Optimal medical therapy with or without PCI for
postoperative outcomes in a renal transplant: a systematic review stable coronary disease. N Engl J Med 2010;356:1503–16.
and meta-analysis. Exp Clin Transplant 2016;14:121–8. 194. Marechal C, et al. Progression of coronary artery calcification and
169. de Vries LV, et al. Twenty-four hour urinary cortisol excretion and thoracic aorta calcification in kidney transplant recipients. Am J Kid-
the metabolic syndrome in prednisolone-treated renal transplant ney Dis 2012;59:258–69.
recipients. Steroids 2017;127:31–9. 195. Kasiske BL, Israni AK, Snyder JJ, Camarena A. Design considerations
170. Ahmadi SF, et al. Body mass index and mortality in kidney trans- and feasibility for a clinical trial to examine coronary screening before
plant recipients: a systematic review and meta-analysis. Am J kidney transplantation (COST). Am J Kidney Dis 2011;57:908–16.
Nephrol 2014;40. 196. Helve S, et al. Even mild reversible myocardial perfusion defects pre-
171. De Lima JJ, et al. Coronary events in obese hemodialysis patients before dict mortality in patients evaluated for kidney transplantation. Eur
and after renal transplantation. Clin Transplant 2015;29:971–7. Hear J Cardiovasc Imaging 2018;19(9):1019–25. Available online
172. Naik AS, et al. The impact of obesity on allograft failure after kidney at: https://doi.org/10.1093/ehjci/jex200.
transplantation. Transplantation 2016;100:1963–9. 197. Henry TD, Herzog CA. Bad kidneys are bad for the heart: but what
173. Potluri K, Hou S. Obesity in kidney transplant recipients and candi- can we do about it? Catheter Cardiovasc Interv 2012;80:358–60.
dates. Am J Kidney Dis 2010;56:143–56. 198. Lentine KL, et al. Cardioprotective medication use after acute myo-
174. Gusukuma LW, et al. Outcomes in obese kidney transplant recipi- cardial infarction in kidney transplant recipients. Transplantation
ents. Transplant Proc 2014;46:3416–9. 2011;91:1120–6.
175. Marszalek R, Ziemiaski P, Lisik W, et al. Bariatric surgery as a 199. Lenihan CR, Montez-Rath ME, Scandling JD, Turakhia MP, Win-
bridge for kidney transplantation in obese subjects. Ann Transplant kelmayer WC. Outcomes after kidney transplantation of patients
2012;17:108–12. previously diagnosed with atrial fibrillation. Am J Transplant
176. Freeman CM, et al. Addressing morbid obesity as a barrier to renal 2013;13:1566–75.
transplantation with laparoscopic sleeve gastrectomy. Am J Trans- 200. Findlay MD, et al. Risk factors and outcome of stroke in renal trans-
plant 2015;15:1360–8. plant recipients. Clin Transplant 2016;30:918–24.
177. Huang E, Bunnapradist S. Pre-transplant weight loss and survival 201. Lenihan CR, et al. Correlates and outcomes of warfarin initiation in
after kidney transplantation. Am J Nephrol 2015;41:448–55. kidney transplant recipients newly diagnosed with atrial fibrillation.
178. Rosas SE, Reese PB, Huan Y, et al. Pretransplant physical activity Nephrol Dial Transplant 2015;30:321–9.
predicts all-cause mortality in kidney transplant recipients. Am J 202. Salerno DM, et al. Direct oral anticoagulant considerations in solid
Nephrol 2012;35:17–23. organ transplantation: a review. Clin Transplant 2017;31.
179. Painter PL, et al. Effects of exercise training on coronary heart dis- 203. Sharma A, Gilbertson DT, Herzog CA. Survival of kidney transplan-
ease risk factors in renal transplant recipients. Am J Kidney Dis tation patients in the united states after cardiac valve replacement.
2003;42:362–9. Circulation 2010;121:2733–9.
180. Johansen KL, Painter P. Exercise in individuals with CKD. Am J Kid- 204. Shroff GR, Skeans M, Herzog CA. Outcomes of renal transplant and
ney Dis 2012;59:126–34. waiting list patients with bacterial endocarditis in the United States.
Nephrol Dial Transplant 2008;23:2381–5.
516 Kidney Transplantation: Principles and Practice

205. Silver SA, Huang M, Nash MM, Prasad GV. Framingham risk score 207. Schnitzler MA, et al. Use of 12-month renal function and baseline
and novel cardiovascular risk factors underpredict major adverse clinical factors to predict long-term graft survival: application to BEN-
cardiac events in kidney transplant recipients. Transplantation EFIT and BENEFIT-EXT trials. Transplantation 2012;93:172–81.
2011;92:183–9. 208. Pilmore H, Dent H, Chang S, McDonald SP, Chadban SJ. Reduction
206. Soveri I, et al. A cardiovascular risk calculator for renal transplant in cardiovascular death after kidney transplantation. Transplanta-
recipients. Transplantation 2011;94:57–62. tion 2010;89:851–7.
31 Infection in Kidney
Transplant Recipients
JAY A. FISHMAN, SYLVIA F. COSTA and BARBARA D. ALEXANDER

CHAPTER OUTLINE Overview Special Infectious Risks and Organ


Risk of Infection Procurement
Epidemiologic Exposures Evaluation of the Transplant Recipient
Donor-Derived Infections Selected Infections of Importance
Recipient-Derived Exposures General Considerations
Community Exposures Viral Pathogens
Nosocomial Exposures Cytomegalovirus
Net State of Immunosuppression Epstein–Barr Virus
Timeline of Infection Polyomaviruses
First Phase (First Month After JC Virus
Transplantation) Human Papillomavirus
Second Phase (1–12 Months After Human Immunodeficiency Virus
Transplantation) Fungal Infections
Third Phase (More Than 12 Months After Candida
Transplantation) Aspergillus
Assessment of Infectious Diseases in Central Nervous System Infections and
Recipients and Potential Donors Before Cryptococcus neoformans
Transplantation
Pneumocystis and Fever with Pneumonitis
Transplant Donor
Urinary Tract Infection
Deceased Donor Evaluation
Conclusions
Living Donor Evaluation

Overview Risk of Infection


Successful management of infections in kidney transplant The risk of infection in a kidney transplant recipient is deter-
recipients is a function of the immune status of the host mined by the interaction of two key factors:
  
and the epidemiology of infectious exposures. Transplant 1. The epidemiologic exposures of the patient, including
recipients are susceptible to a broad spectrum of infectious the timing, intensity, and virulence of the organisms
pathogens while manifesting diminished signs and symp- encountered.
toms of invasive infection. Thus the diagnosis of infection 2. The patient’s “net state of immunosuppression,” a con-
is more difficult in transplant recipients than in immuno- ceptual measure of all the factors that contribute to the
logically normal individuals. The interactions between host’s risk of infection.1,2
infection, immunosuppression, and immune function often   

result in clinical syndromes reflecting multiple simultane- The importance of any infectious exposure is determined
ous processes, such as infection and graft rejection. Immu- by the ability of the host to deal effectively with the patho-
nocompromised patients tolerate invasive infection poorly, gen. Thus the immunosuppressed diabetic with vascular
with high morbidity and mortality, lending urgency to the disease is at greater risk of bacterial skin infections than is a
need for an early, specific diagnosis to guide antimicrobial comparable immunosuppressed nondiabetic. Understand-
therapy. Given the predominant T-lymphocyte dysfunction ing the risk factors for each transplant recipient allows
inherent to transplant immunosuppression, viral infections development of a differential diagnosis for infectious syn-
are a major contributor to morbidity resulting in graft dys- dromes and development of preventive strategies (prophy-
function, graft rejection, systemic illness, and increased risk laxis, vaccination) appropriate to the individual’s unique
for other opportunistic infections (e.g., Pneumocystis and risks.3,4
Aspergillus) and virally mediated cancers.

517
518 Kidney Transplantation: Principles and Practice

EPIDEMIOLOGIC EXPOSURES significant morbidity in recipients form the basis of screen-


ing paradigms for organ donors.5–7 Bloodstream infections
Epidemiologic exposures of importance in the transplant (with bacteria or yeast) in donors at the time of donation
recipient can be divided into four overlapping categories: can cause local (abscess) or systemic infections, and, impor-
(1) donor-derived infections, (2) recipient-derived infec- tantly, may adhere to anastomotic sites (vascular, urinary)
tions, (3) community-derived exposures, and (4) nosoco- to produce leaks or mycotic aneurysms.
mial exposures (Table 31.1). Transmission of some donor-derived viral infections are
Donor-Derived Infections common and expected, including cytomegalovirus (CMV)
and Epstein–Barr virus (EBV), and are associated with
Diverse donor-derived infections have been recognized in specific syndromes in transplant recipients (see section on
transplant recipients. Some of these infections are latent selected infections of importance). The greatest risk of viral
(e.g., viral, parasitic), whereas others are active (e.g., bac- infections is transmission from seropositive donors (latent
teremia, fungemia) in the donor at the time of procure- infection) to seronegative (immunologically naïve) recipi-
ment. Frequent pathogens and endemic organisms causing ents (or D+/R−). Some viruses demonstrate accelerated
progression (lymphocytic choriomeningitis virus [LCMV],
rabies, West Nile virus [WNV]) in transplant recipients.
Latent infections, such as tuberculosis, toxoplasmosis, or
TABLE 31.1 Significant Epidemiologic Exposures
strongyloidiasis, may activate many years after the initial,
Relevant to Transplantation
often unrecognized exposures.
DONOR-DERIVED Donor screening for transplantation is limited by the
Viral available technology and by time constraints within which
Herpesvirus group (CMV, EBV, HHV-6, HHV-7, HHV-8, HSV) organs from deceased donors must be used (discussed
Hepatitis viruses (HBV, HCV) later).5,8,9 Routine screening of donors relies on history,
Retroviruses (HIV, HTLV-I/II) and both antibody detection (serologic tests) and nucleic
Others (rabies, LCMV, WNV)
acid testing (NAT) for common infections. Risk factors
Bacteria for infection in the donor are often unknown. As a result,
Gram-positive and gram-negative bacteria (Staphylococcus, Pseudo- transmission may occur from seronegative donors with
monas, Enterobacteriaceae) active viral or other exposures (before seroconversion in
Mycobacteria (tuberculous and nontuberculous)
Nocardia species the “window period”) or with viral loads below the limits
of detection by the NAT selected. This risk has been dem-
Fungi
onstrated by clusters of donor-derived Trypanosoma cruzi
Candida species
(Chagas’ disease), rabies virus, WNV, and LCMV infec-
Aspergillus
Endemic fungi (Cryptococcus neoformans) tions in organ transplant recipients.7,10–13 NAT for donor
Geographically restricted fungi (Histoplasma capsulatum, Coccidioides screening (e.g., for human immunodeficiency virus [HIV],
immitis, Blastomyces dermatitidis, Paracoccidioides brasiliensis) hepatitis B virus [HBV], hepatitis C virus [HCV], WNV) has
Parasites the capacity to reduce the window period between expo-
Toxoplasma gondii sure and pathogen detection over serologic tests albeit with
Trypanosoma cruzi some risk for false-positive assays given heightened assay
NOSOCOMIAL EXPOSURESa
sensitivity.14–18
Given the risk of transmission of infection from the
Methicillin-resistant Staphylococcus aureus
Vancomycin-resistant enterococci organ donor to recipients, certain syndromes should be
CRE and ESBL gram-negative bacilli considered relative contraindications to organ donation.
Aspergillus species Because kidney transplantation is typically elective sur-
Non-albicans Candida species gery, it is reasonable to avoid donation from individuals
COMMUNITY EXPOSURESa with unexplained fever, rash, or infectious syndromes,
Foodborne and water-borne (Listeria monocytogenes, Salmonella, including meningitis or encephalitis. At some centers,
Cryptosporidium, hepatitis A, Campylobacter) transplantation from donors with untreated HCV or HIV
Respiratory viruses (RSV, influenza, parainfluenza, adenovirus, meta- (to HIV-positive recipients) infections is undertaken. Com-
pneumovirus) mon criteria for exclusion of organ donors are listed in
Common viruses, often with exposure to children (coxsackievirus,
parvovirus, polyomavirus, papillomavirus) Table 31.2.
Atypical respiratory pathogens (Legionella, Mycoplasma, Chlamydia)
Geographically restricted fungi, Cryptococcus, Pneumocystis jirovecii
Recipient-Derived Exposures
Parasites (often remote) (Strongyloides stercoralis, Leishmania, T. Recipient-derived exposures generally reflect coloniza-
gondii, T. cruzi) tion or latent infections that reactivate during immuno-
Amoeba (Naegleria fowleri, Balamuthia mandrillaris) suppression.19 Certain common infections are recognized
CMV, cytomegalovirus; CRE, carbapenem-resistant Enterobacteriaceae; during the evaluation of the transplant candidate, includ-
EBV, Epstein–Barr virus; ESBL, extended-spectrum beta-lactamase; HBV, ing HBV, HCV, and HIV. It is necessary to obtain a care-
hepatitis B virus; HCV, hepatitis C virus; HHV, human herpesvirus; HIV, ful history of prior infections, travel, and exposures to
human immunodeficiency virus; HSV, herpes simplex virus; HTLV, human guide preventive strategies and empirical therapies.
T cell lymphotropic virus; LCMV, lymphocytic choriomeningitis virus; RSV,
respiratory syncytial virus.
Notable among these infections are mycobacterial infec-
aColonization and infection of the recipient in advance of transplantation tion (including tuberculosis), strongyloidiasis, viral infec-
may occur because of these potential pathogens. tions (herpes simplex virus [HSV] and varicella-zoster
31 • Infection in Kidney Transplant Recipients 519

virus [VZV] or shingles), histoplasmosis, coccidioidomy- transplantation because immune response is likely to be
cosis, and paracoccidiomycosis (Fig. 31.1). Vaccination more robust, and live virus vaccines are generally con-
status should be evaluated (childhood vaccines, tetanus, traindicated after transplantation (Table 31.3). Dietary
HBV, influenza, pneumococcus); vaccines not previ- habits also should be considered, including the use of well
ously administered should be considered in advance of water (Cryptosporidium) and consumption of uncooked
meats (Salmonella, Listeria, hepatitis E) and unpasteurized
dairy products (Listeria).
TABLE 31.2 Common Infectious Exclusion Criteria for
Organ Donorsa Community Exposures
CENTRAL NERVOUS SYSTEM INFECTION Common exposures in the community are often related
Unknown or untreated infection of central nervous system (encepha- to contaminated food and water ingestion; exposure to
litis, meningitis) infected family members or coworkers; or exposures related
Herpes simplex encephalitis or other encephalitis to hobbies, travel, or work. Infection caused by common
History of JCV infection respiratory viruses (influenza, parainfluenza, respiratory
WNV infection
Cryptococcal infection syncytial virus [RSV], adenovirus, and metapneumovirus)
Rabies and by more atypical pathogens (HSV) carry the risk of
Creutzfeldt–Jakob disease viral pneumonia and increased risk of bacterial or fungal
Other fungal or viral encephalitis superinfections. Community (contact or transfusion-asso-
Amoebic encephalitis
ciated) exposure to CMV and EBV may produce severe pri-
DISSEMINATED AND UNTREATED INFECTIONS mary infection in the nonimmune host. Recent and remote
HIV (serologic or molecular; may be considered for HIV-positive exposures to endemic, geographically restricted systemic
recipient) mycoses (Blastomyces dermatitidis, Coccidioides immitis,
HSV (with viremia), acute EBV (mononucleosis)
Serologic or molecular evidence of HTLV-I/II
Histoplasma capsulatum, and Paracoccidioides brasiliensis)
Active hepatitis A (may consider HBV and HCV-infected donors for and Mycobacterium tuberculosis can result in localized pul-
appropriate recipients) monary, systemic, or metastatic infection. Asymptomatic
Parasitic infections (Trypanosoma cruzi, Leishmania donovani, Strongy- Strongyloides stercoralis infection may activate more than
loides stercoralis, Toxoplasma gondii) 30 years after initial exposure as a result of immunosup-
INFECTIONS DIFFICULT TO TREAT ON IMMUNOSUPPRESSION pressive therapy (see Fig. 31.1). Such reactivation can
Active tuberculosis result in either a diarrheal illness and parasite migration
SARS, MERS with hyperinfection syndrome (characterized by hemor-
Untreated pneumonia rhagic enterocolitis, hemorrhagic pneumonia, or both) or
Untreated bacterial or fungal sepsis (e.g., candidemia)
Untreated syphilis disseminated infection with accompanying (usually) gram-
Multisystem organ failure resulting from overwhelming sepsis, gan- negative or polymicrobial bacteremia or meningitis. Gas-
grenous bowel troenteritis secondary to Salmonella, Cryptosporidium, and
aThese
a variety of enteric viruses (e.g., norovirus) can result in
must be considered in the context of the individual donor/recipient.
EBV, Epstein–Barr virus; HBV, hepatitis B virus; HCV, hepatitis C virus; HIV,
persistent infection, with more severe and prolonged diar-
human immunodeficiency virus; HSV, herpes simplex virus; HTLV, human rheal disease and an increased risk of primary or secondary
T cell lymphotropic virus; MERS, Middle East respiratory syndrome; SARS, bloodstream invasion and metastatic infection.20
severe acute respiratory syndrome.

A B
Fig. 31.1 Simultaneous Pneumocystis pneumonia and bacterial lung abscess secondary to coinfection by Strongyloides stercoralis in a Vietnamese
kidney transplant recipient. (A) Chest radiograph shows a lung abscess secondary to Enterobacter species. Bronchoscopic examination also revealed
simultaneous Pneumocystis jirovecii and S. stercoralis infections. Migration of Strongyloides across the wall of the gastrointestinal tract during immuno-
suppression (hyperinfection) is associated with systemic signs of sepsis and central nervous system infection (parasitic and bacterial). (B) S. stercoralis
from the lung of the same patient.
520 Kidney Transplantation: Principles and Practice

TABLE 31.3 Vaccinations to Consider Before TABLE 31.4 Factors Contributing to the Net State of
Transplantationa Immunosuppression
Measles/mumps/rubella (MMR) Immunosuppressive therapy: type, temporal sequence, intensity,
Diphtheria/tetanus/pertussis (DTP) cumulative dose
Poliovirus Prior therapies (chemotherapy and antimicrobials)
Haemophilus influenzae b (Hib) Mucocutaneous barrier integrity (catheters, lines, drains)
Hepatitis B, Hepatitis A Neutropenia, lymphopenia (often drug-induced)
Pneumococcus Underlying immunodeficiencies
Influenza (subunit vaccine) Autoimmune diseases
Varicella (Live, attenuated vaccine; zoster vaccine recombinant, adju- Hypogammaglobulinemia from proteinuria or drug therapy
vanted under study) Complement deficiencies
Other disease states (HIV, lymphoma/leukemia)
aLive virus vaccinations are generally precluded in immunosuppressed hosts. Metabolic conditions (uremia, malnutrition, diabetes, cirrhosis)
Viral infections (CMV, HBV, HBC, RSV)
Nosocomial Exposures Graft rejection and treatment
Cancer/cellular proliferation
Nosocomial infections are of increasing importance. Organ-
isms with significant multidrug antimicrobial resistance CMV, cytomegalovirus; HBV, hepatitis B virus; HCV, hepatitis C virus; HIV,
(MDRO) are present in most medical centers, including human immunodeficiency virus; RSV, respiratory syncytial virus.
enterococci that are resistant to vancomycin, linezolid,
daptomycin and/or quinupristin/dalfopristin; methicillin-
resistant staphylococci; gram-negative bacteria producing TABLE 31.5 Immunosuppression and Common
Infections
extended-spectrum beta-lactamases (ESBL) and carbapen-
emases (CRE); and fluconazole-resistant Candida species (see Agent Common Infections/Effects
Table 31.1).21,22 A single case of nosocomial Aspergillus Antilymphocyte globulins Activation of latent viruses, fever,
infection in an immunocompromised host in the absence (lytic) and alloimmune cytokines
of a clear epidemiologic exposure should be viewed as a fail- response
ure of infection control practices. Antimicrobial misuse and Anti-CD20 antibody Unknown to date
inadequate infection control practices have caused increased Plasmapheresis Encapsulated bacteria
Corticosteroids Bacteria, Pneumocystis jirovecii, HBV, HCV
rates of Clostridium difficile colitis. Outbreaks of infections Azathioprine Neutropenia, papillomavirus (?)
secondary to Legionella have been associated with hospital Mycophenolate mofetil Early bacterial infection, late CMV (?)
plumbing and contaminated water supplies or ventilation Calcineurin inhibitors Enhanced viral replication (absence
systems. Nosocomial spread of Pneumocystis jirovecii between of immunity), gingival infection,
immunocompromised patients has been documented.23,24 intracellular pathogens
mTOR inhibitors Poor wound healing, idiosyncratic
Respiratory viral infections may be acquired from medical pneumonitis syndrome
staff and should be considered among the causes of fever and Belatacept Posttransplant lymphoproliferative
respiratory decompensation in hospitalized or institutional- disorder
ized immunocompromised individuals. Each nosocomially
CMV, cytomegalovirus; HBV, hepatitis B virus; HCV, hepatitis C virus; mTOR,
acquired infection should be investigated to ascertain the mammalian target of rapamycin.
source and to prevent subsequent infections. The question mark indicates possible side effect in some literature.

Assessment of the overall degree of immune compromise


NET STATE OF IMMUNOSUPPRESSION remains difficult. The combination of organ dysfunction,
The net state of immunosuppression is a conceptual immunosuppression, viral infections, nutritional status,
measure of the risk factors for infection in an individual, technical factors, and other factors in infectious risk resists
including immunosuppressive medications and iatrogenic quantification. Measures of pathogen-specific (i.e., cellular)
conditions (Table 31.4). Among the most important are as immune function are useful in guiding prophylaxis for spe-
follows: cific infections in individuals. Commercialized assays exist for
  
CMV and tuberculosis including interferon-γ-release assays
1. The specific immunosuppressive therapy, including
(IGRA), ELISpot, major histocompatibility complex (MHC)-
dose, duration, and sequence of agents (Table 31.5)
tetramer staining, or intracellular cytokine staining. Low
2. Technical difficulties during transplantation resulting in
serum antibody levels correlate with the overall risk of infec-
an increased incidence of leaks (blood, lymph, urine) and
tion but specific cutoff values and indications for replace-
fluid collections, devitalized tissue, poor wound healing,
ment therapy are lacking.25,26 Few data exist on functional
and prolonged use of surgical drainage catheters
immune reconstitution after T- or B-lymphocyte depletion or
3. Prolonged instrumentation, including airway intuba-
with costimulatory blockade. Recent data support the impor-
tion and use of vascular access devices (e.g., dialysis
tance of genetic polymorphisms among transplant recipients
catheters)
and risk of microbial colonization and infection.27–29
4. Prolonged use of broad-spectrum antibiotics
5. Renal or hepatic dysfunction, or both (in addition to
graft dysfunction)
6. Presence of infection with an immunomodulating virus,
Timeline of Infection
including CMV, EBV, HBV, HCV, or HIV
   With standardized immunosuppressive regimens, the most
Specific immunosuppressive agents are associated with common infections vary in a predictable pattern depending
increased risk for certain infections (see Table 31.5). on the time elapsed since transplantation (Fig. 31.2). This is
31 • Infection in Kidney Transplant Recipients 521

Nosocomial technical Opportunistic,

TRANSPLANT
Community
Donor-derived relapsed, residual
acquired infections
Recipient-dervied Activation of latent infection

4 Weeks 1–12 Months 12 Months

MRSA, Candida, VRE, HSV, CMV, HBV, HCV, EBV, Community-acquired


Aspergillus, aspiration VZV, HSV, Listeria, TB, PCP, BK pneumonia, Aspergillus,
C. difficile colitis virus, Nocardia, Toxoplasma, dermatophytes, CMV,
Strongyloides, Leishmania colitis, UTI, Cryptococcus

Wounds, Lines, Greatest impact of Skin and anogenital


Leaks, Aspiration, UTI immunosuppression cancers, PTLD

Common variables in risk:


• Antimicrobial prophylaxis
• Anti-rejection therapy (T-cell depletion)
• New immunosuppressive agents
• Neutropenia, lymphopenia
• Immunomodulatory viral infections (CMV, HCV, EBV)
• Antimicrobial resistance

Fig. 31.2 The timeline of posttransplantation infections. Infection after transplantation tends to occur in a predictable pattern based on the epidemio-
logic exposure of the host and the nature of immune deficits. Patients with infections falling outside the usual patterns suggest unusual exposures or
excessive immunosuppression. BKV, BK virus; CMV, cytomegalovirus; EBV, Epstein–Barr virus; HBV, hepatitis B virus; HCV, hepatitis C virus; HSV, herpes
simplex virus; MRSA, methicillin-resistant Staphylococcus aureus; PJP, Pneumocystis jirovecii pneumonia; PTLD, posttransplant lymphoproliferative
disorder; TB, tuberculosis; UTI, urinary tract infection; VRE, vancomycin-resistant enterococcus; VZV, varicella-zoster virus.

primarily a reflection of changing risk factors over time, includ- suspected to have infection; (2) to provide a clue to the pres-
ing surgery and hospitalization, tapering of immunosuppres- ence of an excessive environmental hazard for the individ-
sion, acute and chronic rejection, and exposure to infections in ual, either within the hospital or in the community; and (3)
the community.2,30 The predicted pattern of infection changes to serve as a guide to the design of preventive antimicrobial
with alterations in the immunosuppressive regimen (e.g., strategies. Infections occurring outside the usual period or
increased steroids for graft rejection), intercurrent viral infec- of unusual severity suggest either an intense epidemiologic
tions, neutropenia (drug toxicity), graft dysfunction, or sig- exposure or excessive immunosuppression.
nificant epidemiologic exposures (travel or food). The timeline The prevention of infection must be linked to the risk for
remains a useful starting point for the differential diagnosis of infection at various times after transplantation. Table 31.6
infection after transplantation, although it is altered by the outlines some routine preventive strategies, keeping in mind
introduction of new immunosuppressive agents and patterns that such strategies serve only to delay the onset of infection
of use, including reduced use of corticosteroids and calcineurin in the face of epidemiologic pressure. Use of antimicrobial
inhibitors, increased use of antibody-based (induction) thera- prophylaxis, vaccines, and behavioral modifications (e.g.,
pies or sirolimus, routine antimicrobial prophylaxis, improved routine hand washing or advice against digging in gardens
molecular assays, antimicrobial resistance, transplantation of without masks) may result in a “shift to the right” of the
HIV-infected and HCV-infected individuals, and broader epide- infection timeline, unless the intensity of immunosuppres-
miologic exposures from work or travel. sion is reduced or immunity develops.
There are three overlapping periods of risk for infection
after transplantation (see Fig. 31.2), each associated with FIRST PHASE (FIRST MONTH AFTER
differing patterns of common pathogens, as follows2:
   TRANSPLANTATION)
1. The perioperative period to approximately 4 weeks after
During the first month after transplantation, three types of
transplantation, reflecting surgical and technical com-
infection occur. The first is infection or colonization present
plications and nosocomial exposures
in the recipient before transplantation that emerges in the
2. The period from 1 to 12 months after transplantation
setting of surgery and immunosuppression. Pretransplan-
(depending on the rapidity of taper of immunosuppres-
tation pneumonia and vascular access infections are com-
sion, the use of antilymphocyte “induction” therapy,
mon examples of this type of infection. Colonization of the
and deployment of prophylaxis), reflecting intensive
recipient with resistant organisms that infect intravenous
immunosuppression with viral activation and opportu-
catheters or surgical drains also is common (e.g., methicillin-
nistic infections
resistant Staphylococcus aureus [MRSA]). All infection should
3. The period beyond the first year after transplantation,
be controlled or eradicated to the degree possible before
reflecting community-acquired exposures and some
transplantation.
unusual pathogens based on the level of maintenance
The second type of early infection is donor-derived. This
immunosuppression
   may be nosocomially derived (resistant gram-negative
The timeline can be used in a variety of ways: (1) to bacilli and S. aureus or Candida species) colonization during
establish a differential diagnosis for a transplant patient the donor’s hospitalization, secondary to systemic infection
522 Kidney Transplantation: Principles and Practice

TABLE 31.6 Renal Transplantation: Routine Antimicrobial Protocols


A. PNEUMOCYSTIS JIROVECII PNEUMONIA (PJP) AND GENERAL ANTIBACTERIAL PROPHYLAXIS

REGIMEN
□ One single-strength TMP-SMX tablet (containing 80 mg trimethoprim, 400 mg sulfamethoxazole) orally daily for a minimum of 4–6 months post-

transplantation. Patients infected with cytomegalovirus (CMV), with chronic rejection, or with recurrent infections are maintained on lifelong pro-
phylaxis. A thrice-weekly regimen of TMP-SMX prevents PJP, but does not prevent other infections (e.g., urinary tract infection, Nocardia, Listeria,
Toxoplasma, and other gastrointestinal and pulmonary infections).
ALTERNATIVE REGIMEN
□ For patients proven not to tolerate TMP-SMX, alternative regimens include: (1) a combination of atovaquone, 1500 mg orally daily with meals, plus

levofloxacin, 250 mg orally daily (or equivalent fluoroquinolone without anaerobic activity); (2) pentamidine, 300 mg intravenously or inhaled every
3–4 weeks; or (3) dapsone, 100 mg orally daily twice weekly, with or without pyrimethamine. Each of these agents has toxicities that must be con-
sidered (e.g., hemolysis in G6PD-deficient hosts with dapsone). None of these alternative programs offers the same broad protection of TMP-SMX.

B. CYTOMEGALOVIRUS AND HERPESVIRUS PROPHYLAXISa,b

CMV UNIVERSAL ANTIVIRAL PROPHYLAXIS (KIDNEY OR PANCREAS RECIPIENTS)


Donor (D) and Recipient (R) CMV Possible Regimen Monitoring (Viral Load NAT)
serologic status +/−
T-cell depletion in induction therapy
D+/R−with induction using T cell Valganciclovir 900 mg po × QD (or IV ganciclovir 5 mg/kg Monthly for 6 months after
depletion IV until taking po) (corrected for renal function) for 6 months discontinuation of therapya
(Highest risk)
D+/R− without T cell depletion Valganciclovir 900 mg po × QD (or IV ganciclovir 5 mg/kg IV Monthly for 6 months after
(costimulatory blockade) until taking po) (corrected for renal function) for 3–6 months discontinuation of therapya
(High risk)
R+ without T cell depletion Oral valganciclovir (900 mg/day corrected for renal function) Symptoms only
(costimulatory blockade) × 3 months or preemptive therapy
(Intermediate risk)
R+ with T cell depletion or Oral valganciclovir (900 mg/day corrected for renal function) Symptoms only
desensitization, × 3–6 months or preemptive therapy
(D− at Intermediate risk)
(D + at Higher risk)
D−/R− Oral famciclovir 500 mg po qd × 3–4 months (or valacyclovir Symptoms, fever/neutropenia
(Lowest risk) 500 bid or acyclovir 400 tid); Leukocyte-filtered blood
Target HSV/VZV

Neutropenia: The doses of antiviral therapies are not reduced for neutropenia. Formal creatinine clearance measurement may be useful in dose adjustment.
Alternatives to valganciclovir: High dose valacyclovir (≥8 g/day)—compliance is difficult and efficacy not well studied; po ganciclovir (3 g/day)—lower bioavailability.
First dose of ganciclovir is often intravenous but valganciclovir may be used if taking oral medications. All antiviral agents adjusted for renal function. For
abnormal renal function, formal creatinine clearance measurement may be indicated. The dose of antiviral therapy is generally not reduced for neutropenia.
Consider other options first.
Antifungal Prophylaxis: Mucocutaneous candidiasis can be prevented with oral clotrimazole or nystatin 2–3 times per day during corticosteroid therapy or
in the face of broad-spectrum antibacterial therapy and in diabetic transplant patients. Fluconazole,.200 mg/day for 10–14 days, is used to treat prophylaxis
failures. Routine prophylaxis with fluconazole is used for pancreas and kidney–pancreas transplants. Other prophylaxis is determined based on the presence
or absence of colonization or other risk factors for fungal infection.
aHybrid Prophylaxis: Many centers prefer universal prophylaxis for highest risk kidney recipients (D+/R− or R+ with lymphocyte depletion) and preemptive

therapy for lower risk groups.


bPreemptive Therapy: Preemptive therapy requires a carefully organized monitoring program and patient compliance. Either a molecular CMV viral load test

or a pp65 antigenemia assay may be used for monitoring. Monitoring should be performed once weekly after transplantation for 12–24 weeks. Infections
indicated by positive assays are treated with either oral valganciclovir or intravenous ganciclovir. Therapy is continued at least until viremia is undetectable.
D, donor serology; R, recipient serology; NAT, nucleic acid test normalized to the World Health Organization international standard.

in the donor (e.g., line infection), or contamination dur- patients undergoing comparable surgery. Given immuno-
ing the organ procurement process. Active infections may suppression, the signs of infection may be subtle and the
be transmitted from donor to recipient and emerge earlier severity or duration is usually increased. Thus bowel perfo-
than normally predicted (e.g., HSV, tuberculosis, histo- ration may be clinically silent, marked only by tachycardia,
plasmosis). Most recent clusters of donor-derived infection a rising white blood cell count, abnormal liver function tests,
have been the result of unfortunate timing—a donor who or graft dysfunction. The technical skill of the surgeons and
acquired acute infection (HIV, WNV, rabies, or LCMV) meticulous postoperative care (i.e., wound care and proper
before and unrelated to the cause of death. maintenance and timely removal of endotracheal tubes,
The third and most common source of infection in the vascular access devices, and drainage catheters) determine
early period is related to the transplant procedure. These the degree of risk for these infections. Another important
infections include surgical wound infections, pneumonia infection during this period is Clostridium difficile colitis.31,32
(aspiration), bacteremia associated with vascular access or Limited perioperative antibiotic prophylaxis (i.e., from a
surgical drainage catheters, urinary tract infections, and single dose to 24 hours of an antibiotic such as cefazolin or
superinfection of fluid collections—leaks of vascular or uri- amoxicillin-clavulanate) is usually adequate for renal trans-
nary anastomoses or of lymphoceles. These are nosocomial plantation with additional coverage required for known
infections and, as such, may carry the same antimicrobial- risk factors (e.g., prior colonization with MRSA). For pan-
resistant pathogens observed in nonimmunosuppressed creas transplantation, additional perioperative prophylaxis
31 • Infection in Kidney Transplant Recipients 523

against yeasts is useful with fluconazole, mindful of poten- immunosuppression to maintain graft function, who had
tial increases in sirolimus and calcineurin inhibitor levels a poor technical outcome of transplantation (leaks or vas-
when used concomitantly with azole antifungal agents. cular issues) or poor graft function, or who has persistent
Opportunistic infections are notably absent in the first viral or other infections (e.g., C. difficile colitis), which pre-
month after transplantation, even though the daily doses dict long-term susceptibility to other infections (third phase,
of immunosuppressive drugs may be greatest during this discussed next). Such patients may benefit from prolonged
time. The implication of this observation is important (lifelong) prophylaxis (antibacterial, antifungal, antiviral,
because it suggests the daily dose of immunosuppressive or a combination) to prevent life-threatening infection.
drugs is less important than the cumulative dose (i.e., the Opportunistic infections reflect the immunosuppressive
“area under the curve”) for determining the true state of regimen used, individual epidemiology, and the presence or
immunosuppression. The net state of immunosuppression absence of immunomodulating viral infection. Viral patho-
during the first month after transplant is not great enough gens (and rejection) are responsible for most febrile episodes
to support opportunistic infections, unless exposure has that occur in this period. Anti-CMV strategies and trim-
been excessive. Accordingly, the occurrence of a single ethoprim/sulfamethoxazole (TMP-SMX) prophylaxis are
case of opportunistic infection in this period should trig- effective in decreasing the risk of infection. TMP-SMX pro-
ger an epidemiologic investigation for an environmental phylaxis effectively prevents Pneumocystis pneumonia and
hazard. reduces the incidence of urinary tract infection and urosep-
sis, L. monocytogenes meningitis, Nocardia species infection,
SECOND PHASE (1–12 MONTHS AFTER and T. gondii.
TRANSPLANTATION)
THIRD PHASE (MORE THAN 12 MONTHS AFTER
The second phase of infection was traditionally 1 to 3 TRANSPLANTATION)
months, but has been extended because of two main factors:
successful use of prophylaxis or monitoring programs tar- Recipients who underwent transplantation more than a
geting CMV and the herpesviruses, Pneumocystis, urinary year previously can be divided into three groups in terms of
tract infections, and HBV, and intensification of immuno- infectious risk. Most transplant recipients (70%–80%) have
suppression using more potent agents or antibody-based a technically good procedure with satisfactory allograft
induction therapies with prolonged effects on immune function, reduced immunosuppression, and absence of
function (see Table 31.5).1,2 Infection in the transplant chronic viral infection. These patients resemble the general
recipient 1 to 12 months after transplantation has one of community in terms of infection risk, with community-
three causes: acquired respiratory viruses constituting the major risk.
  
Occasionally, such patients develop primary CMV infection
1. Infection from the perisurgical period, including relapsed
(socially acquired) or infections related to underlying dis-
C. difficile colitis, inadequately treated pneumonia, or
eases (e.g., skin infections in diabetics).
infection related to a technical problem (e.g., urine leak,
A second group of patients has chronic viral infection,
hematoma). Fluid collections in this setting generally
which may produce end organ damage (e.g., BK poly-
require drainage.
omavirus leading to fibrosis, HCV leading to cryoglobu-
2. Viral infections including CMV, HSV, VZV, human her-
linemia and cirrhosis, CMV with chronic graft rejection)
pesvirus (HHV)-6 or HHV-7, EBV, HBV, HCV, and HIV.
or malignancy (e.g., posttransplantation lymphoprolifer-
Viruses are prominent given the importance of T cell
ative disease (PTLD) secondary to EBV, skin or anogeni-
function in antiviral control and the disproportionate
tal cancer related to papillomaviruses). In the absence
degree of T cell inhibition by most immunosuppressive
of specific and effective antiviral therapy, these patients
regimens. Furthermore, these viruses are systemically
often suffer graft rejection with the reduced intensity of
immunosuppressive, predisposing to opportunistic
immunosuppression.
infection or acceleration of other infections and, via
A third group of patients has unsatisfactory allograft
chronic immune stimulation, predispose to graft rejec-
function and suffers the ravages of renal dysfunction,
tion. Useful therapies are now available for most of these
often despite intensified immunosuppression used to pre-
pathogens. The herpesvirus infections are lifelong and
serve graft function. Declining allograft function may
tissue-associated, transmitted with the allograft from
be a result of underlying disease progression (athero-
seropositive donors. Other common viral pathogens
sclerosis, IgA, or diabetes), calcineurin inhibitor toxic-
of this period include BK polyomavirus (in association
ity, or humoral and cellular graft rejection. Thus these
with allograft dysfunction or polyomavirus-associated
patients are overimmunosuppressed relative to the risk
nephropathy [PyVAN]) and community-acquired respi-
of infection. These patients may benefit from lifetime
ratory viruses (adenovirus, influenza, parainfluenza,
maintenance TMP-SMX and often fluconazole prophy-
RSV, metapneumovirus). Bacterial and fungal superin-
laxis. In this group, one also should consider organisms
fection of virally infected hosts is common.
more commonly associated with immune dysfunction of
3. Opportunistic infections secondary to P. jirovecii, L.
acquired immunodeficiency syndrome (AIDS; Bartonella,
monocytogenes, Toxoplasma gondii, Cryptococcus neofor-
Rhodococcus, Cryptosporidium, and microsporidia) and
mans, Nocardia, Aspergillus, and other agents
   invasive fungal pathogens (Aspergillus, Mucorales, and
In this period, the stage also is set for the emergence of dematiaceous or pigmented molds). Even minimal clini-
a subgroup of patients—the “chronic ne’er do well”— cal signs or symptoms warrant careful evaluation in this
the patient who requires higher than usual levels of group of high-risk patients.
524 Kidney Transplantation: Principles and Practice

Assessment of Infectious Diseases penetrate the cerebrospinal fluid before organ procure-
ment. Individuals with unidentified and untreated causes of
in Recipients and Potential meningoencephalitis or sepsis should not be used as organ
Donors Before Transplantation donors. Donor-derived infections caused by Candida species
have resulted from organ contamination or candidemia at
Guidelines for pretransplant screening have been the subject the time of procurement.36,37 Susceptibility testing of the
of several publications, including a consensus conference isolate and prolonged treatment (2–4 weeks) with effective
of the Immunocompromised Host Society, the American agents to avoid pyelonephritis, abscess formation, mycotic
Society for Transplantation Clinical Practice Guidelines aneurysm, or candidemia in the recipient is recommended.
for the evaluation of kidney transplant candidates, and the Vascular involvement by Candida species in the recipient
American Society of Transplant Surgeons Clinical Practice requires at least 6 weeks of therapy. Certain active infections
Guidelines for the evaluation of living kidney transplant (CMV, VZV, HSV, EBV, or HCV) may be unsuspected even
donors.19,33,34 in the seropositive donor and require NAT for diagnosis.
Likewise, the donor’s clinical, social, and medical histories
TRANSPLANT DONOR are essential to reducing the risk of such infections. Known
infection should be treated before procurement if possible.
Deceased Donor Evaluation See previous discussion of unrecognized donor pathogens.
A crucial challenge in screening deceased organ donors is Major exclusion criteria are outlined in Table 31.2.
the narrow time frame for the evaluation. A useful organ
must be procured and implanted before some microbio- Living Donor Evaluation
logic assessments have been completed. Thus bacteremia The living donor procedure should be considered elec-
or fungemia may not be detected until after transplantation tive, and the evaluation should be completed and infec-
has been performed. Such infections generally have not tions treated before donation.38 An interim history must
resulted in transmission of infection if the infection has been be taken at the time of surgery to assess the presence of
adequately treated before procurement using antimicrobial new infections since the initial donor evaluation. Intercur-
agents to which the organism is susceptible for an appro- rent infections (flu-like illness, headache, confusion, myal-
priate duration. In recipients of tissues from 95 bacteremic gias, cough) might be the harbinger of important infection
donors, a mean of 3.8 days of effective therapy after trans- (WNV, severe acute respiratory syndrome [SARS], Trypano-
plantation prevented transmission of susceptible pathogens soma cruzi). Live donors undergo a battery of serologic tests
(in an era of reduced antimicrobial resistance). Surveil- (Table 31.7), purified protein derivative (PPD) skin test or
lance with additional courses of therapy in the recipient are tuberculosis interferon-γ release assay, and, if indicated,
employed, targeting known donor-derived pathogens.2,35 chest radiograph. The testing must be individualized, based
Bacterial meningitis must be treated with antibiotics that on unique risk factors (e.g., travel). Of note in the kidney

TABLE 31.7 Pretransplant Evaluation of Living Donors


Quantitative Molecular Patients With Expsoure
Pathogen Laboratory Test Test Available All Patients to Endemic Area
CMV Serologies X X
HSV Serologies X X
EBV Serologies X
HIV Serologies X X
HBV Serologies including: X X
HBV surface antigen and
HBV surface antibody
HCV Serologies X X
Treponema pallidum RPR or VDRL test X
Toxoplasma gondii Serologies X
Strongyloides stercoralis Serologies X
Stool ova and parasite examination
Leishmania Serologies X
Trypanosoma cruzi Serologies X
Blood smear
Shistosomiasis Urine ova and parasite examination X
with or without endoscopy
Histoplasma capsulatum Serologies X
Coccidioides immitis Serologies X
Bacteria and yeasts Urinalysis and culture X
Mycobacterium tuberculosis Skin test or IGRA X
Latent and active pulmonary Chest x-ray (routine) X
infections

anti-HBs, antibody to hepatitis B surface antigen; CMV, cytomegalovirus; EBV, Epstein–Barr virus; HBsAg, hepatitis B surface antigen; HBV, hepatitis B virus; HCV,
hepatitis C virus; HIV, human immunodeficiency virus; HSV, herpes simplex virus; IGRA, interferon gamma release assay; RPR, rapid plasma reagin; VDRL,
venereal disease research laboratory; VZV, varicella-zoster virus.
31 • Infection in Kidney Transplant Recipients 525

transplant recipient is the exclusion of urinary tract infec- antibody titer indicates either vaccination or prior infec-
tions (including both bacteria and yeasts) and bacteremia tion. HBcAb-IgM positivity suggests active HBV infection,
at the time of donation. The US Public Health Service sug- whereas IgG positivity suggests a more remote or persistent
gests rescreening potential donors as close to donation as is infection. The HBsAg-negative, HBcAb-IgG-positive donor
feasible, using NAT for HIV, HCV, and HBV. will have viral DNA in the liver but may be appropriate as
a kidney donor for HBV-infected or vaccinated, and thus
Special Infectious Risks and Organ Procurement immune, renal recipients; quantitative viral assays for HBV
Tuberculosis. Mycobacterium tuberculosis from the should be obtained to guide further therapy.52 The presence
donor represents approximately 4% of reported post- of HBsAg-negative, HBcAb-IgG-positive assays may be a
transplant tuberculosis cases.39,40 Much higher rates false-positive result or reflect true, latent HBV infection.
occur in endemic regions.41,42 Active disease should be The effect of HCV infection has changed in the era of
excluded in PPD-positive donors with chest radiograph, directly active antiviral agents (DAA); evidence exists that
sputum cultures, and chest computed tomography (CT) if the use of HCV-positive kidneys has clinical benefits for the
the chest radiograph is abnormal. Urine acid-fast bacillus recipient.53 Pan-genotypic DAA therapies cure over 95%
cultures may be useful in a PPD-positive kidney donor. of HCV infections in transplantation.50 Untreated, HCV
Isoniazid prophylaxis of the recipient should be consid- infection progresses to cirrhosis more rapidly with immu-
ered for untreated PPD-positive donors. Factors favoring nosuppression and with CMV coinfection (see Chapter 32
prophylaxis include a donor from an endemic region, for detailed discussion). Side effects of historical therapies
high-risk social environment, or use of a high-dose ste- (pegylated interferon-α and ribavirin) are increased in the
roid regimen in the recipient. transplant population. HCV therapy is initiated based on
center-specific protocols and may rely on quantitative NAT
Parasites. Chagas’ disease (Trypanosoma cruzi) has been for HCV ribonucleic acid (RNA) and liver function testing
transmitted by transplantation in endemic areas and more (see Chapter 32).
recently in the US.7,43 Schistosomiasis and Strongyloides Historically, the progression of untreated HIV infection
stercoralis are generally recipient-derived. Malaria and in transplant recipients is rapid. However, HIV-infected
leishmaniasis have been rarely transmitted with allografts. kidneys (and other organs) have been transplanted into
HIV-infected recipients in South Africa and elsewhere.54
Viral Infections Other Than Cytomegalovirus. Under the US HIV Organ Policy Equity (HOPE) Act, organs
EBV infection is a major risk for development of PTLD. from HIV-infected donors may be transplanted into HIV+
The risk is greatest in the EBV-seronegative recipient of recipients as part of a clinical trial with informed consent.55
an EBV-seropositive allograft (i.e., donor seropositive, The first planned HIV+/HIV+ transplant in the US was per-
recipient seronegative, D+/R−).44,45 This situation is formed in March 2016. Based on current criteria for recipi-
most common in pediatric transplant recipients. Other ents who are not HIV infected, donors are evaluated based
at-risk groups include adults coinfected with CMV or on epidemiology and using fourth-generation enzyme-
those receiving greater intensity of immunosuppres- linked immunosorbent assay (ELISA)-NAT testing.
sion, notably with T cell depletion and possibly with Human T cell lymphotropic virus I (HTLV-I) is endemic in
belatacept.46–48 Monitoring should be considered for the Caribbean and parts of Asia (Japan) and can progress to
at-risk individuals using a quantitative molecular assay HTLV-I-associated myelopathy/tropical spastic paraparesis
for EBV.49 EBV is also a cofactor for other lymphoid or to adult T cell leukemia/lymphoma. HTLV-II serologically
malignancies. cross-reacts with HTLV-I, but it is less clearly associated
VZV screening should be used to identify seronegative with disease. Use of organs from such donors is generally
individuals (no history of chickenpox or shingles) for vacci- avoided; however, serologic testing does not distinguish
nation before transplantation. It is likely, although unstud- between the two types of virus. Donor screening for HTLV
ied, that the new herpes zoster subunit vaccine will replace in the US is no longer mandatory, but some centers con-
live vaccine before and after transplantation. HSV screen- tinue donor screening in endemic regions or use targeted
ing is performed by most centers although active infection screening of donors perceived to be high risk.56–58 WNV is a
is prevented by most anti-CMV prophylaxis (except with flavivirus associated with viral syndromes and meningoen-
newer agents such as maribavir and letermovir) during the cephalitis and may be transmitted by blood transfusion and
posttransplant period. VZV serologic status is particularly organ transplantation.7,36,46 Routine screening of donors
important in nonimmune children who may be exposed is not advocated other than in areas with endemic infec-
at school (for antiviral or VZV immunoglobulin prophy- tion. Donors with unexplained changes in mental status or
laxis) and in adults with atypical presentations of infection recent viral illness with neurologic signs should be avoided.
(pneumonia or gastrointestinal disease). Other herpesvi-
ruses also may reactivate, with HHV-6 and HHV-7 serving EVALUATION OF THE TRANSPLANT RECIPIENT
as cofactors for CMV and fungal infections, and Kaposi’s
sarcoma-associated herpesvirus (HHV-8) causing malig- The pretransplant period is useful for obtaining travel,
nancy, notably in endemic regions in South America and animal, environmental, and exposure histories; updating
surrounding the Mediterranean basin. immunizations; and counseling the recipient regarding
Hepatitis screening has changed with quantitative nucleic travel, food, and other infection risks. Ongoing infection
acid test (QNAT) screening and the advent of effective antivi- must be eradicated to the degree possible before transplan-
ral therapies.18,34,50 HBV surface antigen (HBsAg) and HBV tation. Two forms of infection pose a special risk—blood-
core antibody (HBcAb) are used for screening purposes (see stream infection related to vascular access (including that
Chapter 32 for detailed discussion).51 A positive HBV surface for dialysis) and pneumonia, which puts the patient at high
526 Kidney Transplantation: Principles and Practice

risk for subsequent lung infection with nosocomial organ- drug levels and toxicities must be monitored closely. Some
isms. Several other infections are commonly encountered drugs (e.g., raltegravir) have fewer interactions, and the CC
and should be treated and cleared before transplantation. chemokine receptor type 5 (CCR5) receptor antagonist mara-
Peritonitis must be cleared before surgery and infected viroc may even decrease rates of graft rejection.69,70 In vitro
peritoneal dialysis catheters removed. Urinary tract infec- antiretroviral synergy occurs between sirolimus and earlier
tion must be eliminated with antibiotics, with or without CCR5 antagonists; the use of sirolimus-based immunosup-
nephrectomy. Similarly, skin disease threatens the integ- pression may increase rates of graft rejection.71 Prophylaxis
rity of a primary defense against infection and should be against P. jirovecii pneumonia (PJP), and toxoplasmosis in
corrected even if doing so requires the initiation of immu- seronegative recipients of seropositive organs, should be con-
nosuppression (e.g., to treat psoriasis or eczema) before tinued for life, preferably using TMP-SMX.
transplantation. Finally, a history of more than one episode
of diverticulitis should trigger an evaluation to determine
whether colectomy is indicated before transplant.
Among important considerations in transplant recipi-
Selected Infections of Importance
ents are strongyloidiasis, tuberculosis, and AIDS. Strongy- GENERAL CONSIDERATIONS
loides hyperinfection syndrome (hemorrhagic enterocolitis,
pneumonia, gram-negative or polymicrobic bacteremia, The spectrum of infection in the immunocompromised host
or meningitis) may occur more than 30 years after trans- is broad. Given the potential toxicity of antimicrobial agents
plantation. Patients from endemic areas should be screened and the need for rapid interruption of infection, early, spe-
and Strongyloides-seropositive recipients empirically treated cific diagnosis is essential in this population. Whereas
(ivermectin) pretransplant to prevent active disease after initial, empiric therapy is broad by necessity, a rapid nar-
transplant. rowing of the antimicrobial spectrum as data become avail-
The incidence of active and disseminated tuberculosis is able is essential. Advances in diagnostic modalities (e.g., CT
higher in the transplant recipient than in the general popu- or magnetic resonance imaging, molecular microbiologic
lation and the major antituberculous drugs are potentially techniques, high-throughput sequencing) greatly assist in
hepatotoxic and some significantly interact with immu- this process, but the need for invasive procedures cannot be
nosuppressive agents.39–41,59,60 Thus eradication of tuber- overemphasized because they are often required for specific
culosis in transplant candidates before transplantation is microbiologic diagnosis.
preferred. Patients at greater risk of tuberculosis infection Key among decisions in antiinfective therapy is whether
or exposure include individuals with prior history of active to reduce the intensity of immunosuppression, with the
tuberculosis or significant signs of old tuberculosis on chest understanding that the risks of such an approach are graft
radiograph, recent tuberculin reaction conversion, known rejection and/or an aggressive and sometimes detrimental
exposure to active disease, protein-calorie malnutrition, inflammatory response, the immune reconstitution inflam-
cirrhosis, other immunodeficiency, or exposures related matory syndrome (IRIS), perhaps best demonstrated in
to living conditions (e.g., in a shelter or other group hous- patients with cryptococcal meningitis in whom a “rebound”
ing). PPD-positive individuals from endemic regions or with of inflammatory responses may result in worsening symp-
high-risk exposures should be screened for active disease toms and hydrocephalus despite appropriate antimicrobial
and treated for such if present. For those with latent infec- therapy. For latent viral infections or tuberculosis, activa-
tion, therapy should be initiated before transplantation tion is evidence of excessive immunosuppression relative to
although some judgment may be used as to the optimal tim- the host’s immune function. In contrast, for intercurrent
ing of latent treatment. bacterial or fungal infections, reductions in immunosup-
HIV infection has generally been converted from a pro- pression might be reversed when resolution of infection is
gressively fatal disease to a chronic infection controlled by demonstrated. The specific reduction chosen may depend
complex regimens of antiviral agents or highly active anti- on the organisms isolated (e.g., corticosteroids and bacte-
retroviral therapy (HAART). In the pre-HAART era, organ rial infections). Similarly, reversal of some immune deficits
transplantation generally was associated with a rapid pro- (e.g., neutropenia, hypogammaglobulinemia) may be pos-
gression to AIDS. Kidney transplantation in HIV-infected sible with adjunctive therapies (e.g., colony-stimulating
recipients has been associated with good outcomes in indi- factors or antibody therapy). Finally, coinfection with
viduals with controlled HIV infection and with treatment of immunoregulatory viruses such as CMV is common dur-
HCV coinfection.61–65 After transplantation, HAART must ing other active infections and should be screened for and
be continued despite multiple and reciprocal drug interac- treated.
tions between antiretroviral and immunosuppressive medi-
cations. This necessitates experience in HIV and transplant VIRAL PATHOGENS
drug management.66 Rejection will occur more frequently
than in other hosts and standard intensity of immunosup- Cytomegalovirus
pression is required. The most significant interactions are Invasive infection resulting from CMV has become less com-
between the calcineurin and mammalian target of rapamy- mon because of the availability of effective antiviral thera-
cin (mTOR) inhibitors and HIV protease inhibitors (PIs) pies, and diagnostic and monitoring assays for the virus72
and nonnucleoside analog reverse transcriptase inhibitors (see Table 31.6B). Even latent infection or low-level repli-
(NNRTIs) via the hepatic cytochrome P450 3A4 system and cation has important implications for transplant outcomes,
P-glycoprotein.61,67,68 Adjustment in dosages and dosing and strategies used to prevent (universal versus preemptive
intervals of immunosuppressive medications is required, and therapy) and treat infection vary between centers.73 The
31 • Infection in Kidney Transplant Recipients 527

manifestations of CMV infection have been traditionally immunosuppressive therapy is used without antilympho-
termed “direct” and “indirect” effects. More accurate terms cyte antibody induction treatment, approximately 10% to
might be “viremic/cytopathic” effects and “cellular/immu- 15% experience direct CMV syndrome in the absence of pro-
nologic” effects. The common direct effects or clinical syn- phylaxis with a higher rate, up to 50%, after T cell depletion
dromes include: therapies.74
  

CMV syndrome, viremia associated with fever and neu-


□ 
CMV Superinfection. Virus derived from a seropositive
tropenia, variably associated with features of infectious
donor may reactivate in a seropositive recipient (D+/R+).
mononucleosis, including hepatitis, nephritis, lymphad-
Blood transfusions, even if leukocyte-reduced, have a low
enitis, leukopenia, and/or thrombocytopenia
rate (∼4%) of transmission of CMV infection.75 This obser-
Pneumonitis, which is often difficult to distinguish from
□ 
vation gains importance in patients requiring significant
apparently benign secretion
transfusion in the perioperative setting.
Gastrointestinal invasion with esophagitis, colitis, gastri-
□ 

tis, ulcers, bleeding, or perforation


Pathogenesis of Infection. Multiple factors may drive
Hepatitis, pancreatitis, or myocarditis
□ 
CMV activation, including the intensity of immunosuppres-
Meningoencephalitis
□ 
sion (notably pulsed-dosed corticosteroids), the amount
Hemolytic uremic syndrome or microangiopathic throm-
□ 
of virus in the graft, the use of lytic T cell-depleting thera-
bosis
pies, coinfections with other herpesviruses (HHV-6 and
Chorioretinitis and central nervous system (CNS) vasculi-
□ 
HHV-7), and graft rejection. These events share features of
tis or encephalitis
   inflammation and fever, endothelial activation and injury,
The direct clinical manifestations of CMV infection and secretion of proinflammatory cytokines, including
usually occur 1 to 6 months after transplantation in the tumor necrosis factor-α that activates intracellular NF-κB.
absence of prophylaxis, other than chorioretinitis. Cho- NF-κB translocates to the cell nucleus to activate the CMV
rioretinitis can occur at low levels of viral replication and major immediate-early promoter/enhancer and viral
generally later in the posttransplant course. Viremia and replication.76,77
symptomatic infections are rare during effective antiviral The risk of viral activation with graft rejection is most
prophylaxis. CMV activation may develop despite prophy- prominent in the D+/R− combination with the alloimmune
laxis during intensification of immunosuppression (e.g., for response producing graft inflammation and injury, gener-
rejection). ally the site of greatest viral load and MHC mismatch, inten-
The cellular and immunologic effects of CMV infection sification of immunosuppression, systemic inflammation
(discussed later) are the result of the suppression of a vari- with fever, and enhanced MHC display in the allograft. Thus
ety of host defense mechanisms and predispose to second- a bidirectional linkage exists between CMV replication and
ary invasion by P. jirovecii, Candida and Aspergillus species, graft rejection. Reinke et al.67 showed that 17 of 21 patients
and other bacterial and fungal pathogens.73 CMV infection with biopsy evidence of late acute rejection demonstrated a
also contributes to the risk for graft rejection, PTLD, accel- response to antiviral therapy. Similarly, Lowance and col-
eration of HCV coinfection, HHV-6 and HHV-7 infections, leagues have demonstrated that preventing CMV infection
and increased risk for death. also resulted in a lower incidence of graft rejection.73,78 The
cellular and immunologic effects of CMV (“indirect effects”)
Patterns of Transmission. Transmission of CMV in the are as important to the immunocompromised host as inva-
transplant recipient occurs in one of three patterns: pri- sive viral infection. The mechanisms for these effects are
mary infection, reactivation infection, and superinfection. complex and relate to viral strategies to evade the host’s
responses and allow CMV-infected antigen-presenting cells
Primary CMV Infection. The greatest risk for CMV infec- to travel throughout the host and spread the virus.
tion is in the setting of seronegative individuals receiving
grafts from latently infected, seropositive donors (D+/R−), Diagnosis. Clinical management of CMV, including pre-
with reactivation of virus within the graft. Over 50% of vention and treatment, is based on understanding the
these patients become viremic in the absence of prophy- causes of CMV activation and the available diagnostic tech-
laxis, often without symptoms. Many become viremic after niques. CMV cultures are no longer essential for resistance
cessation of antiviral prophylaxis, with symptomatic “late testing, which can be performed by molecular sequenc-
infection” occurring in up to one-third of recipients previ- ing.72,79,80 Positive CMV cultures (or shell vial culture)
ously treated with prophylaxis.74 Primary CMV infection is derived from respiratory secretions or urine are of little diag-
often severe and may also occur in seronegative individuals nostic value—many immunosuppressed patients secrete
such as in children, after transfusion, or via sexual contacts CMV in the absence of invasive disease. Serologic tests are
in the community. The allograft may be a privileged site for useful before transplantation to predict risk but are of lit-
viral replication because the MHC-restricted, virus-specific, tle value after transplantation in defining clinical disease,
cytotoxic T cells have decreased ability to eliminate virally because seroconversion is generally delayed. Seroconver-
infected cells in the presence of donor and recipient MHC sion to positive CMV IgG provides evidence that the patient
mismatch. has developed some degree of immunity and appears to cor-
relate generally with T cell function.
Reactivation CMV Infection. In reactivation infec- Quantitation of the intensity of CMV viral burden has
tion, seropositive individuals reactivate endogenous virus been linked to the risk for infection in transplant recipi-
after transplantation (D+ or D−/R+). When conventional ents.74,81–84 Two types of quantitative assays have been
528 Kidney Transplantation: Principles and Practice

developed: molecular and antigen detection assays. The therapy incurs extra costs for monitoring and coordination
antigenemia assay is labor-intensive and, being semiquan- of outpatient care, while reducing the cost of drugs and the
titative, it is less commonly used than NAT; circulating neu- inherent toxicities. Prophylaxis has the possible advantage
trophils are immunostained for CMV early antigen (pp65) of not only preventing CMV infection during the period
that is taken up nonspecifically as a measure of the total of greatest risk but also diminishing infections secondary
viral burden. QNAT is generally used for diagnosis, preemp- to HHV-6, HHV-7, and EBV. The indirect effects of CMV
tive management strategies, and monitoring response to (i.e., graft rejection, herpesvirus infections, opportunistic
therapy.85–89 CMV deoxyribonucleic acid (DNA) is gener- infections) also may be reduced by routine prophylaxis.
ally detected earlier in whole blood compared with plasma; In practice, neither universal prophylaxis nor preemp-
one specimen type and one laboratory should be used when tive therapy is perfect. Many centers use a combination of
monitoring patients. The highest viral loads often are asso- both approaches: universal prophylaxis for the highest-risk
ciated with tissue-invasive disease; the lowest are associated recipients (D+/R− and R+ with T cell depletion) and pre-
with asymptomatic CMV infection. Multiple viral strains emptive therapy for others. Infrequently, breakthrough
are often present during infection. Viral loads present dur- disease and ganciclovir resistance have been observed with
ing CMV syndrome vary widely. A single quantitative assay both approaches.
should be used consistently for monitoring each patient. A Given the risk of invasive infection, patients at risk of
World Health Organization (WHO) International Reference primary infection from the donor (CMV D+/R−) and sero-
Standard became available in 2010 from a clinical isolate positive patients receiving depleting anti–T-lymphocyte
(Merlin) with a titer of 5×106 IU/mL. All laboratory tests antibodies are generally given prophylaxis for 3 to 6 months
should be calibrated to the WHO International Standard after transplantation. Other groups are candidates for pre-
with results reported as IU/mL.90 One source of variability is emptive therapy if an appropriate monitoring system is in
different DNA extraction methods. Multiple viral genotypes place and patient compliance is good. Current data support
may be observed, often in patients with more severe disease the use of universal prophylaxis (not preemptive therapy)
and in antiviral resistance. Resistance testing should be in the prevention of indirect effects of CMV infection, includ-
performed in the face of poor clinical and virologic response ing PTLD, opportunistic infections, allograft rejection, and
to adequate therapy for at least 10 to 14 days; responses mortality.73 Increasingly, “late” disease has been observed
may be delayed with high-level viremia. Multiple risk fac- after the completion of prophylaxis.92,93 Thus monitoring
tors for drug resistance often coexist, including prolonged may be useful after prophylaxis. The rate of late disease var-
antiviral drug exposure; notably with inadequate dosing, ies but is thought to be as high as 17% to 37% in D+/R−
lack of immunity, high-level viral replication; and intensive recipients, supporting the value of prophylaxis for 6 months
immunosuppression. in D+/R− renal recipients (the IMPACT study).93
The advent of quantitative assays for the diagnosis and Options for CMV prophylaxis include valganciclovir (900
management of CMV infection has allowed noninvasive mg orally once daily), oral ganciclovir (1 g three times
diagnosis in many patients with two important exceptions: daily), intravenous ganciclovir (5 mg/kg once daily), or
  
high-dose oral valacyclovir (2 g four times daily)—each
1. Neurologic disease, including chorioretinitis
corrected for renal function. Valganciclovir and ganciclo-
2. Gastrointestinal disease, including invasive colitis and
vir are associated with neutropenia; however, dose reduc-
gastritis.
   tion risks breakthrough viremia and the emergence of viral
In these syndromes, CMV viral loads are often low or resistance. Given changing renal function after transplan-
below detection limits. For the diagnosis of gastrointestinal tation and the costs of medication, many regimens employ
CMV disease, demonstration of CMV inclusions in tissues lower doses of valganciclovir.94,95 Such regimens should be
and/or immunohistology for CMV antigens remain essen- coupled with monitoring to assure efficacy. After comple-
tial. The central role of assays is illustrated by the approach tion of treatment for CMV disease (see later), many centers
to the management of CMV risk (see Table 31.6B). The initiate a course of secondary prophylaxis (1–3 months).
schedule for screening is linked to the risk for infection. In An alternative is a strategy of virologic monitoring during
the high-risk patient (D+/R− or R+ with antilymphocyte this period.
globulin), after the completion of prophylaxis, monthly
screening is performed to ensure the absence of viremia for Treatment. The standard of care for treating invasive CMV
3 to 6 months. In the patient being treated for CMV infec- disease is 2 to 4 weeks of intravenous (IV) ganciclovir (5
tion, the assays provide an endpoint for therapy and the ini- mg/kg twice daily, with dosage adjustments for renal dys-
tiation of prophylaxis. function) until a quantitative assay for CMV is negative.91
In patients with mild to moderately severe symptoms, val-
Cytomegalovirus Prevention. Prevention of CMV infec- ganciclovir (900 mg orally twice daily corrected for renal
tion must be individualized for immunosuppressive regi- function) may be used as an alternative. In symptomatic
mens and the patient (see Table 31.6B).74,91 Two strategies patients slow to respond to therapy and who are seronega-
are commonly used for CMV prevention—universal pro- tive, the addition of CMV hyperimmune globulin (150 mg/
phylaxis and preemptive therapy.84 Universal prophylaxis kg/dose IV monthly) for 3 months is controversial, costly,
involves giving antiviral therapy to all at-risk patients after and of uncertain benefit. Relapse does occur, primarily in
transplantation for a defined period. In preemptive therapy, seronegative patients, in those with high viral burdens, if
QNAT assays are used to monitor patients at predefined not treated to the achievement of a negative quantitative
intervals (generally weekly for weeks 1–12) to detect assay, and in gastrointestinal disease treated with an oral
early disease. Positive assays result in therapy. Preemptive regimen. Repeat endoscopy may be considered with poor
31 • Infection in Kidney Transplant Recipients 529

clinical response, or if other processes are present (ischemia, Epstein–Barr Virus


cancer). In practice, it is reasonable to initiate therapy with EBV is a ubiquitous herpesvirus that infects B lymphocytes.
intravenous ganciclovir, monitor weekly to assure a viro- In immunosuppressed transplant recipients, primary EBV
logic response and treat until after monitoring is negative infection (and relapses in the absence of antiviral immu-
(often two negative weekly assays). Such patients may ben- nity) causes a mononucleosis-type syndrome, generally
efit from 2 to 4 months of oral valganciclovir (900 mg daily manifesting as a lymphocytosis (B cell) with or without
based on creatinine clearance) administered as secondary lymphadenopathy or pharyngitis.106–108 Meningitis,
prophylaxis after the completion of intravenous therapy. hepatitis, and pancreatitis also are observed. Remitting-
This approach has resulted in rare symptomatic relapses relapsing EBV infection is common in children and may
and has been associated uncommonly with the emergence reflect the interplay between evolving antiviral immu-
of antiviral resistance. It may be worth measuring a formal nity and immunosuppression.109–111 Regardless of its
creatinine clearance to assure adequate dosing. mode of expression, this syndrome should suggest relative
The incidence of ganciclovir resistance in CMV is gen- overimmunosuppression.
erally low.79,80 The risk of resistance is greatest in D+/R− EBV also plays a central role in the pathogenesis of
recipients with higher viral loads, who received inadequate PTLD.106,112 PTLD represents a spectrum of disease from
dosing of prophylactic or therapeutic ganciclovir, more benign B cell mononucleosis-like syndrome to monomor-
intensive immunosuppression including antilymphocyte phic B cell lymphoma and tumors of T cell, natural killer
antibody induction, and with prolonged antiviral prophy- cell, and null-cell origins (Fig. 31.3).113,114 The most clearly
laxis. Clinically, the patient’s viral load or clinical syndrome defined risk factor for PTLD is primary EBV infection, which
fails to respond to appropriate therapy, including a reduc- increases the risk for PTLD by 10- to 76-fold. PTLD may
tion in immunosuppression over 10 to 14 days. Genetic occur in the absence of EBV infection or in seropositive
resistance testing is useful in managing resistant CMV infec- patients, and the role of EBV in the pathogenesis of non-B
tion. Many mutations have now been identified, including cell tumors is less clear.45 Other risk factors include CMV
mutations in the viral UL97 (thymidine kinase) or UL54 coinfection, T cell depletion therapy, duration of immuno-
(DNA polymerase) genes that confer ganciclovir resis- suppression, and, in adults, older age. Lymphomas consti-
tance. Some of the common mutations in the UL97 gene tute 15% of tumors among adult transplant recipients (51%
respond to higher doses of intravenous ganciclovir. Com- in children) with mortality of 40% to 60%. Many deaths are
bined mutations (UL97 and UL54) may manifest high-level associated with allograft failure after withdrawal of immu-
resistance to ganciclovir. Alternative therapies have been nosuppression during treatment of malignancy.
available in intravenous form only. These include foscarnet Compared with the general population, PTLD has
and cidofovir. Foscarnet is active against many ganciclovir- increased extranodal involvement, poor response to con-
resistant strains of CMV, although associated with marked ventional therapies, and poor outcomes. The spectrum of
magnesium and potassium wasting, seizures (notably with disease is broad, as noted previously.81 EBV-negative PTLD
calcineurin inhibitor therapy), and renal toxicity. Cidofovir has been described, and T cell PTLD has been shown in
may also be used, but often incurs significant nephrotoxic- allografts thought to have rejection or other viral infection.
ity and ocular toxicity. Viral polymerase UL54 mutations PTLD late (>1–2 years) after transplantation is more often
may cause resistance to foscarnet and cidofovir depending EBV-negative in adults.115
on the nature of the mutation. Multiple courses of antivi- The clinical presentations of EBV-associated PTLD vary
ral therapy may be needed to cure resistant CMV infection. widely and include:
Given the toxicity of available medications, newer drugs are   

becoming available. Unexplained fever (fever of unknown origin) with


□ 

A new agent, letermovir, has demonstrated promise for viremia


the management of CMV infection in stem cell transplant A mononucleosis-type syndrome, with fever and malaise,
□ 

recipients. Letermovir appears to have good oral bioavail- with or without pharyngitis or tonsillitis (often diagnosed
ability, and a low rate of adverse effects and drug–drug incidentally in tonsillectomy specimens); often no lym-
interactions.96,97 Letermovir exerts its antiviral effect by phadenopathy is observed
interfering with the viral pUL56 gene product to disrupt the Gastrointestinal bleeding, obstruction, or perforation
□ 

viral terminase complex.98 The addition of hyperimmune Abdominal mass lesions


□ 

globulin may be beneficial. Most centers try to reduce over- Infiltrative disease of the allograft
□ 

all immunosuppression during therapy. Alternative agents Hepatocellular or pancreatic dysfunction


□ 

include the dihydroorotate dehydrogenase inhibitors (leflu- CNS mass lesions.


□ 

nomide) approved for immunosuppression in treatment


of rheumatologic diseases with useful, incidental activ- Diagnosis. Serologic testing is not useful for the diagnosis
ity against CMV (and possibly BK polyomavirus).99–101 of acute EBV infection or PTLD in transplantation. Quanti-
Effective use requires monitoring of drug levels and liver tative EBV viral load testing is required for the diagnosis and
function tests. In phase III trials, maribavir, a competitive management of EBV-positive PTLD.106,107,116 Serial assays
inhibitor of the pUL97 protein, appeared to be safe, but effi- are more useful in an individual patient than specific viral
cacy in CMV prevention was suboptimal.102–104 CMX001 load measurements. Some data suggest that assays using
is a broad-spectrum oral lipid antiviral conjugate of cido- unfractionated whole blood are preferable to plasma sam-
fovir with activity against adenovirus as well as CMV; ples for EBV viral load surveillance. The diagnosis of PTLD
the drug has good bioavailability and some dose-limiting may be suggested by the presence of a compatible clini-
diarrhea.105 cal syndrome with demonstration of EBV viral load. WHO
530 Kidney Transplantation: Principles and Practice

standards now exist for EBV viral load assays; however, and obstruction. In such patients, the etiologies of decreased
assays are not directly comparable between laboratories renal function must be carefully evaluated (e.g., mechani-
given laboratory-specific differences in performance.117,118 cal obstruction, drug toxicity, pyelonephritis, rejection,
Trends in individual patients over time using a single assay thrombosis, recurrent disease), and choices must be made
are most useful. The demonstration of EBV-specific nucleic between increasing immunosuppression to treat suspected
acids in tissues may diagnose EBV-associated PTLD. RNA graft rejection or reducing immunosuppression to allow the
in situ hybridization against EBV-encoded small nuclear immune system to control infection.131,132 Patients with
RNAs is more sensitive than the detection of viral DNA. The BKV nephropathy treated with increased immunosuppres-
EBV-latent antigens EBNA-1, EBNA-2, and LMP-1 can be sion have a high incidence of graft loss. Reduced immuno-
detected by immunohistochemistry.119 suppression may stabilize renal allograft function but risks
graft rejection. Other clinical syndromes may be associated
Management. Clinical management depends on the stage with BKV, including pneumonitis, hemophagocytic syn-
of disease. In the polyclonal form, particularly in children, drome, encephalitis, or PML.
reestablishment of immune function may suffice to cause Risk factors for BKV nephropathy include high-dose
PTLD to regress.120 At this stage, it is possible that antivi- immunosuppression (particularly T cell depletion, tacro-
ral therapy might have some utility given the viremia and limus, and mycophenolate mofetil), pulse-dose steroids
role of EBV, and of CMV, if present, as an immunosuppres- for treatment of (presumed) graft rejection, ischemia-
sive agent.121 With progression of disease to extranodal reperfusion injury, increased number of human leukocyte
and monoclonal malignant forms, reduction in immuno- antigens (HLA) mismatches between donor and recipient,
suppression may be useful, but alternative therapies are and the intensity of viremia in the pathogenesis of disease.
often required. In kidney transplantation, the failure to Renal retransplantation for PyVAN is a risk factor for rein-
regress with significant reductions in immunosuppression fection.133,134 JCV may also cause nephropathy. The role of
may suggest the need to sacrifice the allograft for patient specific immunosuppressive agents has not been confirmed.
survival. Combinations of anti–B cell therapy (anti-CD20, The greatest incidence of BKV nephropathy is at centers
rituximab), chemotherapy (CHOP: cyclophosphamide, with the most intensive immunosuppressive regimens.
hydroxydaunomycin, vincristine, prednisone), irradiation
especially for CNS tumors, or adoptive immunotherapy Screening, Prevention, and Diagnosis. BKV infection
with stimulated T cells have been used.122–125 is generally asymptomatic. Renal tubular cell injury in
PyVAN is reflected in a rising serum creatinine. Most cen-
Polyomaviruses ters have developed screening programs to document early
Polyomaviruses have been identified in transplant recipi- disease. The use of urine cytology to detect the presence
ents in association with tubulointerstitial nephritis and of infected decoy cells in the urine has high sensitivity for
nephropathy (PyVAN with BK virus [BKV], and JC virus BKV infection but a low (29%) predictive value for PyVAN.
[JCV]) and ureteric stenosis (BKV), and in association with Detection of urine BKV by electron microscopy, urine BKV
demyelinating disease of the brain (JCV-associated progres- viral (DNA) loads greater than 7 log gEq/mL or BKV VP1
sive multifocal leukoencephalopathy [PML]). Less com- gene mRNA of >6 log copies/ng total urine RNA are use-
monly, polyomaviruses have been identified in association ful diagnostically, keeping in mind that BKV molecular tests
with trichodysplasia spinulosa (TSV), in some malignancies vary depending on the target and the matrix tested. Patients
(Merkel cell carcinoma [MCV]), and in condylomas. Tis- with BKV nephropathy tend to have higher plasma viral
sue receptors for these human viruses are ubiquitous. The loads; in one study >7700 BKV copies per mL of plasma, P
seroprevalence in adults for all polyomaviruses ranges from < 0.001, 50% positive predictive value, 100% negative pre-
40% to 90%. BKV resides in latency in renal tubular epi- dictive value compared with patients without disease.135
thelial cells. JCV also has been isolated from renal tissues A high serum BKV viral load is considered a basis for
but has preferred tropism for neural tissues. Reactivation reduction in immunosuppression, especially if serum creati-
occurs with immunodeficiency and immunosuppression nine has risen. The diagnosis should be made by the demon-
and tissue injury (e.g., ischemia-reperfusion). stration of BKV cytopathic changes with cellular infiltration
consistent with the diagnosis of interstitial nephritis in the
BK Polyomavirus Infection. BKV is associated with a allograft and by immunohistology for BKV proteins, or by
range of clinical syndromes in immunocompromised hosts, in situ hybridization for BKV nucleic acids in a renal biopsy.
including viruria and viremia, ureteral ulceration and ste- There is a semiquantitative scoring system for histologic
nosis, and hemorrhagic cystitis.126–130 Active infection of changes of PyVAN.135 For immunohistochemistry, cross-
renal allografts has been associated with progressive loss reacting antibodies against the large T antigen of the sim-
of graft function (BKV nephropathy) in approximately ian virus 40 or antibodies against BKVVP1 or agnoprotein
4% (range 1%–8%) of kidney transplant recipients; this is have been used. PyVAN is characterized by intranuclear
referred to as PyVAN (polyomavirus-associated nephropa- polyomavirus inclusion bodies in tubular epithelial and/or
thy). BKV nephropathy is rarely recognized in recipients of glomerular cells. Fibrosis is often prominent, occasionally
extrarenal organs. The clinical presentation of disease is with calcification. PyVAN is often focal, with false-negative
usually as asymptomatic, sterile pyuria, reflecting shedding biopsies in some cases. Graft rejection may accompany
of infected tubular and ureteric epithelial cells. These cells PyVAN, and complicates both diagnosis and management.
contain sheets of virus and are detected by urine cytology Recommendations regarding screening for BKV infection
as “decoy cells.” In some cases, the patient presents with vary, but generally suggest testing once every 3 months
diminished renal allograft function or with ureteric stenosis during the first 2 years after transplantation, and at least
31 • Infection in Kidney Transplant Recipients 531

annually for years 2 to 5. A urinary test for BKV (cytology (0.25–1 mg/kg every 2 weeks). Significant renal toxicity may
for decoy cells or urine BKV viral load over 7 log gEq/mL) is be observed with cidofovir despite probenecid which may
adequate for screening. Patients with high urinary BKV viral decrease efficacy. Leflunomide, an immunosuppressant used
loads require testing for plasma BKV DNA. Screening can in rheumatoid arthritis, and fluoroquinolones have some
also be performed using plasma BKV DNA loads. For patients anti-BKV activity in vitro, but little efficacy in vivo. Repletion
with plasma BKV DNA loads of >4 log10 gEq/mL on dupli- of serum immunoglobulins may be considered.
cate testing 2 to 3 weeks apart, a presumptive diagnosis of Retransplantation has been successful in PyVAN patients
PyVAN should be made and immunosuppression reduced with failed allografts, possibly as a reflection of immunity
(see later). If screening is performed by plasma viral load, developing after reduction of immunosuppression. Some
the interval between screening assays should be reduced to centers allow retransplantation after immunosuppression
monthly for the first 6 months posttransplant. This reflects has been discontinued for some period (e.g., 6 months)
the faster onset of permanent renal injury in patients with and BKV is undetectable in blood and low in urine. Surgi-
circulating viremia compared with urinary excretion. cal removal of the allograft does not protect against future
BKV infection or PyVAN but may be needed if immunosup-
Treatment. There is no accepted treatment for PyVAN pression cannot be reduced (double transplants, allosensi-
other than reduction in the intensity of immunosuppres- tization) and/or elevated viral loads persist. In the future,
sion. It is useful to monitor the response to such maneuvers measurements of BKV-specific cellular immunity after dis-
using plasma viral load measurements.135 Despite contro- continuation of immunosuppression may help determine
versy, it is reasonable to reduce dosing of both calcineurin the optimal time for retransplantation.
inhibitors and antimetabolites in a stepwise fashion while
monitoring BKV plasma loads. Given the toxicity of calci- JC Virus
neurin inhibitors for tubular cells, the role of renal tubular Infection of the CNS by JC polyomavirus has been observed
injury in the activation of BKV, and the need for anti-BKV uncommonly in transplant recipients as PML (see Fig. 31.3).
T cell activity, these agents should be included in initial This infection may present with focal neurologic deficits or
reductions. General targets include tacrolimus trough lev- seizures and more slowly progressive neurologic lesions and
els of <6 ng/mL, cyclosporine trough levels <150 ng/mL, may progress to death after extensive demyelination. PML
sirolimus trough levels of <6 ng/mL, and/or mycopheno- may be confused with calcineurin neurotoxicity; both may
late mofetil daily dose equivalents of ≤1000 mg. Regard- respond to a reduction in drug levels. No proven therapies
less of the approach, renal function (at least 1–2 times per exist, although reduction of immunosuppression is com-
week), drug levels, and viral loads (alternate weeks) must monly employed, an analogy to immune reconstitution in
be monitored carefully during reductions. Rebiopsy may be AIDS patients with PML.
needed for poor virologic and clinical responses.
The use of adjunctive antiviral therapies remains con- Human Papillomavirus
troversial. Some centers advocate the use of cidofovir, with Human papillomavirus (HPV) infection is associated
or without probenecid, for BKV nephropathy in low doses with anogenital and skin precancers, cancers, and warts

A B
Fig. 31.3 (A) Central nervous system lymphoma with positive staining for Epstein–Barr virus (posttransplantation lymphoproliferative disorder) in a
63-year-old man 6 years postrenal transplantation. (B) Acute stroke syndrome in a 65-year-old man with progressive multifocal leukoencephalopathy.
532 Kidney Transplantation: Principles and Practice

associated with human HPV infections. Anogenital warts infection—tissue infarction, hemorrhage, and systemic
should be considered a marker for possible carriage of risk dissemination with metastatic invasion. Soon after trans-
types for cervical and anal cancers including HPV16 and plantation, CNS fungal infection is most often a result of
18. Routine gynecologic and skin screening is mandatory Aspergillus; 1 year or later after transplantation, other fungi
for transplant recipients. (Mucorales, dematiaceous fungi) become more prominent.
The drug of choice for documented Aspergillus infection is
Human Immunodeficiency Virus voriconazole, despite its significant interactions with calci-
Successful organ transplantation in HIV-infected individu- neurin inhibitors and rapamycin.138,139 Isavuconazole, an
als (discussed earlier) has been achieved with effective anti- azole antifungal with activity against yeasts, Aspergillus,
retroviral therapies and full transplant immunosuppression and some Mucorales, has been shown to be safe and effica-
including appropriate T cell depletion therapy. Rejection rates cious in solid-organ transplant recipients although it may
remain higher than in non-HIV-infected groups. Management require dose adjustment and therapeutic drug monitoring
requires careful tracking of immunosuppressive drug lev- of immunosuppressive agents.140–142 Liposomal ampho-
els, avoidance of protease inhibitors in antiretroviral therapy tericin is an effective and fungicidal alternative, though a
(ART) selection if possible, and knowledge of HIV susceptibility more toxic alternative in renal transplantation and should
patterns. Prophylaxis for opportunistic infections depends on be reserved for cases in which azoles are contraindicated
the epidemiology of the individual (e.g., mycobacteria, Coccidi- or not tolerated. Although not as toxic as amphotericin
oides, or Histoplasma exposures) and routine therapies. Donors B preparations, echinocandins can be used in situations
with HIV or HCV infections are used at some centers with where azole and polyene antifungals are contraindicated,
expertise in antiviral management in transplantation. but they are not recommended as primary therapy for inva-
sive aspergillosis, including renal infections. Combination
therapy using voriconazole and anidulafungin has been
FUNGAL INFECTIONS
studied in the hematologic malignancy population but did
Transplant recipients are at risk for opportunistic infections not show a survival advantage. Surgical debridement is
with a variety of fungal pathogens, the most important of sometimes required for successful clearance of such inva-
which are Candida, Aspergillus, and Cryptococcus neofor- sive infections.138,139
mans, but also including the endemic mycoses.
Central Nervous System Infections and
Candida Cryptococcus Neoformans
The most common fungal pathogen in transplant patients is CNS infection in the transplant recipient may result from a
Candida, with more than 50% being non-albicans species.136 broad spectrum of organisms.2,143 Infections are often met-
Mucocutaneous candidal infection (e.g., oral thrush, esoph- astatic to the CNS from the blood and lungs. Viral etiologies
ageal infection, cutaneous infection at intertriginous sites, include CMV (nodular angiitis), HSV meningoencephalitis,
candidal vaginitis) is most common in diabetics, with high- JCV (progressive multifocal leukoencephalopathy), and
dose steroid therapy, and during broad-spectrum antibacte- VZV. Locally epidemic pathogens (WNV, Eastern equine
rial therapy. These infections are usually treatable through encephalitis) also must be considered. Bacterial pathogens
correction of the underlying metabolic abnormality and can include S. pneumoniae, Borrelia burgdorferi (Lyme dis-
topical therapy with clotrimazole or nystatin (see Table ease), L. monocytogenes, tuberculosis, Nocardia, and occa-
31.6C). Thrush also may complicate viral (HSV, CMV) or sionally Salmonella. Brain and epidural abscesses have
toxic (drugs including mycophenolate mofetil) esophagi- been observed and may be particularly problematic when
tis. Optimal management of Candida infection occurring in secondary to an antibiotic-resistant pathogen. As noted
association with surgical drains or vascular access cathe- earlier, fungi may be metastatic from lungs but also may
ters requires removal of the foreign body and systemic anti- spread from sinuses, skin, and the blood. Parasites with
fungal therapy with fluconazole or echinocandin. A single potential for CNS involvement include Toxoplasma gondii
positive blood culture result for Candida species necessitates and Strongyloides stercoralis.
systemic antifungal therapy; this finding carries a signifi- Given the spectrum of etiologies, precise diagnosis is essen-
cant risk of dissemination in this population. tial (Table 31.8). A reasonable empirical regimen would
A special problem in kidney transplant recipients is can- treat pneumococcus and Haemophilus influenzae (ceftriaxone
diduria, including in asymptomatic patients. Notably in and vancomycin), Listeria (ampicillin), Cryptococcus (flucon-
individuals with poor bladder function, obstructing fungal azole or lipid formulation of amphotericin), and HSV (acyclo-
balls can develop at the ureteropelvic junction, resulting in vir) while awaiting data (lumbar puncture, blood cultures,
obstructive uropathy, ascending pyelonephritis, and the and radiographic studies). Noninfectious etiologies, includ-
possibility of systemic dissemination. ing calcineurin inhibitoOO)r toxicity, lymphoma, and meta-
static cancer, should be included in the differential diagnosis.
Aspergillus Molecular (HSV, VZV, Toxoplasma) and antigen (cryptococ-
Invasive aspergillosis is a medical emergency in the trans- cal) assays on cerebrospinal fluid and biopsy (for noninfec-
plant recipient, with the portal of entry being the lungs tious etiologies) may be needed for diagnosis.
and sinuses in more than 90% of patients and the skin in
most of the others. The predominant species depends on the Cryptococcus Neoformans. Cryptococcal infection is
clinical center and prior azole exposure.137 The pathologic rarely seen in the transplant recipient until more than
hallmark of invasive aspergillosis is blood vessel invasion, 6 months after transplantation. In the relatively intact
which accounts for the three clinical characteristics of this transplant recipient, the most common presentation of
31 • Infection in Kidney Transplant Recipients 533

TABLE 31.8 Cerebrospinal Fluid Analysis in


Transplantation
Opening pressure
Cell count with differential
Glucose and total protein concentrations
Gram stain and bacterial culture
India ink (or other fungal stain) and fungal culture
Viral culture (save sample if indicated)
Cryptococcal polysaccharide antigen
Fungal culture
Also consider: Histoplasma polysaccharide antigen (urine)
Also consider: Coccidioides immitis complement fixation antibodies
(serum or CSF)
NUCLEIC ACID DETECTION (IN CLINICAL CONTEXT)
Herpes simplex virus 1 and 2
Varicella-zoster virus
Epstein–Barr virus
Cytomegalovirus
Human herpesvirus 6
JC virus
Enterovirus Fig. 31.4 Cryptococcus neoformans meningitis—India ink preparation
of cerebrospinal fluid from a 53-year-old man, 16 months after renal
Toxoplasma gondii (CSF or serum)
transplantation, who presented with confusion and 7th nerve palsy
CYTOLOGY AND FLOW CYTOMETRY (CONSIDER FOR PTLD) without fever or meningismus.

CSF, cerebrospinal fluid; PTLD, posttransplant lymphoproliferative disorder.

cryptococcal infection is that of an asymptomatic pulmo- including Africa, Asia, Latin America, and the Caribbean
nary nodule with the organism present. In the “chronic should be screened with Strongyloides IgG serology before
ne’er-do-well” patient, pneumonia and meningitis are com- transplantation and should be treated with ivermectin pre-
mon, with skin involvement at sites of tissue injury (cath- emptively if seropositive.
eters) and prostate or bone involvement also reported.
Cryptococcosis should be suspected in transplant recipi- Pneumocystis and Fever With Pneumonitis
ents who present more than 6 months after transplantation The spectrum of potential pulmonary pathogens in the
with unexplained headaches (especially when accompa- transplant recipient is broad. Some general concepts are
nied by fevers), decreased state of consciousness, failure to worth consideration. The depressed inflammatory response
thrive, or unexplained focal skin disease (which requires of the immunocompromised transplant patient may greatly
biopsy for culture and pathologic evaluation).144–146 Diag- modify or delay the appearance of a pulmonary lesion on
nosis is often achieved by serum cryptococcal antigen detec- radiograph. Focal or multifocal consolidation of acute
tion, but all such patients should have lumbar puncture for onset is likely to be caused by bacteria. Similar multifo-
cell count, Gram stain, culture, India ink preparation, and cal lesions with subacute to chronic progression are more
cryptococcal antigen studies (Fig. 31.4). Initial treatment likely secondary to fungi, tuberculosis, or Nocardia. Large
for CNS disease is liposomal amphotericin and 5-flucytosine nodules are usually a sign of fungal or Nocardia infection.
(monitoring serum levels) followed by high-dose flucon- Subacute disease with diffuse abnormalities, either of the
azole. Extended courses of fluconazole suppression may be peribronchovascular type or miliary micronodules, are
required for patients based on clinical progress or net degree usually caused by viruses (especially CMV) or Pneumocys-
of immunosuppression.147,148 A gradual tapering of immu- tis. Additional clues can be found by examining pulmonary
nosuppression should also be considered with initiation of lesions for cavitation, which suggests necrotizing infection
antifungal therapy, recognizing that IRIS, the clinical man- as may be caused by fungi (Aspergillus or Mucoraceae),
ifestations that mimic worsening cryptococcal disease, may Nocardia, Staphylococcus, and certain gram-negative bacilli,
occur in patients for whom reduction in immunosuppres- most commonly Klebsiella pneumoniae and Pseudomonas
sion has been too rapid.149–151 This ultimately may require aeruginosa.
adjunctive use of corticosteroids as IRIS or scarring may CT of the chest is useful when the chest radiograph is
cause obstruction, with increased cerebrospinal fluid pres- negative or when the radiographic findings are subtle or
sure and hydrocephalus. nonspecific. CT is also essential to determine the extent of
the disease process, to discern the possibility of a simultane-
Strongyloides Stercoralis. With immunosuppressive ous process (superinfection), and to select the optimal pro-
therapy, S. stercoralis infection may activate more than 30 cedure for achieving a pathologic diagnosis.
years after initial exposure. Such reactivation can result in
either diarrheal illness or parasite migration with hyperin- Pneumocystis Pneumonia. The risk of infection with PJP
fection syndrome (characterized by hemorrhagic entero- is greatest in the first 6 months after transplantation (up
colitis, hemorrhagic pneumonia, or both) or disseminated to 10% without TMP-SMX or other prophylaxis) and dur-
infection with accompanying (usually) gram-negative bac- ing periods of increased immunosuppression.152–155 There
teremia or meningitis. Immigrants, refugees, travelers to is a continued risk of infection in three overlapping groups
and military personnel stationed in hyperendemic regions of transplant recipients: (1) recipients who require higher
534 Kidney Transplantation: Principles and Practice

than normal levels of immunosuppression (notably corti- atovaquone, and inhaled or intravenous pentamidine, are
costeroids) for prolonged periods because of poor allograft less effective than TMP-SMX but are useful in patients with
function or chronic rejection; (2) recipients with chronic significant allergy to sulfa drugs. The advantages of TMP-
CMV infection; and (3) recipients undergoing treatments SMX include increased efficacy; lower cost; availability of
(e.g., cancer chemotherapy, drug toxicity) that increase the oral preparations; and possible protection against other
level of immunodeficiency or neutropenia. The expected organisms, including T. gondii, Isospora belli, Cyclospora
mortality secondary to PJP is increased in patients on cyclo- cayetanensis, many Nocardia species, and common urinary,
sporine compared with other immunocompromised hosts. respiratory, and gastrointestinal bacterial pathogens. Alter-
The hallmark of PJP is the presence of marked hypox- native agents lack this spectrum of activity.
emia, dyspnea, and cough with a paucity of physical or
radiologic findings. In the transplant recipient, PJP is Urinary Tract Infection
generally acute to subacute in development. Atypical Most urinary tract infections occur in the first year after
Pneumocystis infection (radiographically or clinically) kidney transplant.161,162 A subset of patients experience
may be seen in patients who have coexisting pulmonary recurrent disease and may suffer from pyelonephritis or
infections or who develop disease while receiving prophy- bacteremia. Urinary tract infection beyond 6 months after
laxis with second-line agents (e.g., pentamidine or ato- transplantation is associated with reduced renal graft sur-
vaquone). Patients presenting with PJP outside the usual vival and increased mortality.163 The risk of urinary tract
highest risk period may have indolent disease that can be infection after renal transplant is increased in women,
confused radiographically with heart failure. mTOR inhib- with prolonged bladder catheterization, with increased
itors (sirolimus and everolimus) have also been associated intensity of immunosuppression, in recipients of deceased
with the development of interstitial lung disease that can donor grafts, and, possibly, with vesicoureteral reflux. The
be confused radiographically and clinically with PJP.156 risk for vesicoureteral reflux is dependent in part on the
Further, two case-control studies reported an associa- surgical approach to implantation of the ureter. The risk
tion between the use of sirolimus and the development of for candiduria is increased in patients who have received
PJP.157,158 In such patients, invasive procedures are often prior antimicrobial therapy, with neurogenic bladder,
required for definitive diagnosis. with indwelling urethral catheters, and in intensive care
units. Most kidney transplant recipients with bacteriuria
Diagnosis, Therapy, and Prophylaxis. The characteris- are asymptomatic, whereas pain with pyelonephritis rep-
tic hypoxemia of PJP produces a broad alveolar-arterial par- resents transmural infection with local inflammation out-
tial pressure of oxygen gradient. The level of serum lactate side the denervated allograft causing what is perceived as
dehydrogenase is elevated in most patients with PJP (>300 allograft tenderness.
IU/mL) although many other diffuse pulmonary processes The major causative organisms include gram-negative
also increase serum lactate dehydrogenase levels. No diag- bacilli (Escherichia coli, Klebsiella, Pseudomonas, Enterobac-
nostic pattern exists for PJP on routine chest radiograph. ter, Proteus) and gram-positives (largely enterococci) and
The chest radiograph may be entirely normal or reveal a fungi (Candida species). Each of these groups may mani-
classic pattern of perihilar and interstitial ground-glass fest important antimicrobial resistance; therapy should
infiltrates (see Fig. 31.1). Chest CT scans are more sensitive be based on susceptibility patterns and by the presence or
to the diffuse interstitial and nodular pattern than routine absence of structural abnormalities (obstruction, delayed
radiographs. The clinical and radiologic manifestations of bladder emptying).164 Thus cultures and imaging (ultra-
PJP are virtually identical to the manifestations of CMV. sound to exclude hydronephrosis) are required in patients
The clinical challenge is to determine whether both patho- with upper tract infection. Initial empiric therapy should
gens are present; bronchoalveolar lavage may be helpful include antimicrobial agents not used previously for pro-
and the serum based (1 → 3)-β-D-glucan test shows excel- phylaxis and, where possible, not used in prior episodes
lent sensitivity and very good specificity in the diagnosis of of infection given the risk for antimicrobial resistance.
PJP.159,160 Significant extrapulmonary disease is uncom- Therapy can be narrowed based on susceptibility data.
mon in the transplant recipient. Short-course therapy is not used for treatment of uncom-
Early therapy with TMP-SMX is preferred; few kidney plicated urinary tract infection after transplantation; a
transplant patients tolerate full-dose TMP-SMX for pro- 7-day minimum course with an effective agent is recom-
longed periods.154 This reflects the elevation of creatinine mended. Upper tract disease (pyelonephritis) may require
by trimethoprim (competing for secretion in the kidney), intravenous therapy initially and a 2- to 3-week total
and the toxicity of sulfa agents for the renal allograft. course. Clinical response should be documented. Asymp-
Hydration and the gradual initiation of therapy may help. tomatic candiduria should be treated in patients with renal
Alternative therapies are less desirable but have been used allografts (although data are limited) with fluconazole
with success, including intravenous pentamidine, atova- (200 mg orally per day for 7–14 days). Upper tract disease
quone, clindamycin with primaquine or pyrimethamine, with Candida species suggests obstruction and requires
and trimetrexate. The use of short courses of adjunctive ste- more intensive therapy (fluconazole 400 mg daily for 3–4
roids with a gradual taper is generally useful. weeks). Echinocandins are not useful for treatment of most
The importance of preventing Pneumocystis infection urinary tract infections because they achieve poor concen-
cannot be overemphasized. Low-dose TMP-SMX is well trations in the urine. Removal of stents and catheters is
tolerated and should be used in the absence of concrete generally required for cure.
data showing true allergy or interstitial nephritis.154,155 The prevention of urinary tract infections is altered by
Alternative prophylactic strategies, including dapsone, TMP-SMX prophylaxis. In the absence of instrumentation
31 • Infection in Kidney Transplant Recipients 535

or obstruction, TMP-SMX given for 6 months to 1 year post- 18. US Public Health Service. PHS guideline for reducing human
transplant is generally effective. Few recent studies address immunodeficiency virus, hepatitis B virus, and hepatitis C virus
transmission through organ transplantation. Pub Health Rep
whether the changing ecology of bacteria has reduced the 2013;128:247–343.
efficacy of prophylaxis. In TMP-SMX intolerant patients, a 19. Kasiske BL, Cangro CB, Hariharan S, et al. The evaluation of renal
fluoroquinolone may be used as prophylaxis with the addi- transplantation candidates: clinical practice guidelines. Am J Trans-
tion of another agent against PJP. plant 2001;1(Suppl. 2):3–95.
20. Schorn R, Hohne M, Meerbach A, et al. Chronic norovirus infection
after kidney transplantation: molecular evidence for immune-driven
viral evolution. Clin Infect Dis 2010;51:307–14.
Conclusions 21. Patel G, Huprikar S, Factor SH, Jenkins SG, Calfee DP. Outcomes of
carbapenem-resistant Klebsiella pneumoniae infection and the impact
Transplant infectious disease is increasingly character- of antimicrobial and adjunctive therapies. Infect Cont Hosp Epide-
miol 2008;29:1099–106.
ized by the ability to monitor and prevent infection based 22. Brizendine KD, Richter SS, Cober ED, van Duin D. Carbapenem-
on prophylaxis, new antimicrobial agents, and vaccina- resistant Klebsiella pneumoniae urinary tract infection follow-
tion. Despite significant advances, infection poses a life- ing solid organ transplantation. Antimicrob Agents Chemother
threatening challenge for many recipients. In the future, 2015;59:553–7.
23. Martin SI, Fishman JA. Pneumocystis pneumonia in solid organ
increased availability of pathogen-specific immune function transplant recipients. Am J Transplant 2009;9(Suppl. 4):S227–33.
tests, enhanced donor and recipient screening, and a better 24. de Boer MG, de Fijter JW, Kroon FP. Outbreaks and clustering of
understanding of infection risks such as genetic polymor- Pneumocystis pneumonia in kidney transplant recipients: a system-
phisms should combine with advances in transplant immu- atic review. Med Mycol 2011;49:673–80.
nosuppression to further reduce infection risks. 25. Florescu DF. Solid organ transplantation: hypogammaglobulinae-
mia and infectious complications after solid organ transplantation.
Clin Exp Immunol 2014;178(Suppl. 1):54–6.
References 26. Florescu DF, Kalil AC, Qiu F, Schmidt CM, Sandkovsky U. What is the
1. Fishman JA. Infection in solid-organ transplant recipients. N Engl J impact of hypogammaglobulinemia on the rate of infections and sur-
Med 2007;357:2601–14. vival in solid organ transplantation? A meta-analysis. Am J Trans-
2. Fishman JA. Infection in organ transplantation. Am J Transplant plant 2013;13:2601–10.
2017;17:856–79. 27. Manuel O, Asberg A, Pang X, et al. Impact of genetic polymorphisms
3. Avery RK, Ljungman P. Prophylactic measures in the solid-organ in cytomegalovirus glycoprotein B on outcomes in solid-organ
recipient before transplantation. Clin Infect Dis 2001;33(Suppl. transplant recipients with cytomegalovirus disease. Clin Infect Dis
1):S15–21. 2009;49:1160–6.
4. Avery RK, Michaels M. Update on immunizations in solid organ 28. Wojtowicz A, Gresnigt MS, Lecompte T, et al. IL1B and DEFB1 poly-
transplant recipients: what clinicians need to know. Am J Trans- morphisms increase susceptibility to invasive mold infection after
plant 2008;8:9–14. solid-organ transplantation. J Infect Dis 2015;211:1646–57.
5. Grossi PA, Fishman JA. Donor-derived infections in solid organ 29. Wojtowicz A, Lecompte TD, Bibert S, et al. PTX3 polymorphisms and
transplant recipients. Am J Transplant 2009;9(Suppl. 4):S19–26. invasive mold infections after solid organ transplant. Clin Infect Dis
6. Ison MG, Hager J, Blumberg E, et al. Donor-derived disease trans- 2015;61:619–22.
mission events in the United States: data reviewed by the OPTN/ 30. Fishman JA. Infection in renal transplant recipients. Sem Nephrol
UNOS Disease Transmission Advisory Committee. Am J Transplant 2007;27:445–61.
2009;9:1929–35. 31. Len O, Rodriguez-Pardo D, Gavalda J, et al. Outcome of Clostridium
7. Chin-Hong PV, Schwartz BS, Bern C, et al. Screening and treatment difficile-associated disease in solid organ transplant recipients: a
of Chagas disease in organ transplant recipients in the United States: prospective and multicentre cohort study. Transpl Int 2012;25:
recommendations from the Chagas in Transplant Working Group. 1275–81.
Am J Transplant 2011;11:672–80. 32. Dubberke ER, Burdette SD, AST Infectious Diseases Community of
8. Fishman JA, Greenwald MA, Kuehnert MJ. Enhancing transplant Practice. Clostridium difficile infections in solid organ transplanta-
safety: a new era in the microbiologic evaluation of organ donors? tion. Am J Transplant 2013;13(Suppl. 4):42–9.
Am J Transplant 2007;7:2652–4. 33. Avery RK. Recipient screening prior to solid-organ transplantation.
9. Humar A, Morris M, Blumberg E, et al. Nucleic acid testing (NAT) of Clin Infect Dis 2002;35:1513–9.
organ donors: is the “best” test the right test? A consensus confer- 34. Len O, Garzoni C, Lumbreras C, et al. Recommendations for screen-
ence report. Am J Transplant;10:889–899. ing of donor and recipient prior to solid organ transplantation and
10. Fischer SA, Avery RK. Screening of donor and recipient prior to solid to minimize transmission of donor-derived infections. Clin Microbiol
organ transplantation. Am J Transplant 2009;9(Suppl. 4):S7–18. Infect 2014;20(Suppl. 7):108.
11. Fischer SA, Graham MB, Kuehnert MJ, et al. Transmission of lym- 35. Freeman RB, Giatras I, Falagas ME, et al. Outcome of transplanta-
phocytic choriomeningitis virus by organ transplantation. N Engl J tion of organs procured from bacteremic donors. Transplantation
Med 2006;354:2235–49. 1999;68:1107–11.
12. Srinivasan A, Burton EC, Kuehnert MJ, et al. Transmission of rabies 36. Pappas PG, Silveira FP. Candida in solid organ transplant recipients.
virus from an organ donor to four transplant recipients. N Engl J Med Am J Transplant 2009;9(Suppl. 4):S173–9.
2005;352:1103–11. 37. Patel R, Portela D, Badley AD, et al. Risk factors of invasive Candida
13. Maier T, Schwarting A, Mauer D, et al. Management and outcomes and non-Candida fungal infections after liver transplantation. Trans-
after multiple corneal and solid organ transplantations from a donor plantation 1996;62:926–34.
infected with rabies virus. Clin Infect Dis 2010;50:1112–9. 38. Levi ME, Kumar D, Green M, et al. Considerations for screening live
14. Fishman JA, Grossi PA. Donor-derived infection—the challenge for kidney donors for endemic infections: a viewpoint on the UNOS pol-
transplant safety. Nat Rev Nephrol 2014;10:663–72. icy. Am J Transplant 2014;14:1003–11.
15. Fishman JA, Greenwald MA, Grossi PA. Transmission of infection 39. Singh N, Paterson DL. Mycobacterium tuberculosis infection in solid-
with human allografts: essential considerations in donor screening. organ transplant recipients: impact and implications for manage-
Clin Infect Dis 2012;55:720–7. ment. Clin Infect Dis 1998;27:1266–77.
16. Grossi PA, Fishman JA, Practice ASTID Co. Donor-derived infections 40. Bumbacea D, Arend SM, Eyuboglu F, et al. The risk of tuberculosis in
in solid organ transplant recipients. Am J Transplant 2009;9(Suppl. transplant candidates and recipients: a TBNET consensus statement.
4):S19–26. Euro Resp J 2012;40:990–1013.
17. Kiberd BA, Forward K. Screening for West Nile virus in organ 41. Munoz P, Rodriguez C, Bouza E. Mycobacterium tuberculosis infec-
transplantation: a medical decision analysis. Am J Transplant tion in recipients of solid organ transplants. Clin Infect Dis 2005;40:
2004;4:1296–301. 581–7.
536 Kidney Transplantation: Principles and Practice

42. Aguado JM, Torre-Cisneros J, Fortun J, et al. Tuberculosis in solid- 66. Locke JE, Shelton BA, Reed RD, et al. Identification of optimal
organ transplant recipients: consensus statement of the group for the donor-recipient combinations among human immunodeficiency
study of infection in transplant recipients (GESITRA) of the Spanish virus (HIV)-positive kidney transplant recipients. Am J Transplant
Society of Infectious Diseases and Clinical Microbiology. Clin Infect 2016;16:2377–83.
Dis 2009;48:1276–84. 67. Frassetto L, Floren L, Barin B, et al. Changes in clearance, vol-
43. Zayas CF, Perlino C, Caliendo A, et al. Chagas disease after organ ume and bioavailability of immunosuppressants when given with
transplantation—United States 2001. MMWR 2002;51:210–2. HAART in HIV-1 infected liver and kidney transplant recipients.
44. Allen UD, Preiksaitis JK, AST Infectious Diseases Community of Biopharma Drug Disp 2013;34:442–51.
Practice. Epstein-Barr virus and posttransplant lymphoprolif- 68. Sawinski D, Shelton BA, Mehta S, et al. Impact of protease inhibitor-
erative disorder in solid organ transplantation. Am J Transplant based anti-retroviral therapy on outcomes for HIV+ kidney trans-
2013;13(Suppl. 4):107–20. plant recipients. Am J Transplant 2017;17:3114–22.
45. Luskin MR, Heil DS, Tan KS, et al. The impact of EBV status on char- 69. Tricot L, Teicher E, Peytavin G, et al. Safety and efficacy of raltegravir
acteristics and outcomes of posttransplantation lymphoproliferative in HIV-infected transplant patients cotreated with immunosuppres-
disorder. Am J Transplant 2015;15:2665–73. sive drugs. Am J Transplant 2009;9:1946–52.
46. Opelz G, Daniel V, Naujokat C, Dohler B. Epidemiology of pretrans- 70. Boyle SM, Malat G, Harhay MN, et al. Association of tenofovir diso-
plant EBV and CMV serostatus in relation to posttransplant non- proxil fumarate with primary allograft survival in HIV-positive kid-
Hodgkin lymphoma. Transplantation 2009;88:962–7. ney transplant recipients. Transpl Infect Dis 2017;19.
47. Opelz G, Daniel V, Naujokat C, Fickenscher H, Dohler B. Effect of 71. Malat GE, Ranganna KM, Sikalas N, Liu L, Jindal RM, Doyle A.
cytomegalovirus prophylaxis with immunoglobulin or with antivi- High frequency of rejections in HIV-positive recipients of kidney
ral drugs on post-transplant non-Hodgkin lymphoma: a multicentre transplantation: a single center prospective trial. Transplantation
retrospective analysis. Lancet Oncol 2007;8:212–8. 2012;94:1020–4.
48. Opelz G, Dohler B. Impact of HLA mismatching on incidence of post- 72. Kotton CN, Kumar D, Caliendo AM, et al. International consensus
transplant non-hodgkin lymphoma after kidney transplantation. guidelines on the management of cytomegalovirus in solid organ
Transplantation 2010;89:567–72. transplantation. Transplantation 2010;89:779–95.
49. Tsai DE, Nearey M, Hardy CL, et al. Use of EBV PCR for the diagno- 73. Kaminski H, Fishman JA. The cell biology of cytomegalovirus: impli-
sis and monitoring of post-transplant lymphoproliferative disorder cations for transplantation. Am J Transplant 2016;16:2254–69.
in adult solid organ transplant patients. Am J Transplant 2002;2: 74. Humar A, Snydman D. Cytomegalovirus in solid organ transplant
946–54. recipients. Am J Transplant 2009;9(Suppl. 4):S78–86.
50. Levitsky J, Doucette K, AST Infectious Diseases Community of Prac- 75. Nichols WG, Price TH, Gooley T, Corey L, Boeckh M. Transfusion-
tice. Viral hepatitis in solid organ transplantation. Am J Transplant transmitted cytomegalovirus infection after receipt of leukoreduced
2013;13(Suppl. 4):147–68. blood products. Blood 2003;101:4195–200.
51. Huprikar S, Danziger-Isakov L, Ahn J, et al. Solid organ transplanta- 76. Fietze E, Prosch S, Reinke P, et al. Cytomegalovirus infection in
tion from hepatitis B virus-positive donors: consensus guidelines for transplant recipients. The role of tumor necrosis factor. Transplan-
recipient management. Am J Transplant 2015;15:1162–72. tation 1994;58:675–80.
52. Wright AJ, Fishman JA, Chung RT. Lamivudine compared with newer 77. Reinke P, Fietze E, Ode-Hakim S, et al. Late-acute renal allograft
antivirals for prophylaxis of hepatitis B core antibody positive livers: a rejection and symptomless cytomegalovirus infection. Lancet
cost-effectiveness analysis. Am J Transplant 2014;14:629–34. 1994;344:1737–8.
53. Reese PP, Abt PL, Blumberg EA, Goldberg DS. Transplanting hepati- 78. Lowance D, Neumayer HH, Legendre CM, et al. Valacyclovir for the
tis C-positive kidneys. N Engl J Med 2015;373:303–5. prevention of cytomegalovirus disease after renal transplantation.
54. Blumberg EA, Stock P. Solid organ transplantation in the HIV- International Valacyclovir Cytomegalovirus Prophylaxis Transplan-
infected patient. Am J Transplant 2009;9(Suppl. 4):S131–5. tation Study Group. N Engl J Med 1999;340:1462–70.
55. US Department of Health and Human Services, Health Resources 79. Chou S. Antiviral drug resistance in human cytomegalovirus. Trans-
and Services Administration. Final Human Immunodeficiency Virus plant Infect Dis 1999;1:105–14.
(HIV) Organ Policy Equity (HOPE) Act Safeguards and Research Cri- 80. Lurain NS, Chou S. Antiviral drug resistance of human cytomegalo-
teria for Transplantation of Organs Infected with HIV. Washington, virus. Clin Microbiol Rev 2010;23:689–712.
DC: DHHS; 2016. 81. Emery VC, Sabin CA, Cope AV, Gor D, Hassan-Walker AF, Griffiths
56. Tanabe K, Kitani R, Takahashi K, et al. Long-term results in human PD. Application of viral-load kinetics to identify patients who
T-cell leukemia virus type 1-positive renal transplant recipients. develop cytomegalovirus disease after transplantation. Lancet
Transplant Proc 1998;30:3168–70. 2000;355:2032–6.
57. US Department of Health and Human Services. Guidance for HTLV-1 82. Caliendo AM, St George K, Kao SY, et al. Comparison of quantitative
Screening and Confirmation in Potential Donors and Reporting cytomegalovirus (CMV) PCR in plasma and CMV antigenemia assay:
Potential HTLV-1 Infection. Washington, DC: DHHS; 2011. clinical utility of the prototype AMPLICOR CMV MONITOR test in
58. Kaul DR, Taranto S, Alexander C, et al. Donor screening for human transplant recipients. J Clin Microbiol 2000;38:2122–7.
T-cell lymphotrophic virus 1/2: changing paradigms for changing 83. Hakki M, Chou S. The biology of cytomegalovirus drug resistance.
testing capacity. Am J Transplant 2010;10:207–13. Curr Opin Infect Dis 2011;24:60511.
59. Aguado JM, Torre-Cisneros J, Fortun J, et al. [Consensus document 84. Kalil AC, Levitsky J, Lyden E, Stoner J, Freifeld AG. Meta-analysis:
for the management of tuberculosis in solid organ transplant recipi- the efficacy of strategies to prevent organ disease by cytomegalovi-
ents]. Enfermed Infecs Microbiol Clin 2009;27:465–73. rus in solid organ transplant recipients. Ann Intern Med 2005;143:
60. Manuel O, Humar A, Preiksaitis J, et al. Comparison of quantiferon-TB 870–80.
gold with tuberculin skin test for detecting latent tuberculosis infection 85. Lisboa LF, Asberg A, Kumar D, et al. The clinical utility of whole
prior to liver transplantation. Am J Transplant 2007;7:2797–801. blood versus plasma cytomegalovirus viral load assays for monitor-
61. Frassetto LA, Browne M, Cheng A, et al. Immunosuppressant phar- ing therapeutic response. Transplantation 2011;91:231–6.
macokinetics and dosing modifications in HIV-1 infected liver and 86. Abdul-Ali D, Kraft CS, Ingersoll J, Frempong M, Caliendo AM. Cyto-
kidney transplant recipients. Am J Transplant 2007;7:2816–20. megalovirus DNA stability in EDTA anti-coagulated whole blood
62. Roland ME, Barin B, Carlson L, et al. HIV-infected liver and kidney and plasma samples. J Clin Virol 2011;52:222–4.
transplant recipients: 1- and 3-year outcomes. Am J Transplant 87. Pang XL, Fox JD, Fenton JM, Miller GG, Caliendo AM, Preiksaitis JK.
2008;8:355–65. Interlaboratory comparison of cytomegalovirus viral load assays.
63. Roland ME, Barin B, Huprikar S, et al. Survival in HIV-positive trans- Am J Transplant 2009;9:258–68.
plant recipients compared with transplant candidates and with HIV- 88. Hirsch HH, Lautenschlager I, Pinsky BA, et al. An international
negative controls. AIDS 2016;30:435–44. multicenter performance analysis of cytomegalovirus load tests. Clin
64. Stock PG, Barin B, Murphy B, et al. Outcomes of kidney transplanta- Infect Dis 2013;56:367–73.
tion in HIV-infected recipients. N Engl J Med 2010;363:2004–14. 89. Hayden RT, Gu Z, Sam SS, et al. Comparative performance of
65. Locke JE, Gustafson S, Mehta S, et al. Survival benefit of kidney trans- reagents and platforms for quantitation of cytomegalovirus DNA by
plantation in HIV-infected patients. Ann Surg 2017;265:604–8. digital PCR. J Clin Microbiol 2016;54:2602–8.
31 • Infection in Kidney Transplant Recipients 537

90. Rychert J, Danziger-Isakov L, Yen-Lieberman B, et al. Multicenter 112. Loren AW, Porter DL, Stadtmauer EA, Tsai DE. Post-transplant
comparison of laboratory performance in cytomegalovirus and lymphoproliferative disorder: a review. Bone Marrow Transplant
Epstein-Barr virus viral load testing using international standards. 2003;31:145–55.
Clin Transplant 2014;28:1416–23. 113. Swerdlow SH. T-cell and NK-cell posttransplantation lymphoprolif-
91. Kotton CN, Kumar D, Caliendo AM, et al. Updated international con- erative disorders. Am J Clin Pathol 2007;127:887–95.
sensus guidelines on the management of cytomegalovirus in solid- 114. Swerdlow SH, Campo E, Pileri SA, et al. The. 2016 revision of the
organ transplantation. Transplantation 2013;96:333–60. World Health Organization classification of lymphoid neoplasms.
92. Humar A. Reactivation of viruses in solid organ transplant Blood 2016;127:2375–90.
patients receiving cytomegalovirus prophylaxis. Transplantation 115. Ghobrial IM, Habermann TM, Macon WR, et al. Differences between
2006;82:S9–14. early and late posttransplant lymphoproliferative disorders in solid
93. Humar A, Limaye AP, Blumberg EA, et al. Extended valganciclovir organ transplant patients: are they two different diseases? Trans-
prophylaxis in D+/R− kidney transplant recipients is associated with plantation 2005;79:244–7.
long-term reduction in cytomegalovirus disease: two-year results of 116. Tsai DE, Douglas L, Andreadis C, et al. EBV PCR in the diagnosis and
the IMPACT study. Transplantation 2010;90:1427–31. monitoring of posttransplant lymphoproliferative disorder: results of
94. Gabardi S, Asipenko N, Fleming J, et al. Evaluation of low- ver- a two-arm prospective trial. Am J Transplant 2008;8:1016–24.
sus high-dose valganciclovir for prevention of cytomegalovirus 117. Semenova T, Lupo J, Alain S, et al. Multicenter evaluation of whole-
disease in high-risk renal transplant recipients. Transplantation blood Epstein-Barr viral load standardization using the WHO inter-
2015;99:1499–505. national standard. J Clin Microbiol 2016;54:1746–50.
95. Halim MA, Al-Otaibi T, Gheith O, et al. Efficacy and Safety Of Low- 118. Preiksaitis JK, Pang XL, Fox JD, Fenton JM, Caliendo AM, Miller GG.
Dose Versus Standard-Dose Valganciclovir For Prevention Of Interlaboratory comparison of Epstein-Barr virus viral load assays.
Cytomegalovirus Disease In Intermediate-Risk Kidney Transplant Am J Transplant 2009;9:269–79.
Recipients. Exper Clin Transplant 2016;14:526–34. 119. Young L, Alfieri C, Hennessy K, et al. Expression of Epstein-Barr virus
96. Kropeit D, Scheuenpflug J, Erb-Zohar K, et al. Pharmacokinetics transformation-associated genes in tissues of patients with EBV lym-
and safety of letermovir, a novel anti-human cytomegalovirus drug, phoproliferative disease. N Engl J Med 1989;321:1080–5.
in patients with renal impairment. Br J Clin Pharmacol 2017;83: 120. Reshef R, Vardhanabhuti S, Luskin MR, et al. Reduction of immuno-
1944–53. suppression as initial therapy for posttransplantation lymphoprolif-
97. Verghese PS, Schleiss MR. Letermovir treatment of human cyto- erative disorder (bigstar). Am J Transplant 2011;11:336–47.
megalovirus infection antiinfective agent. Drugs Future 2013;38: 121. AlDabbagh MA, Gitman MR, Kumar D, Humar A, Rotstein C, Husain
291–8. S. The role of antiviral prophylaxis for the prevention of Epstein-Barr
98. Chou S. A third component of the human cytomegalovirus termi- virus-associated posttransplant lymphoproliferative disease in solid
nase complex is involved in letermovir resistance. Antiviral Res organ transplant recipients: a systematic review. Am J Transplant
2017;148:1–4. 2017;17(3):770–81. Available online at: https://doi.org/10.1111/
99. Josephson MA, Gillen D, Javaid B, et al. Treatment of renal allograft ajt.14020.
polyoma BK virus infection with leflunomide. Transplantation 122. Daniel PT, Kroidl A, Kopp J, et al. Immunotherapy of B-cell lym-
2006;81:704–10. phoma with CD3x19 bispecific antibodies: costimulation via CD28
100. Waldman WJ, Knight DA, Blinder L, et al. Inhibition of cytomegalo- prevents “veto” apoptosis of antibody-targeted cytotoxic T cells.
virus in vitro and in vivo by the experimental immunosuppressive Blood 1998;92:4750–7.
agent leflunomide. Intervirology 1999;42:412–8. 123. Jacobson CA, LaCasce AS. Lymphoma: risk and response after solid
101. Avery RK, Mossad SB, Poggio E, et al. Utility of leflunomide in the organ transplant. Oncology (Williston Park) 2010;24:936–44.
treatment of complex cytomegalovirus syndromes. Transplantation 124. Porcu P, Eisenbeis CF, Pelletier RP, et al. Successful treatment of
2010;90:419–26. posttransplantation lymphoproliferative disorder (PTLD) following
102. Marty FM, Boeckh M. Maribavir and human cytomegalovirus: what renal allografting is associated with sustained CD8(+) T-cell restora-
happened in the clinical trials and why might the drug have failed? tion. Blood 2002;100:2341–8.
Curr Opin Virol 2011;1:555–62. 125. Elstrom RL, Andreadis C, Aqui NA, et al. Treatment of PTLD with
103. Marty FM, Ljungman P, Papanicolaou GA, et al. Maribavir pro- rituximab or chemotherapy. Am J Transplant 2006;6:569–76.
phylaxis for prevention of cytomegalovirus disease in recipients of 126. Comoli P, Cioni M, Basso S, et al. Immunity to polyomavirus BK
allogeneic stem-cell transplants: a phase 3, double-blind, placebo- infection: immune monitoring to regulate the balance between risk
controlled, randomised trial. Lancet Infect Dis 2011;11:284–92. of BKV nephropathy and induction of alloimmunity. Clin Dev Immu-
104. Avery RK, Marty FM, Strasfeld L, et al. Oral maribavir for treatment nol 2013;2013:256923.
of refractory or resistant cytomegalovirus infections in transplant 127. Hirsch HH, Randhawa P. BK polyomavirus in solid organ transplan-
recipients. Transplant Infect Dis 2010;12:489–96. tation. Am J Transplant 2013;13(Suppl. 4):179–88.
105. Marty FM, Winston DJ, Rowley SD, et al. CMX001 to prevent cyto- 128. Vats A, Randhawa PS, Shapiro R. Diagnosis and treatment of
megalovirus disease in hematopoietic-cell transplantation. N Engl J BK virus-associated transplant nephropathy. Adv Exp Med Biol
Med 2013;369:1227–36. 2006;577:213–27.
106. Allen UD, Preiksaitis JK. Epstein-Barr virus and posttransplant lym- 129. Munoz P, Fogeda M, Bouza E, et al. Prevalence of BK virus replica-
phoproliferative disorder in solid organ transplantation. Am J Trans- tion among recipients of solid organ transplants. Clin Infect Dis
plant 2013;13(Suppl. 4):107–20. 2005;41:1720–5.
107. Green M, Michaels MG. Epstein-Barr virus infection and posttrans- 130. Hirsch HH, Babel N, Comoli P, et al. European perspective on human
plant lymphoproliferative disorder. Am J Transplant 2013;13(Suppl. polyomavirus infection, replication and disease in solid organ trans-
3):41–54. plantation. Clin Microbiol Infect 2014;20(Suppl. 7):74–88.
108. Paya CV, Fung JJ, Nalesnik MA, et al. Epstein-Barr virus-induced 131. Mylonakis E, Goes N, Rubin RH, Cosimi AB, Colvin RB, Fishman
posttransplant lymphoproliferative disorders. ASTS/ASTP EBV- JA. BK virus in solid organ transplant recipients: an emerging syn-
PTLD Task Force and The Mayo Clinic Organized International Con- drome. Transplantation 2001;72:1587–92.
sensus Development Meeting. Transplantation 1999;68:1517–25. 132. Fishman JA. BK virus nephropathy—polyomavirus adding insult to
109. Dharnidharka VR, Araya CE. Post-transplant lymphoproliferative injury. N Engl J Med 2002;347:527–30.
disease. Pediatr Nephrol 2009;24:731–6. 133. Hirsch HH, Ramos E. Retransplantation after polyomavirus-associated
110. Dharnidharka VR, Lamb KE, Gregg JA, Meier-Kriesche HU. Asso- nephropathy: just do it? Am J Transplant 2006;6:7–9.
ciations between EBV serostatus and organ transplant type in PTLD 134. Ramos E, Vincenti F, Lu WX, et al. Retransplantation in patients
risk: an analysis of the SRTR National Registry Data in the United with graft loss caused by polyoma virus nephropathy. Transplanta-
States. Am J Transplant 2012;12:976–83. tion 2004;77:131–3.
111. Dharnidharka VR, Sullivan EK, Stablein DM, Tejani AH, Har- 135. Hirsch HH, Randhawa P, AST Infectious Diseases Community of
mon WE. Risk factors for posttransplant lymphoproliferative dis- Practice. BK polyomavirus in solid organ transplantation. Am J
order (PTLD) in pediatric kidney transplantation: a report of the Transplant 2013;13(Suppl. 4):179–88.
North American Pediatric Renal Transplant Cooperative Study
(NAPRTCS). Transplantation 2001;71:1065–8.
538 Kidney Transplantation: Principles and Practice

136. Horn DL, Neofytos D, Anaissie EJ, et al. Epidemiology and outcomes 150. Singh N. Immune recovery gone rogue: microbe-associated immune
of candidemia in 2019 patients: data from the prospective antifungal reconstitution syndrome in neutropenic host. J Infect 2015;70:
therapy alliance registry. Clin Infect Dis 2009;48:1695–703. 563–4.
137. Lamoth F, Chung SJ, Damonti L, Alexander BD. Changing epidemi- 151. Singh N, Perfect JR. Immune reconstitution syndrome associated
ology of invasive mold infections in patients receiving azole prophy- with opportunistic mycoses. Lancet Infect Dis 2007;7:395–401.
laxis. Clin Infect Dis 2017;64:1619–21. 152. Fishman JA. Treatment of infection due to Pneumocystis carinii: anti-
138. Patterson TF, Thompson III GR, Denning DW, et al. Executive microbial agents and chemotherapy. 1998;42:1309–14.
summary: practice guidelines for the diagnosis and management 153. Fishman JA. Prevention of infection caused by Pneumocystis carinii in
of aspergillosis: 2016 update by the Infectious Diseases Society of transplant recipients. Clin Infect Dis 2001;33:1397–405.
America. Clin Infect Dis 2016;63:433–42. 154. Martin SI, Fishman JA. Pneumocystis pneumonia in solid organ
139. Patterson TF, Thompson III GR, Denning DW, et al. Practice guide- transplantation. Am J Transplant 2013;13(Suppl. 4):272–9.
lines for the diagnosis and management of aspergillosis: 2016 155. Rodriguez M, Fishman JA. Prevention of infection due to Pneumocys-
update by the Infectious Diseases Society of America. Clin Infect Dis tis spp. in human immunodeficiency virus-negative immunocom-
2016;63:e1–60. promised patients. Clin Microbiol Rev 2004;17:770–82.
140. Maertens JA, Raad II , Marr KA, et al. Isavuconazole versus 156. Lopez P, Kohler S, Dimri S. Interstitial lung disease associated with
voriconazole for primary treatment of invasive mould disease mTOR inhibitors in solid organ transplant recipients: results from a
caused by Aspergillus and other filamentous fungi (SECURE): a large phase III clinical trial program of everolimus and review of the
phase 3, randomised-controlled, non-inferiority trial. Lancet literature. J Transplant 2014;2014:305931.
2016;387:760–9. 157. De Castro N, Xu F, Porcher R, Pavie J, Molina JM, Peraldi MN. Pneu-
141. Marty FM, Ostrosky-Zeichner L, Cornely OA, et al. Isavuconazole mocystis jirovecii pneumonia in renal transplant recipients occur-
treatment for mucormycosis: a single-arm open-label trial and case- ring after discontinuation of prophylaxis: a case-control study. Clin
control analysis. Lancet Infect Dis 2016;16:828–37. Microbiol Infect 2010;16:1375–7.
142. Rivosecchi RM, Clancy CJ, Shields RK, et al. Effects of isavucon- 158. Neff RT, Jindal RM, Yoo DY, Hurst FP, Agodoa LY, Abbott KC. Anal-
azole on the plasma concentrations of tacrolimus among solid- ysis of USRDS: incidence and risk factors for Pneumocystis jiroveci
organ transplant patients. Antimicrob Agent Chemother 2017 Aug pneumonia. Transplantation 2009;88:135–41.
24;61(9):e00970-17. Available online at: https://doi.org/10.1128/ 159. Damiani C, Le Gal S, Da Costa C, Virmaux M, Nevez G, Totet A. Com-
AAC.00970-17. bined quantification of pulmonary Pneumocystis jirovecii DNA and
143. Fishman JA, Gonzalez RG, Branda JA. Case records of the Massa- serum (1->3)-beta-D-glucan for differential diagnosis of pneumo-
chusetts General Hospital. Case 11-2008: a 45-year-old man with cystis pneumonia and Pneumocystis colonization. J Clin Microbiol
changes in mental status after liver transplantation. N Eng J Med 2013;51:3380–8.
2008;358:1604–13. 160. Karageorgopoulos DE, Qu JM, Korbila IP, Zhu YG, Vasileiou VA,
144. Davis JA, Horn DL, Marr KA, Fishman JA. Central nervous system Falagas ME. Accuracy of beta-D-glucan for the diagnosis of Pneu-
involvement in cryptococcal infection in individuals after solid mocystis jirovecii pneumonia: a meta-analysis. Clin Microbiol Infect
organ transplantation or with AIDS. Transplant Infect Dis 2009;11: 2013;19:39–49.
432–7. 161. Alangaden GJ, Thyagarajan R, Gruber SA, et al. Infectious complica-
145. Nguyen MH, Husain S, Clancy CJ, et al. Outcomes of central nervous tions after kidney transplantation: current epidemiology and associ-
system cryptococcosis vary with host immune function: results from ated risk factors. Clin Transplant 2006;20:401–9.
a multi-center, prospective study. J Infect 2010;61:419–26. 162. Lee JR, Bang H, Dadhania D, et al. Independent risk factors for
146. Singh N, Lortholary O, Dromer F, et al. Central nervous system cryp- urinary tract infection and for subsequent bacteremia or acute
tococcosis in solid organ transplant recipients: clinical relevance cellular rejection: a single-center report of 1166 kidney allograft
of abnormal neuroimaging findings. Transplantation 2008;86: recipients. Transplantation 2013;96:732–8.
647–51. 163. Ariza-Heredia EJ, Beam EN, Lesnick TG, Cosio FG, Kremers WK,
147. Osawa R, Alexander BD, Lortholary O, et al. Identifying predictors of Razonable RR. Impact of urinary tract infection on allograft func-
central nervous system disease in solid organ transplant recipients tion after kidney transplantation. Clin Transplant 2014;28:683–90.
with cryptococcosis. Transplantation 2010;89:69–74. 164. Munoz P. Management of urinary tract infections and lymphocele
148. Baddley JW, Forrest GN, AST Infectious Diseases Community of in renal transplant recipients. Clin Infect Dis 2001;33(Suppl. 1):
Practice. Cryptococcosis in solid organ transplantation. Am J Trans- S53–7.
plant 2013;13(Suppl. 4):242–9.
149. Fishman JA. Editorial commentary: Immune reconstitution syn-
drome: how do we “tolerate” our microbiome? Clin Infect Dis
2015;60:45–7.
32 Liver Disease Among Renal
Transplant Recipients
ADNAN SAID, NASIA SAFDAR, and MICHAEL R. LUCEY

CHAPTER OUTLINE Overview of Incidence and HCV Species


Clinicopathologic Associations Clinical Manifestations of Hepatitis C
Combined Liver and Kidney Diseases Infection in Immunocompetent Hosts
Polycystic Disease Incidence/Prevalence and Transmission of
Drug-Induced Hepatotoxicity Hepatitis C in Renal Transplant Patients
Specific Immunosuppressive Agents Allograft Transmission of HCV
in Renal Transplantation and Effect of Pretransplant HCV on
Hepatotoxicity Posttransplant Outcomes
Azathioprine HCV and Posttransplant Diabetes in the
Calcineurin Inhibitor-Induced Hepatotoxicity Renal Transplant Recipient
Sirolimus HCV and Posttransplant Nephropathy
Mycophenolate Mofetil, Mycophenolic Acid Immunosuppressive Strategies in Renal
Monoclonal Antibodies Transplant Patients Infected with HCV
T Cell Costimulatory Inhibitor Hepatitis C Antiviral Therapy
Hepatitis Viruses Associated with Renal Treatment of HCV in the Renal Transplant
Transplantation Candidate and Recipient
Hepatitis B Virus Hepatitis E
HBV Viral Structure and Proteins Nonalcoholic Fatty Liver Disease and
Renal Transplant
Tests for Detection of Hepatitis B
NAFLD Post Renal Transplant
Epidemiology of HBV
NAFLD and Outcomes in Renal Transplant
Hepatitis B Infection in Patients Awaiting Recipients
Renal Transplant on Dialysis
Treatment of NAFLD
Pretransplant Management of Hepatitis
B-Positive Dialysis Patients Hepatocellular Carcinoma After Renal
Transplantation
Posttransplant Prognosis in Hepatitis B
Recipients Systemic Infections Resulting in Hepatitis
and Liver Disease
De Novo HBV Infection After Kidney
Transplantation Liver Abscess
Antiviral Therapy of Chronic Hepatitis B in Mycobacterial Infection
Renal Transplant Candidates/Recipients Viral Infections
Specific Antiviral Agents for HBV Used in Herpesviruses
Renal Transplant Recipients Cytomegalovirus
Treatment of Fibrosing Cholestatic Hepatitis Epstein–Barr Virus
B in Renal Transplant Recipients Herpes Simplex Virus
Summary Varicella-Zoster Virus
Hepatitis C Virus Human Herpesvirus 6 and 7
Viral Structure

Overview of Incidence and of chronic liver injury in otherwise healthy subjects sug-
Clinicopathologic Associations gest that the burden of unrecognized liver disease in the
apparently healthy community is high. A study by Ioannou
Theoretically, the spectrum of liver disease in renal trans- et al.1 used the National Health and Nutrition Examination
plant recipients should mimic the spectrum of disease seen Survey (NHANES) conducted between 1999 and 2002 to
in society. It is axiomatic that renal transplant recipients assess the prevalence of elevated serum transaminase activ-
are at risk for all the acute and chronic liver disorders seen ities in a cohort of 6823 American adults. The prevalence of
in the nontransplant population. Surveys of the prevalence elevated alanine aminotransferase (ALT) was 8.9%, a result
539
540 Kidney Transplantation: Principles and Practice

that is more than double that of previously available esti- and liver polycystic disease and mutations in AD-PKD2
mates in similar populations. Recently another NHANES account for the majority of the remainder.5,6 Patients with
study of American adolescents identified the presence of an mutations in PKD2 tend to have later onset of disease and
elevated ALT, defined as a value above 30 U/mL, in 8.0% of approximately 16 years of increased life expectancy com-
the population.2 Risk factors for an elevated ALT included pared with patients who have mutations in PKD1, but
higher waist circumference, body mass index, fasting blood otherwise the natural history is identical, regardless of
glucose, and fasting triglycerides. whether PKD1 or PKD2 is the mutated gene. Renal cystic
These studies indicate the potential hazards in esti- disease associated with autosomal dominant polycystic dis-
mating the likely prevalence of liver disease in a special ease may develop renal failure that requires hemodialysis or
population, such as recipients of renal transplantation in renal transplantation. The severity of hepatic cystic disease
the absence of good data. The rise in nonalcoholic steato- correlates with both the severity of renal cystic disease and
hepatitis, the advent of highly effective antiviral therapy the degree of renal dysfunction.
to eradicate chronic hepatitis C virus (HCV) infection, and Hepatic cysts are lined with secretory biliary epithe-
possible changing use of alcohol mean that a contempo- lium. The cysts are first noted after puberty and increase in
rary assessment of the spectrum of liver disease might be prevalence with age.7 In addition, hepatic cyst prevalence
quite different from previous reports and in one coun- is correlated with renal cyst volume.7 The lifetime risk for
try compared with another.3 Consequently, it should be expression of hepatic cysts is equal in male and female hold-
noted that there have been no comprehensive attempts ers of the genetic defect, but hepatic cysts tend to be larger
to characterize liver disease in renal transplant recipients and more numerous in women, possibly because of the
since Allison et al.4 examined the prevalence and nature influence of estrogen on hepatic cyst growth.8
of chronic liver disease among 538 patients with func- Symptoms caused by hepatic cysts in adult-onset auto-
tioning renal allografts managed in Scotland between somal dominant polycystic disease are the result of a com-
1980 and 1989. The authors reported that biochemical partment disorder in which the abdominal cavity is unable
evidence of liver dysfunction was observed in 37 patients to accommodate the cystic mass. Patients with massive
(7%), 19 (4%) of whom were seropositive for HCV. The hepatic cysts can experience abdominal pain, early satiety,
work of Allison et al. is most likely an underestimate or dyspnea (Fig. 32.1). These “bulk” symptoms may be so
given that it was undertaken just as HCV infection was troubling as to warrant liver transplantation. In addition,
discovered, and, as will be discussed later, HCV preva- uncommon complications, such as cyst rupture, infection,
lence in renal transplant cohorts has been reported to be torsion, or hemorrhage, can occur.9 Hepatic function and
as high as 40%. portal hemodynamics are usually normal. Biliary obstruc-
In the subsequent sections of this chapter we will discuss tion, portal hypertension, ascites, variceal hemorrhage,
in more detail some liver disorders that appear to occur in and encephalopathy are rare features of autosomal domi-
greater frequency in renal transplant recipients compared nant polycystic disease.
with the background population. In some circumstances, There is no good medical therapy for the abdominal
such as autosomal dominant polycystic disease, the liver symptoms associated with autosomal dominant polycys-
and kidney disorder are part of the same underlying dis- tic disease. Agents such as somatostatin and sirolimus
ease. In other patients in whom renal failure coexists with have been tried without much success.5 For women with
liver disease, the two conditions are acquired separately. symptomatic cysts, stopping oral contraceptive or hor-
Chronic infections with hepatotropic viruses (hepatitis B mone replacement therapy should be considered, but data
virus [HBV] and HCV) fall into this category. We will con- on efficacy are anecdotal. There are many procedures
sider liver diatheses that are consequences of the inher- described to ameliorate the discomfort associated with
ent risks of the transplant process, including drug-related
injury secondary to immunosuppressant medications or
hepatic manifestations of opportunistic infections second-
ary to immunosuppression. Finally, the high prevalence
of metabolic syndrome and obesity in the renal transplant
population has led to the increasing recognition of nonal-
coholic fatty liver disease (NAFLD) in the renal transplant
population. Current knowledge about this common condi-
tion and its consequences and treatment are addressed.

Combined Liver and Kidney


Diseases
POLYCYSTIC DISEASE
Autosomal dominant polycystic disease is a condition aris-
ing from mutations in two distinct genes that result in the
development of the renal and liver cysts. Mutations in AD- Fig. 32.1 The liver has innumerable cysts, ranging from small to large,
PKD1 account for up to 90% adult-onset combined kidney in a patient with autosomal dominant polycystic liver/kidney disease.
32 • Liver Disease Among Renal Transplant Recipients 541

liver cysts. Cyst aspiration under sonographic guidance The mechanisms of drug injury are multiple as well.
may provide temporary relief, but the cysts inevitably Toxic metabolites produced by detoxification of medica-
recur. Continuous or intermittent drainage through a tions through the liver, most commonly via the cyto-
permanent percutaneous catheter should be strongly dis- chrome P-450 mechanisms, may contribute to dose-related
couraged because it runs the risk of converting a sterile hepatotoxicity such as seen with acetaminophen.16,19
cyst into a pyogenic abscess. Surgical approaches include Other medications may have immunologic mechanisms
open or laparoscopic cyst fenestration, hepatic resection, of injury that are not dose-related and considered idiosyn-
and liver transplantation. The results of liver transplanta- cratic.16,18,19 Most patients present asymptomatically or
tion for polycystic liver disease are mixed with a higher with nonspecific symptoms. Occasionally, a hypersensitiv-
than expected incidence of posttransplant complications, ity reaction of fever, lymphadenopathy, and leukocytosis,
including infections. Nevertheless, these patients have often with eosinophilia, may be seen.20,21 Liver test abnor-
had improved access to liver transplant via approval of malities are variable. The most common pattern is acute
model for end-stage liver disease (MELD) exception scores. hepatocellular injury with elevations of aminotransferases
In an audit of United Network for Organ Sharing (UNOS) greater than twofold normal with lesser elevations of alka-
data from 2002 to 2015, 620 patients with polycystic line phosphatase; however, cholestasis and bile duct loss
liver disease were 5.7 times more likely to be transplanted (e.g., amoxillcin-clavulinic acid toxicity) and bland fibrosis
than patients with chronic liver failure and patients with (methotrexate) are also seen.14,15
liver cancer.10 In transplant patients the opportunities for drug-related
Autosomal recessive polycystic kidney disease (ARPKD) hepatotoxicity abound because of the use of multiple
is caused by mutations in the PKHD-1 gene that encodes medications, many of which are metabolized via the same
the protein fibrocystin.11 Congenital hepatic fibrosis pathways in the liver, thereby increasing the risk of accu-
(CHF), caused by ductal plate malformation of the devel- mulation of hepatotoxic metabolites. Common medication
oping biliary system, is invariably present in patients with classes used in transplant that have been implicated in DILI
ARPKD.12 Clinical presentation is related to age. Renal include immunosuppressive medications,22–24 antibiot-
disease predominates in patients that present neonatally. ics,14,15 antihyperlipidemics,14,15 and drugs for hyperten-
Hepatic manifestations predominate in older children sion and diabetes.14,15 In addition, numerous herbal and
and adults, although overlap is common.12 The predomi- nonprescription agents have also been implicated in the
nant manifestations of CHF are the development of portal development of DILI. Finally, more than one agent may
hypertension, dilation of the intrahepatic bile ducts (also be implicated as the etiology for DILI in a given patient.15
known as Caroli’s syndrome), and vascular anomalies. Table 32.1 shows some common medications that stimu-
Variceal formation and hemorrhage, splenomegaly, and late or block the cytochrome P-450 system within the liver
thrombocytopenia are common.12 Dilation of the intra- and may influence the serum concentrations of other drugs
hepatic bile ducts can result in recurrent bile stasis and and their metabolites.
cholangitis. Finally, anomalies of portal venous anatomy The main treatment for DILI is withdrawal of the offend-
are frequent. Treatment for CHF is focused on prevention ing drug. There are few therapies that have been shown
of variceal hemorrhage and promotion of adequate biliary to improve outcomes in clinical trial. Two exceptions are
drainage to prevent cholangitis.13 N-acetylcysteine for acetaminophen toxicity and l-car-
nitine for valproic acid toxicity.25,26 Corticosteroids are of
unproven benefit. In cases that progress to liver failure, liver
DRUG-INDUCED HEPATOTOXICITY
transplantation should be considered.26
Drug-induced liver injury (DILI) can have a wide spectrum,
ranging from asymptomatic elevations of liver enzymes to
acute liver failure. With rare exceptions, the serum bio-
chemical and liver histologic patterns are not diagnostic of TABLE 32.1 Medications that Stimulate or Inhibit the
drug-related injury. Rather, DILI is often diagnosed based Cytochrome P-450 System and Can Influence the Level of
upon a combination of temporal relationship to a particu- Other Medications (Such as Cyclosporine)
lar drug use, exclusion of other pathology (such as viral MEDICATIONS THAT STIMULATE CYTOCHROME P-450 AND CAN
hepatitis), and knowledge of the common pattern of liver DECREASE THE LEVEL OF CALCINEURIN INHIBITOR
test abnormalities associated with particular drugs.14–18 Trimethoprim-sulfamethoxazole
Improvement of liver tests with discontinuation of the Isoniazid
Nafcillin
offending medications offers further evidence of DILI, but Phenytoin
improvement may take weeks. Carbamazepine
The severity of drug-related injury may be predicted by Omeprazole
the degree of impairment of hepatic function. In particular MEDICATIONS THAT INHIBIT CYTOCHROME P-450 AND CAN IN-
the presence of jaundice in association with elevated ami- CREASE THE LEVEL OF CALCINEURIN INHIBITOR
notransferases (known as “Hy’s rule”) is often an ominous Diltiazem
sign of significant hepatocellular injury and risk of pro- Fluconazole
gression to liver failure.14,15,18 In the two largest series to Tetracycline
date, mortality or liver transplantation from idiosyncratic Tacrolimus
Sex hormones
(excluding acetaminophen) drug reactions occurred in Metoclopramide
11.7% and 15% of cases.14,15
542 Kidney Transplantation: Principles and Practice

21 fulfilled the criteria for azathioprine hepatitis with jaun-


Specific Immunosuppressive dice at presentation. Viral hepatitis markers (HCV, HBV, or
Agents in Renal Transplantation both) were present in all 20 that were tested. The jaundice
and Hepatotoxicity disappeared and liver enzymes normalized in all within 4
to 12 weeks of azathioprine discontinuation or dose reduc-
AZATHIOPRINE tion. Rechallenge with azathioprine was performed in four
patients, with recurrence of jaundice in all cases.42,43 In
Azathioprine is an antimetabolite agent that inhibits purine some of these patients, histologic findings were more con-
synthesis. It is the prodrug of 6-mercatopurine (6-MP) and sistent with azathioprine toxicity than viral hepatitis with
inhibits deoxyribonucleic acid (DNA) and ribonucleic acid intrahepatic cholestasis, centrilobular hepatocellular necro-
(RNA) synthesis. A broad range of hepatotoxicity has been sis, and vascular lesions. Most did have chronic liver disease
associated with the use of azathioprine in renal transplant secondary to viral hepatitis on histology (18 out of 21).
recipients, although it is considered rare.22,27–31 The patho- Some have suggested that patients with viral hepatitis
genesis of azathioprine hepatotoxicity is multifactorial, and associated chronic inflammation have reduced catabo-
resulting from endothelial damage,32 direct hepatotoxic- lism and higher levels of toxic azathioprine metabolites in
ity,33 and interlobular bile duct injury.34 In addition, serum the liver, with resultant increases in rates of fibrosis, cirrho-
levels of the 6-MP metabolite 6-methylmercaptopurine sis, and hepatotoxicity.42,43 Other potential mechanisms
ribonucleotide have been associated with the development include accelerated course of viral hepatitis because of the
of hepatotoxicity.35 use of more potent immunosuppressive regimens (pred-
The most severe manifestation of azathioprine toxicity is nisone–azathioprine–cyclosporine) with improvements
sinusoidal obstruction syndrome (SOS), previously known occurring as a result of withdrawal of immunosuppression.
as veno-occlusive disease. The hallmark of SOS is oblitera- These theories are difficult to prove. Nevertheless, in trans-
tion and fibrosis of the central hepatic venule and sinusoi- planting patients with viral hepatitis it is a good policy to
dal congestion.30 SOS is manifested by jaundice, ascites, use minimal immunosuppression (single or dual regimens
hepatomegaly, weight gain, and elevated liver enzymes rather than triple regimens) to minimize acceleration of
(typically alkaline phosphatase with minimal increases in viral hepatitis-associated liver disease.
aminotransferases). In the first few months after kidney
transplantation it can present with asymptomatic hyper- CALCINEURIN INHIBITOR-INDUCED
bilirubinemia and elevated liver enzymes, but progresses to
HEPATOTOXICITY
jaundice, hepatomegaly, and ascites after the first year.36
The diagnosis can be made clinically, but it is often difficult Cyclosporine and tacrolimus are immunosuppressive medi-
to make. In the hematopoietic stem cell population, SOS cations that belong to the class of calcineurin inhibitors.44,45
is diagnosed by two of the three criteria being met: serum Cyclosporine-induced hepatotoxicity is uncommon and
bilirubin greater than 2 mg/dL, hepatomegaly or right the mechanisms of cyclosporine toxicity are incompletely
upper quadrant pain, and sudden weight gain of 2% body understood. Cyclosporine is metabolized via the cytochrome
weight.31 However, these criteria were established in the P-450 system and interactions with medications that
hematopoietic stem cell transplant population and not vali- inhibit or stimulate this pathway can result in increased or
dated in solid-organ transplantation. Doppler ultrasound decreased cyclosporine levels respectively, thereby increas-
is useful for documenting ascites and hepatomegaly, and ing the risk for hepatotoxicity.46 Cyclosporine-induced
for ruling out biliary obstruction or infiltrative processes. decrease in bile flow can result from reduced bile acid secre-
Liver biopsy can be used to help make a diagnosis, as can tion and is associated with risk of bile duct stones and sludge
measurement of the wedged hepatic venous portal gradi- formation in 2% to 5% of transplant recipients.47 Rarely,
ent (HVPG).37,38 Poor outcomes are associated with higher increases in aminotransferases have occurred, mostly in the
bilirubin, degree of weight gain, aminotransferase eleva- first 90 days, and these respond to a reduction in doses. Per-
tion, and HVPG elevation.38 With cessation of azathioprine sistent elevations in aminotransferases are rare and occur
it rarely has been reported to regress.39 Specific therapy in less than 5% to 10% of renal transplant recipients.48,49
for SOS, including defibrotide, heparin, ursodeoxycholic Transient elevations of bilirubin or aminotransferases are
acid, and prostaglandin E1, has produced mixed results.38 more common, occur early (within the first 3 months post-
Transjugular intrahepatic portosystemic shunt and liver transplantation), and are reversible with dose reductions or
transplantation have been reported in small series and case discontinuation.47 Among renal transplant recipients with-
reports, respectively.40 Other vascular diseases of the liver out preexisting liver disease, azathioprine-treated patients
have also been attributed to azathioprine, including pelio- had a higher incidence of posttransplant chronic liver dis-
sis hepatis (dilated blood-filled cavities within the liver), ease compared with cyclosporine-treated patients.50
presumably secondary to endothelial injury within the Tacrolimus has a similar immunosuppressive mechanism
liver, leading to sinusoidal dilation. Nodular regenerative of action to cyclosporine.45 In liver transplant recipients it is
hyperplasia can be associated with peliosis. Veno-occlu- associated with fewer episodes of acute rejection, need for
sive disease is rarely seen and by the time it appears, por- salvage immunosuppressive therapy, or ductopenic rejec-
tal hypertension with complications of ascites and variceal tion than cyclosporine. The overall patient and graft sur-
hemorrhage are often present.41 vival rates are similar to those seen with cyclosporine.48
Azathioprine-induced hepatitis has been reported more Similar to cyclosporine, tacrolimus levels were higher in
frequently in kidney transplant recipients with chronic HCV-positive renal transplant recipients, presumably sec-
viral hepatitis. In one study of 1035 transplant recipients, ondary to impaired cytochrome P-450-related metabolism
32 • Liver Disease Among Renal Transplant Recipients 543

of tacrolimus.51 Unlike cyclosporine, tacrolimus is not asso- high affinity, preventing T cell activation by blocking inter-
ciated with reductions in bile flow and choledocholithiasis. action of CD80/86 with CD28. To date, there have been no
Also tacrolimus was associated with less hyperbilirubinemia reports of hepatotoxicity related to belatacept.
(0.3%) compared with cyclosporine (3.3%) in renal trans-
plant recipients in a large comparative trial.52 Elevations in
aminotransferases are generally mild, even with suprath-
erapeutic levels, and reversible with dose reduction.53
Hepatitis Viruses Associated With
Renal Transplantation
SIROLIMUS HEPATITIS B VIRUS
Sirolimus (rapamycin) is an mammalian target of rapamy-
cin (mTOR) inhibitor that is structurally related to tacro- HBV Viral Structure and Proteins
limus. Sirolimus-induced hepatotoxicity is uncommon. Hepatitis B is a hepatotropic enveloped, partially double-
Elevations of aminotransferases with nonspecific histologic stranded DNA virus that is a member of the hepadnavirus
changes have been reported.54,55 The liver test abnor- family.60 The core of the virus comprises an RNA-dependent
malities have resolved with discontinuation of sirolimus. DNA polymerase plus a partially double-stranded DNA.
Sirolimus hepatotoxicity has been better described in liver After entry into the hepatocyte, the HBV enters the nucleus
transplant recipients. Of 10 patients treated with sirolimus, and forms what is known as covalently closed circular DNA
two had sinusoidal congestion and one had eosinophilia (cccDNA). This DNA is produced by repair of the gapped
consistent with a drug-related allergic reaction. Increases virion DNA and is the likely source of the transcripts used to
in aminotransferases were mild and normalized in all produce the viral proteins. The genome of the HBV encodes
patients by 1 month.24 Another study analyzed a cohort of four different genes. The C gene encodes core protein, the
97 patients treated with sirolimus-based immunosuppres- P gene encodes the hepatitis B polymerase, the S gene
sion post liver transplant. Surprisingly, 61 patients discon- encodes three different polypeptides of the envelope (pre-S1,
tinued treatment because of adverse effects, including 21 pre-S2, and S), and the X gene encodes proteins potentially
patients that discontinued treatment because of hepatotox- involved in the transactivation of viral replication.
icity.56 Cyclosporine, but not tacrolimus, can interfere with The hepatitis B viral antigens consist of the hepatitis B
sirolimus pharmacokinetics, and caution must be exercised core antigen (HBcAg) and a subunit of the core called the
when combining these agents. hepatitis B e antigen (HBeAg). The HBeAg is released in
high concentrations in the plasma during viral replication
MYCOPHENOLATE MOFETIL, MYCOPHENOLIC and is an indirect marker of active viral replication. The
envelope protein is referred to as the hepatitis B surface
ACID antigen (HBsAg) and is likely responsible for viral binding to
Mycophenolate mofetil is an ester of mycophenolic acid that the hepatocyte. HBsAg is released in excess in the serum in
is readily absorbed. It inhibits purine synthesis by noncom- individuals with chronic hepatitis B infection. Its presence
petitively inhibiting a key enzyme in the de novo purine in individuals 6 months after exposure to HBV defines the
pathway, inosine monophosphate dehydrogenase. Hepato- presence of chronic hepatitis B infection.
toxicity is exceedingly uncommon but has been reported in Presently, there are eight distinct genotypes of HBV. The
isolated cases.23 prevalence of these distinct genotypes varies geographi-
cally. Although there is growing evidence that the HBV
genotype may have implications for treatment success,
MONOCLONAL ANTIBODIES seroconversion, severity of liver disease, and development
Monoclonal antibodies are commonly used as induction of hepatocellular carcinoma (HCC), current management
immunosuppression in kidney transplantation. Use of does not change with HBV genotype and thus is not rou-
alemtuzumab (Campath; anti-CD52 humanized antibody) tinely determined.61
has been shown to accelerate hepatic fibrosis in HCV-
infected transplant recipients and should generally be Tests for Detection of Hepatitis B
avoided in solid-organ recipients with chronic viral hepa- HBV can cause acute and chronic infections. Acute infec-
titis.57 Anti-CD3 antibodies are used less often now for sal- tion is associated with acute hepatitis characterized by
vage of refractory rejection but have rarely been associated inflammation and hepatocellular necrosis. The diagnosis
with severe hepatitis and elevation of aminotransferases up rests on detecting HBsAg in the serum of a patient with clin-
to 20-fold.58 Cytokine-mediated reactions presumably can ical and laboratory evidence of acute hepatitis (Table 32.2).
cause the occasional hepatotoxicity seen with anti-CD3 Patients with a silent, self-limiting infection are able to pro-
antibodies. The interleukin-2 receptor antibody basilix- duce protective antibody (HBsAb) and ultimately clear the
imab has only been reported to cause hepatotoxicity in case virus. These patients are negative for HBsAg but positive for
reports in children.59 HBsAb and HBcAb.
Chronic HBV infection is accompanied by evidence of
hepatocellular injury and inflammation and is associated
T CELL COSTIMULATORY INHIBITOR with chronic hepatitis. The diagnosis is made by show-
Belatacept is a fusion protein designed to inhibit T cell acti- ing persistently elevated serum transaminases and HBsAg
vation by blocking a costimulatory pathway. Belatacept in the serum at least 6 months after exposure to HBV
binds CD80 and CD86 on antigen-presenting cells with infection.61
544 Kidney Transplantation: Principles and Practice

TABLE 32.2 Commonly Used Tests for Detection of and decreasing HBV DNA. Immune clearance can fail
Hepatitis B Infection and Their Interpretation and lead to recurrent phases of HBV replication accompa-
nied by surges of serum HBV DNA and aminotransferases,
HBsAg Anti-HBs Anti-HBc Interpretation which increase the risk of fibrosis progression toward cir-
+ – – Early acute infection rhosis and HCC. Some patients can further enter into the
+ – + Acute or chronic infection “inactive carrier state” with disappearance of the HBeAg
– + + Cleared HBV infection—immune from serum and development of anti-HBe antibodies. These
– + – Vaccine response—immune
patients have detectable HBsAg and may have low levels of
HBc, hepatitis B core; HBsAg, hepatitis B surface antigen; HBV, hepatitis B virus. HBV viremia, but aminotransferases are normal or near-
normal and there is little to no necroinflammation on liver
biopsy. Even in the inactive carrier state, patients can revert
Epidemiology of HBV to HBeAg positivity and develop evidence of chronic hepa-
Routes of Transmission. Hepatitis B is widespread world- titis. Therefore they require lifelong follow-up. In addition,
wide with more than a billion individuals estimated to be some patients remain HBeAg-negative, but develop evi-
carrying the virus. Areas of high incidence include China, dence of ongoing chronic hepatitis marked by HBV viremia,
Southeast Asia, and sub-Saharan Africa.62,63 Worldwide, elevated aminotransferases, and ongoing necroinflamma-
more than 350 million people have chronic HBV infection, tion on liver biopsy.65,66 Most of these patients are felt to
and in the US alone more than 1 million individuals are esti- have virus with a mutation in the precore or core promoter
mated to have chronic infection.63 HBV is transmitted via region of the viral genome. Serum HBsAg positivity is lost
perinatal, parenteral, or sexual exposure; transmission via infrequently.
the fecal-oral route does not occur. In countries with a high The outcomes of chronic HBV infection vary from an
prevalence of hepatitis B infection the route of transmis- inactive carrier state to cirrhosis and its attendant compli-
sion is mainly vertical, at childbirth or, to a lesser degree, cations, such as variceal hemorrhage, ascites, and enceph-
horizontally among household contacts in the first decade alopathy. Risk for liver disease progression is increased in
of life. In countries with a lower prevalence of hepatitis B older patients, patients with higher HBV DNA levels, in
infection, the majority of infections occurs in adulthood, patients coinfected with human immunodeficiency virus
and they are transmitted sexually and to a lesser extent by (HIV), HCV, or HDV, and with concomitant toxin exposures
intravenous drug use.63 such as alcohol, smoking, or aflatoxin.61 In addition, the
risk of HCC is elevated in chronic HBV, even in the absence
Natural History of HBV Infection. Hepatitis B can result of cirrhosis.
either in a self-limited acute infection or progress to chronic
liver disease. Progression to chronic hepatitis B infection
Hepatitis B Infection in Patients Awaiting Renal
after acute infection depends on the age of exposure to the Transplant on Dialysis
virus. The risk of developing chronic HBV infection is over The incidence and prevalence of hepatitis B infection among
90% for vertically acquired virus. The risk of chronic HBV patients awaiting renal transplantation have declined in
infection in young children (<5 years old) is 25% to 30%. recent decades, in large measure because of hepatitis B
Clinically symptomatic infection is rare in children. Con- vaccination of patients on dialysis and improved infection
versely, transmission in adulthood is associated with clini- control measures during dialysis. Before hepatitis B vac-
cally apparent hepatitis in over 30% of individuals (>90%).63 cination, 3% to 10% of patients on dialysis developed this
Acute infection in adults when clinically apparent is often disease,67,68 with even higher incidences reported from
associated with jaundice and elevated aminotransferases countries with a high prevalence of HBV infection. Pres-
with liver histology revealing portal inflammation, inter- ently, about 1% of patients on dialysis in the US are infected
face hepatitis, and lobular inflammation. Eventually, often with HBV, with a higher prevalence seen in developing
over several weeks, the jaundice resolves and aminotrans- countries.69-71
ferases are more modestly elevated. Eventually, over 80% of HBV vaccination is important for the prevention of HBV
nonimmunosuppressed adults who develop acute hepatitis transmission during hemodialysis. One case control study
B will not progress to chronic infection (HBsAg-negative, demonstrated a 70% reduction in risk of acquiring HBV
HBsAb-positive, HBcAb-positive). However, in dialysis among hemodialysis patients that underwent HBV vaccina-
patients, exposure to acute HBV results in chronic infection tion.72 Universal vaccination of dialysis patients, although
in the majority of nonvaccinated individuals (80%), likely recommended, is not universally undertaken. One survey of
because of their immunocompromised state and inability to 12 centers from 11 countries showed routine vaccination
mount protective antibody and T cell responses.64 of nonimmune subjects in only 66.7% (8 of 12) of centers.
The natural history of chronic hepatitis B infection Vaccination has a lower response rate in end-stage
depends on the age at which infection occurs. After peri- renal disease (ESRD) patients, with 50% to 60% of dialysis
natally transmitted infection there is an immune-tolerant patients developing adequate titers of anti-HBs antibod-
phase in which high levels of viral replication (with high ies.73,74 Similarly, success of HBV vaccination correlates
serum HBV DNA levels) are accompanied by minimal with glomerular filtration rate (GFR) and thus “earlier”
injury on liver biopsy and normal serum liver enzymes. vaccination is more successful.75 Despite lower rates of anti-
The immune-tolerant phase can last from the first up to HBs development, there is some evidence that vaccination
the third decade of life, after which transition occurs to the confers protective T cell responses and there are reduced
immune clearance phase.63 In this phase immune activ- rates of HBV infection even if anti-HBs antibodies are not
ity against HBV is noted by elevated levels of liver enzymes detected in vaccinated dialysis patients.76
32 • Liver Disease Among Renal Transplant Recipients 545

There are several additional strategies to improve the and renal allograft survival was reduced compared with
success of HBV vaccination, including intramuscular injec- recipients not infected with chronic hepatitis B.64 Over-
tions, doubling of vaccine dose, giving additional booster all mortality was not different between HBV-positive and
doses, and prompt revaccination in nonresponders.77 In HBV-negative recipients in this study. A study of 51 renal
addition to the previous strategies, doubling the vaccine transplant recipients with chronic hepatitis B infection
dose, giving an additional booster dose, and promptly found reduced patient survival and a higher incidence of
repeating the HBV vaccination series in nonresponders death caused by liver failure in the hepatitis B group (44%)
can be considered. Nonresponse is defined as an antiHBs compared with nonhepatitis-infected controls (0.6%). In
antibody titer less than 10 IU/L 1 to 2 months post series multivariable analysis in the hepatitis B group the presence
completion.78 Annual testing of anti-HBs titers should be of hepatitis B antigen was not an independent predictor of
undertaken with boosters given whenever the anti-HBs death; patient age, serum creatinine, and proteinuria at
titer falls under 10 IU/L. 3 months after transplant were independent predictors of
Clinical and histologic outcomes in dialysis patients with reduced patient survival.82
HBV infection are generally similar to that seen in immu- Other large studies have found significant reductions in
nocompetent individuals. The majority of these individuals long-term patient and graft survival in HBsAg-positive kid-
do not die of liver disease. In one study of dialysis patients ney transplant recipients compared with noninfected renal
in which 30% were infected with HBV, fewer than 5% died transplant recipients. In a cohort of 128 renal transplant
from liver disease. This may be as a result of the presence recipients infected with HBV, the 10-year survival was 55%
of other comorbidities (competing causes of mortality) in compared with 80% in non-HBV-infected renal transplant
their patients such as cardiovascular disease or infections recipients.83 Age at transplant and presence of cirrhosis
in addition to insufficient length of follow-up.79 The effect were independent prognostic factors for survival in this
of antiviral therapy on the natural history of chronic HBV study. Another study found a significant difference in long-
infection on hemodialysis patients has not been studied. term survival between hepatitis B-positive recipients com-
pared with recipients without chronic viral hepatitis66 with
Pretransplant Management of Hepatitis B-Positive a relative risk (RR) of mortality of 2.36 for 42 HBsAg-posi-
Dialysis Patients tive recipients. Finally, a meta-analysis that included 6050
Liver enzymes (aminotransferases) do not accurately reflect renal transplant recipients found increased mortality (RR of
the stage of liver disease in patients with chronic viral death with HBsAg positivity 2.49) associated with chronic
hepatitis and ESRD. Patients with chronic HBV on dialysis hepatitis B infection and reduced graft survival (RR of graft
should have imaging for assessment of liver fibrosis before loss 2.49).84
renal transplantation. Newer imaging based on noninva- Differences in outcome between studies may result from
sive measurements of fibrosis such as transient elastrog- small numbers in some studies, length of follow-up, hetero-
raphy is more accurate in distinguishing minimal or no geneity of patient characteristics such as age at transplant,
fibrosis from advanced fibrosis and cirrhosis than serum replicative state of hepatitis B, presence or absence of cirrho-
markers, although in hepatitis B these data are extrapolated sis at time of transplant, and the confounding effect of anti-
mostly from nondialysis patients.80 Patients with fibroscan viral therapy for hepatitis B. Studies with larger numbers,
demonstrating elevated values of fibrosis (F2 or greater) longer follow-up, and with matched case-control design
should proceed to a liver biopsy. Patients with cirrhosis on and multivariate analysis have tended to show a reduction
the biopsy should be considered for a combined liver–kid- in patient and graft survival associated with chronic hepa-
ney transplant when portal hypertension develops. titis B infection in renal transplant recipients.
Criteria for antiviral therapy in nontransplant patients Several studies have documented the progression of
include evidence of chronic necroinflammation of the liver, fibrosis in HBsAg-positive kidney transplant recipients after
evidenced by an elevated ALT and aspartate aminotrans- transplant. In a study of 151 HBsAg-positive kidney trans-
ferase (AST) in the setting of HBeAg positivity or in the set- plant recipients, 28% had a histologic diagnosis of cirrhosis
ting of an elevated serum HBV DNA in HBeAg-negative at a mean of 66 months posttransplant.64 HCV coinfection
patients.81 However, in patients undergoing renal trans- was the only identifiable risk factor for fibrosis progression.
plantation there is increased risk of reactivation of viral More recently, a cohort of 55 HBsAg-positive kidney trans-
replication and increased viral replication after transplan- plant recipients underwent liver biopsy at a mean of 5 years
tation with exposure to immunosuppressive agents. In after transplantation. On logistic regression, the only risk
addition, HBV-positive renal allograft recipients have worse factor for the development of cirrhosis was the time interval
outcomes in terms of liver disease and renal allograft func- between kidney transplant and liver biopsy.85
tion (discussed later). Therefore it is prudent to start anti- In rare cases viral replication may become uncontrolled
viral therapy before renal transplantation for patients with in the setting of immunosuppression after renal transplan-
evidence of active viral replication. This includes patients tation. In this state the virus may become directly cytopathic
with positivity for HBsAg and/or any detectable viral load. and lead to a state of hepatocellular failure with profound
cholestasis. The liver biopsy is characteristic with hepato-
Posttransplant Prognosis in Hepatitis B Recipients cyte ballooning, cholestasis, and perisinusoidal fibrosis.
Post–renal transplantation, hepatitis B-infected recipients This condition is called fibrosing cholestatic hepatitis, and
are generally felt to have decreased survival compared with was first described in liver transplant recipients infected
noninfected recipients, although this finding is controver- with HBV.86 Once established, the prognosis is poor, even
sial. In one study of 1250 renal allograft recipients, with a with antiviral therapy. Preemptive suppressive antivi-
median follow-up of 125 months, cirrhosis occurred in 30% ral therapy is the judicious strategy to prevent this feared
546 Kidney Transplantation: Principles and Practice

outcome. In rare cases the suppression of viral replication the rituximab group, two patients experienced HBV-related
with long-term antiviral therapy has resulted in salvage of severe hepatitis, and one patient died as a result of hepatic
liver and graft function (discussed later). failure. The median time from rituximab desensitization to
The natural history of chronic HBV infection in kidney HBV reactivation was 11 months (range, 5–22 months).
transplant recipients in the era of antiviral therapy is less By contrast, no patients in the control group experienced
well studied. A recent, small study of 63 HBsAg-positive severe hepatitis. Rituximab desensitization (hazard ratio
kidney transplant recipients revealed an improved 20-year [HR], 9.18; 95% confidence interval [CI], 1.74–48.86;
mortality (83% vs. 34%, P = 0.006) in patients treated with P = 0.009) and hepatitis B surface antibody status (HR,
antiviral therapy.87 4.74; 95% CI, 1.05–21.23, P = 0.04) were significant risk
factors for HBV reactivation.93
De Novo HBV Infection After Kidney Ultimately, prevention of de novo hepatitis B in renal
Transplantation transplant recipients is best achieved by universal vacci-
Development of de novo hepatitis B after renal transplan- nation of all dialysis patients. Alternatively, organs from
tation can be associated with rapid viral replication and HBsAg-positive donors can be offered only to recipients with
progression of liver disease.88 The hepatitis B serologic and preexisting HBV infection or those individuals who have
virologic status of the donor and recipient are important been successfully vaccinated for HBV. Use of HBcAb-posi-
risk factors that predict development of de novo hepatitis tive donors is often center-specific. If such organs are used,
B infection after renal transplantation. The highest risk of posttransplant usage of prophylaxis with antiviral medica-
de novo hepatitis exists in recipients who are nonimmune tion or hepatitis B immune globulin should be considered,
for hepatitis B (HBsAb-negative) and receive an organ especially in patients without evidence of HBV immunity.
from HBsAg/HBeAg-positive donors. The risk of transmis-
sion from an HBcAb-positive-only donor (HBsAg-negative, Antiviral Therapy of Chronic Hepatitis B in Renal
HBcAb-positive, negative serum HBV DNA donor) to a hep- Transplant Candidates/Recipients
atitis B-negative recipient also exists, although it is reduced Data regarding the optimal timing of antiviral therapy for
compared with that seen in liver transplant recipients.89,90 HBV in renal transplant candidates are scarce (Table 32.3).
It is important to note that isolated HBcAb positivity could The risks of liver disease progression and severe hepatitis
represent an early, acute HBV infection or possibly a long- B reactivation posttransplant have to be weighed against
standing, chronic infection with low-level HBV viremia. the risk of antiviral toxicity and viral resistance develop-
Determination of donor IgM HBcAb should be performed ing. However, with the development of the newer-genera-
in patients with an isolated positive HBcAb. Positive titers tion antinucleos(t)ide analogs entecavir and tenofovir (see
for IgM suggest a recent infection and should be consid- later), the risk of viral resistance is much lower than with
ered high risk for transmission of HBV to the recipient. HBV lamivudine or adefovir. Data for antiviral therapy posttrans-
DNA should also be determined in the isolated HBcAb-pos- plant have mostly been performed using lamivudine. In one
itive donor with consideration of HBV prophylaxis for the trial, the efficacy of lamivudine in preventing viral replica-
recipient. tion after renal transplantation was compared in HBsAg-
The risk of de novo HBV infection is considerably reduced positive recipients using three strategies: (1) preemptive
if the recipient is positive for HBsAb, although it is not com- lamivudine therapy (HBV DNA-positive recipients, received
pletely eliminated. In one series where HBcAb-positive- lamivudine therapy 0–9 months before renal transplant,
only donors were used for recipients with a prior history of n = 7); (2) prophylactic lamivudine therapy (HBV DNA-
hepatitis B or HBV vaccination, none developed clinically negative, received lamivudine therapy before transplant,
evident hepatitis B, although 27% did develop HBcAb and/ n = 3); and (3) salvage therapy (HBV DNA-positive, advanced
or HBsAb positivity after transplant.91 In a more recent hepatic dysfunction after transplant, received lamivudine
study from Italy, 344 patients received anti-HBcAb-posi- after transplant after hepatic dysfunction, n = 6).94 HBV
tive allografts and no recipient developed HBsAg positivity, DNA disappeared in all recipients in all groups on therapy.
including 62 patients that had not undergone HBV vacci- The recurrence rate of HBV viremia was 10% (1 out of 10)
nation.90 Finally, a cohort of 46 patients that received an in the preemptive and prophylactic group compared with
anti-HBcAb-positive donor kidney were followed for 36 42% (11 out of 25) in a nonlamivudine-treated group. In
months posttransplant. Anti-HBsAb-positive (immunized) the group treated for hepatic dysfunction HBV DNA dis-
recipients received no prophylaxis. Naïve patients received appeared in all 6 cases but recurred in 50% (3 out of 6)
1 year of lamivudine prophylaxis. No patients developed while on lamivudine. In another trial of lamivudine ther-
evidence of HBV viremia or development of HBsAg.92 apy, HBV DNA levels were measured and lamivudine was
HBV reactivation is a well-known complication of immu- started before renal transplantation if the HBV DNA rose
nosuppressive therapy. Although rituximab is increasingly to more than 2.83 × 108 copies/mL alone or to >2.83 ×
used for desensitization of ABO-incompatible or positive 107 copies/mL with elevated AST/ALT from 1996 to 2000
crossmatch kidney transplantation, the risk of HBV reac- (so-called de novo group).95 This strategy was compared
tivation in HBsAg-negative/hepatitis B core antibody with preemptive use of lamivudine for patients who had
(anti-HBc)-positive kidney transplant patients receiving undergone transplantation before 1996 (when lamivu-
rituximab desensitization remains undetermined. In a study dine became commercially available) and thus received
of 172 resolved HBV patients who underwent living donor therapy later after transplantation than the de novo group.
kidney transplant 5 of 49 patients who received rituximab Even though suppression of HBV DNA and normalization
(10.2%) had HBV reactivation compared with 2 of 123 of aminotransferases were achieved in all patients, the sur-
control patients who did not receive rituximab (1.2%). In vival of the de novo treated group was comparable to that of
TABLE 32.3 Selected Pretransplant and Posttransplant (Nonliver) Studies of Antiviral Therapy in HBV Patients
HBeAg
Number HBV Antiviral Duration of HBV DNA Seroconversion
Study Patient Population in Study Therapy Therapy Suppression to Anti-HBe Virologic Breakthrough
PRETRANSPLANT
Fontaine et al.266 Dialysis patients 5 Lamivudine 10 12 months 5/5 1/5 2/5 (at months 7, 18 of
mg daily in 3, (7–28) lamivudine)
50 mg thrice
weekly in 2
Duarte et al.267 Dialysis patients 2 Interferon-alfa 3 months 2/2 2/2 None
3 mu thrice
weekly
POSTTRANSPLANT
Fontaine et al.266 Postrenal transplant patients 26 Lamivudine 16.5 months 26/26 undetectable 6/26 8/26
with HBV infection 100 mg/day (4–31)
Hu et al., 2012107 Postrenal transplant patients 27 Entecavir 104 weeks 100% undetectable 3/5 HBeAg-positive 0/27
with HBV infection after 104 weeks seroconverted
after 35 weeks
Fontaine et al.103 Post kidney transplantation 11 Adefovir 15 months Median change –5.6 0/6 that were Not detected
with lamivudine-resistant 10 mg/day (3–19) log copies/mL initially HBeAg+
HBV (–2.2 to –7.7)
Han et al. 200194 Post kidney transplantation Group 1: After developing Lamivudine Group 1: On treatment Group 1: 0/6 Group 1: 3/6
with HBV (HBsAg+) recurrent hepatic dysfunction 100 mg/day Follow-up Group 1: 6/6 Group 2: 0/11 Group 2: 1/10
after renal transplant (6) 15–60 months On treatment
Group 2: Preemptive or prophylactic Group 2: Group 2: 11/11
treatment for HBsAg-+ recipients Follow-up
beginning before renal 9–30 months
transplantation (10)
Chan et al. 200295 Post kidney transplantation Period II: Post 1996. De novo Lamivudine Period I: 26/26 undetectable Not mentioned. 11 (40.7%) became
with HBV (HBsAg+) preemptive therapy before renal 100 mg/day 36.3 ± 11.4 3/14 HBeAg+ lamivudine-resistant
transplantation and continued months patients became at 9.5–24 months
after transplantation (11) Period II: undetectable after starting treat-
Period I: pre-1996. Preemptive ther- 27.6 ± 14.5 ment

32 • Liver Disease Among Renal Transplant Recipients


apy after renal transplantation months
Puchhammer- Post kidney transplantation 11 Lamivudine >12 months HBV undetectable in Not reported Lamivudine resistance
Stockl et al., with HBV (HBsAg+) 100 mg/day 10/11 undetect- in 5/11 from 9 to 15
200099 in 7, reduced able by PCR months after starting
dose in 4 per lamivudine
renal function
Thabut et al. Post kidney transplantation 14 Lamivudine Median duration 11/11 undetectable 0 of 4 HBeAg+ Lamivudine resis-
2004268 with HBV (HBsAg+) 100 mg/day 64.5 months on treatment patients tance with virologic
(6–93) breakthrough in 8/14
patients from 9 to 24
months after starting
lamivudine
Chan et al., 2004269 Post kidney transplantation 29 Lamivudine 56.7 ± 12.5 29/29 undetectable 5/15 who were 14/29 (48%) developed
with HBV 100 mg/day months on treatment HBeAg + lamivudine resistance
(HBsAg+) initially (10–35 months after
starting treatment)
Fabrizi et al., Post kidney transplantation 184 Lamivudine Variable 91% HBV DNA 27% in 4 of 14 18% in 8 of 14 studies
200496 with HBV (HBsAg+) (meta-analysis of 14 studies) 50–150 mg/ undetectable studies
day
Kamar et al., 2008108 Posttransplantation with 10 (8 kidney) Entecavir 0.5 Median 16.5 5/8 kidney recipients Not reported Not reported
lamivudine or adefovir-­ mg/day titrated months developed unde-
resistant HBV (HBsAg+) to 1.0 mg/day tectable HBV DNA

547
after 1 month

HBeAg, hepatitis B e antigen; HBsAg, hepatitis B surface antigen; HBV, hepatitis B virus; mu, million units; PCR, polymerase chain reaction.
548 Kidney Transplantation: Principles and Practice

HBsAg-negative controls, whereas HBsAg-positive patients was associated with an increased serum creatinine and
who were transplanted before 1996 and received preemp- increased proteinuria at 2-year follow-up. In addition,
tive therapy with rising HBV DNA after renal transplanta- there was evidence for proximal tubular dysfunction with
tion had a higher risk of overall (RR 9.7) and liver-related adefovir usage.105 Given the risk of nephrotoxicity, adefovir
mortality (RR 68.0). More recently, a meta-analysis of 14 should be used with caution in kidney transplant recipients.
clinical trials using lamivudine in kidney transplant recipi-
ents demonstrated that HBV DNA clearance occurred in Entecavir. Entecavir, an analog of 2′-deoxyguanosine, is
91% and normalization of ALT occurred in 81% of treated a nucleoside analog with potent activity against HBV repli-
patients.96 cation. In a randomized, controlled trial of HBeAg-positive
Antiviral therapy should thus be offered to all hepatitis nontransplant patients, 48 weeks’ treatment with enteca-
B-positive (HBsAg-positive) kidney transplant recipients, vir, dosed at 0.5 mg/day, resulted in higher rates of histo-
including those on the waiting list. This recommendation logic, virologic (undetectable HBV DNA), and biochemical
applies even to surface antigen-positive patients with a (normalization of ALT) response compared with lamivu-
negative HBV DNA. The optimal duration of therapy is yet dine 100 mg/day.106
to be determined and in an immunocompromised host may In another study in renal transplant recipients, entecavir
need to be indefinite. Cessation of antiviral therapy in the was compared with 3-TC. Of the 27 recipients, 18 (67%)
immunocompromised host is associated with an increased were NUC-naïve patients, and 9 (33%) were 3TC-experi-
risk of flare-up of liver disease and, rarely, decompensated enced without YMDD mutations. HBV DNA levels became
liver disease in both the transplant recipient and patients undetectable in 70%, 74%, 96%, and 100% of patients
without organ transplantation.95,97 after 12, 24, 52, and 104 weeks, respectively, of entecavir
treatment without viral resistance. By comparison with the
Specific Antiviral Agents for HBV Used in Renal 19 3TC-treated patients, ETV-treated recipients presented
Transplant Recipients higher rates of undetectable HBV DNA than 3TC-treated
Lamivudine. The cytosine analog lamivudine has been recipients (32%, 37%, 63%, and 63% at 12, 24, 52, and 104
the most extensively studied antiviral for HBV. A dose of weeks, respectively; P < 0.005). There was no change of GFR
100 mg/day has been shown to be highly effective in sup- and no lactic acidosis or myopathy during treatment.107
pression of HBV replication and normalization of amino- In a study where 10 transplant recipients (8 kidney) with
transferases in over 80% of individuals.96,98–100 Cessation adefovir or lamivudine resistance were treated with enteca-
of antiviral therapy has been associated with virologic and vir, mean HBV DNA levels decreased and HBV DNA clear-
clinical relapse.100 ance was achieved in 50%.108
The major drawback with lamivudine is the high rate of Unlike lamivudine, resistance to entecavir is low in treat-
viral resistance. The risk of resistance increases with dura- ment-naïve patients. In phase III trial data, viral break-
tion of lamivudine therapy. In a meta-analysis of 14 clinical through was only seen in 3.6% of treatment-naïve patients
trials (184 recipients) of lamivudine after renal transplan- at 96 weeks of entecavir therapy.109 However, entecavir
tation, the majority of recipients had HBV DNA clearance should be used with caution in patients with lamivudine
(91%) and biochemical normalization (81%), and the risk resistance or viral breakthrough while on lamivudine
of lamivudine resistance was 18%.98 Although HBeAg loss therapy. In a study of nontransplant patients on 5 years
was higher with prolonged therapy, the resistance was also of entecavir therapy for chronic HBV, entecavir resistance
higher, thereby limiting its efficacy. Given the high risk of developed in 51% of patients with documented lamivudine
viral resistance, lamivudine is no longer preferred as first- resistance.110
line therapy for HBV.81 Entecavir should be considered a first-line treatment for
kidney transplant candidates and recipients with chronic
Adefovir. Adefovir dipivoxil, an oral prodrug of adefovir, is HBV that have no concern for lamivudine resistance.
a nucleotide analog of adenosine monophosphate. Adefovir
therapy has demonstrated efficacy in treatment-naïve and Tenofovir. Tenofovir disoproxil fumarate is a nucleotide
lamivudine-resistant patients with HBV.42,101,102 Standard analog originally approved for therapy against HIV. Teno-
adefovir dosage is 10 mg/day. The dose should be adjusted fovir is structurally similar to adefovir, but less nephrotoxic,
based on GFR. In patients with a renal transplant it has allowing for higher dosing and a more potent antiviral effect.
been used in small studies, mostly reported in lamivudine- In randomized, controlled trials of nontransplant patients
resistant recipients. In one study of 11 renal transplant with chronic HBV, tenofovir has been shown to be an effective
recipients, there was a significant reduction in HBV DNA antiviral against HBV. In a phase III trial of tenofovir versus
after initiation of adefovir with a median decline of 5.5 adefovir in HBeAg-positive patients, after 48 weeks of therapy
log in HBV DNA after 12 months of therapy. No virologic a greater proportion of tenofovir-treated patients achieved a
breakthrough was observed and no significant changes in negative HBV DNA (76% vs. 13%), ALT normalization (68%
creatinine occurred.103 Adefovir resistance is much less vs. 54%), and surface antigen loss (3% vs. 0%).
common than with lamivudine, even after prolonged ther- Tenofovir resistance appears rare. In the original phase
apy.104 If adefovir is used in patients with lamivudine resis- III clinical trials, no patients had genotypic evidence of
tance, lamivudine should continue to be administered and mutations known to cause tenofovir resistance. Unlike
dual therapy continued indefinitely. entecavir, tenofovir is effective in the setting of lamivudine
The principal drawback to adefovir therapy is the risk resistance. In a randomized trial of HIV/HBV-coinfected
of nephrotoxicity. In a recent study of 11 renal trans- patients known to be lamivudine-resistant, both adefovir
plant recipients with chronic HBV, adefovir therapy and tenofovir were found to be efficacious in decreasing
32 • Liver Disease Among Renal Transplant Recipients 549

HBV DNA at 48 weeks.42 Tenofovir should be used with immunized against HBV. HBV vaccination is more success-
caution in cases of known adefovir resistance. ful at higher GFR and therefore should ideally be adminis-
Data regarding tenofovir in transplant recipients are tered well before the onset of hemodialysis.
scarce. A recent study of seven transplant (3 kidney) All patients who are candidates or who have undergone
patients with chronic HBV treated with tenofovir showed a kidney transplantation and are positive for HBsAg should
decline in HBV DNA during treatment, with three patients undergo a liver biopsy and be given antiviral therapy to
achieving serum DNA clearance.111 Despite the lack of data decrease the risk of liver disease progress or severe HBV
in kidney transplant recipients, tenofovir should be consid- exacerbation after initiation of immunosuppression. Teno-
ered a first-line agent for the treatment of chronic HBV in fovir and entecavir should be considered first-line antiviral
kidney transplant candidates and recipients. therapy because of their potency, tolerability, and the low
risk of resistance development.
Interferon. Use of interferon is associated with an unac-
ceptably high risk of precipitating renal allograft rejection, HEPATITIS C VIRUS
sometimes irreversibly, despite salvage immunosuppressive
therapy. Its use in the renal transplant recipient should thus Viral Structure
be avoided given the availability of other antiviral agents The discoveries of hepatitis A virus (HAV) and HBV
for hepatitis B.112,113 between the years of 1967 and 1973116 were a medical
breakthrough; however, it left many unanswered ques-
Treatment of Fibrosing Cholestatic Hepatitis B in tions. For the next 16 years, patients with non-A non-B
Renal Transplant Recipients hepatitis became increasingly recognized as having a form
Fibrosing cholestatic HBV is a histologic and clinical vari- of chronic liver disease. In 1989 Choo et al.117 published
ant of hepatitis B characterized by hepatocyte ballooning, the first account of HCV, which was further described as a
cholestasis, minimal inflammation, periportal fibrosis, and single-stranded, enveloped, positive-sense RNA virus. It is
massive viral replication (Fig. 32.2). It was first described classified in the Flaviviridae family.
in HBV-infected recipients of liver allografts but has also
been subsequently described in other immunosuppressed HCV Species
states.114 Patients often develop rapidly progressive liver HCV can be thought of as a spectrum of similar viruses.
failure and spontaneous recovery is rare. Lamivudine has Seven HCV genotypes with several distinct subtypes have
been reported to be useful in case reports, resulting in suc- been identified throughout the world.118 Within a genotype
cessful resolution of the severe acute hepatitis and hepatic or subtype, the genome of HCV is highly mutable because
failure associated with this condition.115 With appropriate of the lack of efficient proofreading capabilities. As the virus
antiviral therapy, fibrosing cholestatic HBV should occur replicates over time, selective pressures from the immune
extremely infrequently. system and/or antiviral treatments cause the viral popu-
lations to evolve. These mutant versions of genotypes are
Summary called “quasispecies.” The heterogeneity of this virus is
In summary, chronic HBV infection in kidney transplant what allows it to evade immunologic detection and elimi-
candidates and recipients has become less common in nation thus far, preventing the development of a vaccine.
developed countries. This decrease in prevalence and inci- Epidemiologic studies on the HCV genotypes have been
dence of new cases can be attributed to improved public performed demonstrating significant regional variation.
health efforts, particularly infection control measures dur- Genotype 1 is found worldwide although it is by far the
ing hemodialysis, and widespread HBV immunization. most common (60%–70% of isolates) in the US, Europe,119
All patients with chronic kidney disease (CKD) should be Japan, and Taiwan. Although less common genotypes 2
and 3 are also found in these areas, genotypes 4, 5, and 6
are rarely encountered. Genotype 3 is predominant in India,
the Far East, and Australia.120,121 Genotype 4 is present in
North Africa and the Middle East, with a particularly high
incidence in Egypt. Genotype 5 has been most frequently
detected in South Africa, whereas genotype 6 has been
rather isolated to Hong Kong.122
The significance of viral genotypes is not entirely clear,
but important clinical differences have been shown. Amo-
roso et al.123 followed patients with acute viral hepatitis
and found that those infected with genotype 1 developed
chronic infection at a significantly higher rate compared
with those with genotypes 2 and 3.
Clinical Manifestations of Hepatitis C Infection in
Immunocompetent Hosts
In general, HCV is a chronic infection and its acute form
Fig. 32.2 Perisinusoidal fibrosis and hepatocyte ballooning with- often goes unrecognized. Of patients with acute HCV 20%
out inflammatory infiltration. Characteristic histologic appearance of to 30% have symptoms 2 to 12 weeks after exposure.124,125
fibrosing cholestatic hepatitis B. The symptoms are generally mild and include lethargy,
550 Kidney Transplantation: Principles and Practice

nausea, vomiting, jaundice, and anorexia. Serum amino- Given the prevalence of chronic HCV among patients on
transferases can range from 2- to 10-fold above normal. hemodialysis, a significant number of patients on the renal
Rarely, acute HCV can lead to acute hepatic failure,126 transplant waiting list are infected with HCV. Accurate
although this is exceedingly uncommon. Diagnosis of acute data regarding infection rates in this transplant-associated
HCV is made by testing for HCV RNA, which can be iden- population are complicated by several factors, including the
tified in serum a few days to weeks after exposure.126,127 insidious and indolent nature of the disease in the setting of
Anti-HCV antibodies are typically not detected for weeks uremia,139 regional variations of the HCV genome, the use
to months after exposure and may not develop in immuno- of nonstandardized diagnostic methods, and the absence
compromised individuals or in patients on dialysis.128 of good prospective, well-powered studies. Risk factors for
Chronic HCV develops in about 85% of those who are HCV among transplant candidates include length of time
exposed. In the majority of patients, the clinical course is on hemodialysis, exposure to blood products before univer-
remarkably nonspecific. Fatigue and nonspecific arthral- sal screening, and the prevalence of chronic HCV infection
gias are common complaints and typically improve with in the dialysis center.
eradication of the virus.129 Studies have estimated 20% to
35% of patients will have progression of liver disease to cir- Allograft Transmission of HCV
rhosis over 20 to 30 years.130 A study by Cacoub et al.129 As transplant waiting lists soar to record levels, programs
found that 38% of HCV patients presented with at least one of all organ types are faced with decisions regarding the
clinical extrahepatic manifestation. The associated findings use of extended criteria (previously called marginal) donor
include hematologic disorders such as cryoglobulinemia, organs, including those positive for HCV antibody. Histori-
lymphoma, and porphyria cutanea tarda and other rashes. cally, allocation of HCV-positive organs has been restricted
Dry eyes and mouth, pruritus, renal disease including to HCV-positive recipients. This recommendation is based
membranoproliferative glomerulonephritis (MPGN), and on evidence that transplantation of HCV-positive organs
diabetes are often present. into HCV-negative recipients is a risk factor for poorer
outcomes in renal transplant patients.140 In contrast, out-
Incidence/Prevalence and Transmission of come data regarding kidney transplantation from HCV
Hepatitis C in Renal Transplant Patients antibody-positive donors to HCV-positive recipients are
­
It is estimated that 180 million people are infected with mixed. Recipient wait time may be substantially reduced
HCV worldwide, with 4 million people in the US thought and there appears to be no effect on short-term mortal-
to be HCV antibody carriers. Among those with anti-HCV ity.141–143 Similarly, registry studies have shown that
antibodies, about 80% are viremic.131 The principal risk kidney transplantation from deceased anti-HCV anti-
factors for HCV infection are transfusion of unscreened body-positive donors has a survival advantage compared
blood products and intravenous drug use. With the with staying on dialysis for HCV-positive recipients.140
development of blood donor screening in the 1990s, Recently with the availability of potent and highly effec-
transfusion-related HCV transmission is now exceedingly tive direct-acting antiviral agents for hepatitis C there has
rare.132 Other risk factors for HCV transmission include been interest in using organs from HCV-positive donors for
nosocomial transmission, including via hemodialysis and HCV-negative recipients. In a pilot randomized controlled
occupational exposure. Transmission of HCV via hemodi- trial HCV-positive kidneys were transplanted in 10 HCV-
alysis and occupationally is less frequent with the use of negative recipients who were then given early posttrans-
improved universal precautions. Sexual transmission is plant HCV antiviral therapy (preemptive treatment). All
felt to be rare. patients cleared the HCV with 12 weeks of therapy and had
The prevalence of HCV in patients with CKD is higher preserved renal allograft function and good liver function
than in the general population, particularly in patients on without significant adverse events.142,144
hemodialysis. HCV prevalence in hemodialysis units across Currently, the Kidney Disease Improving Global Out-
seven countries was reported in the Dialysis Outcomes and comes (KDIGO) practice guidelines recommend restrict-
Practice Patterns Study and showed a mean HCV preva- ing the use of allografts from HCV-infected donors to
lence of 13.5% with a range between the countries of HCV-infected recipients and this practice is still considered
2.6% to 22.9%. HCV prevalence is higher in Japan, Italy, investigational.145,146
and Spain and lower in Germany and the United King-
dom. The US had a 14% HCV prevalence and a hemodialy- Effect of Pretransplant HCV on Posttransplant
sis seroconversion rate of 2.5% per 100 patient years.133 Outcomes
However, in the US, there is high variability in the preva- Patient and Graft Survival. Some controversy exists
lence of chronic HCV among hemodialysis units based on regarding the effect of pretransplant HCV infection on the
location.134 Historically, blood products were the major outcome of renal transplantation (Table 32.4). Initially,
contributor to infection in these patients. As mentioned studies of short follow-up periods suggested that neither
previously, in the past decade this method of transmis- patient nor graft survival were altered posttransplant
sion has been virtually eliminated with reliable screening despite a logarithmic increase in HCV RNA levels.147–149
methods135,136 and decreased transfusion requirements Orloff et al. reported the liver biopsy findings at 3 to 7 years
directly related to the increased use of hematopoietic after kidney transplantation in HCV-positive subjects. Of
growth factors.135,137 Despite these improvements, studies these 12% had chronic active hepatitis, 50% showed mild
show de novo infections do occur in dialysis units, though hepatitis, and 38% had normal histology. Furthermore,
clearly identifiable risk factors have not been reproducibly hepatitis C conferred no adverse effect on patient or graft
demonstrated.138 survival.147 Lee et al. also determined that HCV infection
32 • Liver Disease Among Renal Transplant Recipients 551

TABLE 32.4 Outcomes in HCV-Positive Recipients Assessment of fibrosis stage in the patient with ESRD can
(Compared With HCV-Negative Recipients) Undergoing be done noninvasively using transient elastography-based
Renal Transplantation techniques such as fibroscan. This modality is good at dis-
tinguishing minimal fibrosis from advanced fibrosis and cir-
Type of Transplant Outcome
rhosis and can obviate the need for staging liver biopsy in
Renal transplant recipients Decreased long-term patient sur- those with low fibrosis scores.162
vival (follow-up >10 years) Most studies regarding posttransplant HCV outcomes
Decreased graft survival are directed to chronically infected recipients, usually sub-
De novo or recurrent glomeru-
lopathy jects who acquired HCV during hemodialysis. However,
Cirrhosis the subset of solid-organ transplant recipients who become
Posttransplant diabetes infected with HCV in the perioperative period have a mark-
edly different course. Delladetsima et al.163 followed 17
HCV, hepatitis C virus.
such patients by biochemical and histologic markers for a
mean of 7 years. Six (35%) patients died a median of 6 years
did not reduce renal allograft or patient survival; however, posttransplant because of: fibrosing cholestatic hepatitis,
they identified more liver disease and a greater prevalence vanishing bile duct syndrome, cirrhosis, miliary tubercu-
of life-threatening sepsis in the HCV-infected recipient losis, and myocardial infarction. Overall the yearly fibrosis
population.150 progression rate was five times that of age-matched immu-
In contrast, studies with more lengthy follow-up after nocompetent HCV-infected patients. These studies suggest
transplantation have found decreased patient and/or that HCV acquired at the time of transplantation may have
graft survival in HCV-positive renal transplant recipi- a particularly aggressive course.
ents.83,151–153 Periera et al.206 compared the prevalence
of posttransplantation liver disease and graft and patient HCV and Posttransplant Diabetes in the Renal
survival in HCV-positive and HCV-negative kidney trans- Transplant Recipient
plant recipients. Among recipients who were HCV-posi- The association of diabetes mellitus and HCV has become
tive before transplantation, the RR was 5.0, 1.3, and 3.3 increasingly apparent both in the immune-competent HCV
for posttransplantation liver disease, graft loss, and death, population and particularly after solid-organ transplanta-
respectively. There was a significant increase in death tion in HCV-infected patients. The overall incidence of post-
caused by sepsis with an RR of 9.9.154 Similarly, Hana- transplant diabetes mellitus (PTDM) has been reported to
fusa et al. found clinically significant hepatitis in 55% vary from 10% to 54% and has shown similar long-term
of HCV-positive kidney transplant recipients. They also effects as diabetes mellitus types 1 and 2, with cardiac and
found a significant decline in the 20-year survival in the renal dysfunction in a significant proportion.164 Yildiz et al.
HCV-positive patients compared with the HCV-negative reported a case-controlled study of 43 renal transplant
cohort (64% vs. 88%).153 recipients with PTDM in which 72% were HCV-infected,
The most common reasons for increased mortality in compared with a prevalence of 37% in the recipients with-
HCV-positive renal transplant recipients are excess risk of out PTDM (P = 0.002).165 This association was further
diabetes, cardiovascular disease, systemic infections, and observed by Bloom et al.166 where PTDM occurred more
cancer.155 frequently in HCV-positive than HCV-negative patients
In a meta-analysis of observational studies after renal (39.4% vs. 9.8%; P = 0.0005). Their data further found
transplantation that included eight studies, the presence that among the HCV-positive patients there was an eight-
of HCV antibody was an independent risk factor for death fold increased incidence of PTDM in those treated with
and graft failure after renal transplant (RR for death 1.79, tacrolimus (58%) compared with cyclosporine (7.7%).
95% CI 1.57–2.03) and for renal graft failure 1.56 (95%
CI 1.35–1.80). HCC and liver cirrhosis were more frequent HCV and Posttransplant Nephropathy
causes of mortality in HCV-positive than HCV-negative Posttransplant renal disease is common among HCV-pos-
recipients.156 itive recipients of any organ. Whereas the causes of renal
Despite the finding that graft and overall survival are injury after transplantation are multifactorial in nature,
probably decreased in kidney transplant recipients with chronic allograft nephropathy among renal transplant
chronic HCV infection, overall mortality has been shown to recipients and nephrotoxicity resulting from calcineurin
be improved with transplantation over long-term dialysis. inhibitors are the most common etiologies.167
HCV infection is associated with increased hospitalization, Kidney transplant recipients with chronic HCV infection
need for transfusions, and reduced quality of life in patients are at risk of additional immune-mediated nephropathies,
with ESRD.157 Dialysis patients with HCV have a 15% to with MPGN being the most common, followed by mem-
30% increased risk of mortality compared with patients branous nephropathy, minimal change disease, and renal
without HCV on dialysis.158,159 thrombotic microangiopathy. These may be recurrent or
HCV infection thus should not be considered a contra- present de novo.168 MPGN has been reported in 45% of
indication to consideration of kidney transplantation.160 HCV-positive renal transplant recipients who underwent
Particularly with the availability of highly effective antivi- renal biopsy for worsening renal function. In the HCV-neg-
ral therapy for hepatitis C that can be used both in ESRD ative group, the incidence was only 5.9%. De novo disease
and after renal transplantation, posttransplant outcomes was found in 18% of the MPGN patients and chronic renal
in HCV-positive recipients are expected to improve to those allograft nephropathy was similar in both HCV-positive and
seen in non-HCV recipients.161 negative recipients.169
552 Kidney Transplantation: Principles and Practice

Immunosuppressive Strategies in Renal (sustained virologic response [SVR]) or cure rates in excess
Transplant Patients Infected With HCV of 90% with treatment durations of 8 to 24 weeks.181
Specific antiviral treatments vary by genotype, presence
No studies have been performed to determine optimal
or absence of cirrhosis, and according to GFR. Recently
immunosuppressive regimens in renal transplant recipients
approved mediations include pangenotypic regimens that
infected with HCV. Viral replication is increased with the
can be used in patients with CKD, patients on dialysis, and
use of immunosuppressive agents, but the effect on patient
posttransplant.182
survival, progression of liver disease, and graft function is
The specific regimen chosen is likely to be a moving tar-
unknown. As mentioned previously, studies have clearly
get with rapid advances in HCV therapies. Up-to-date infor-
demonstrated tacrolimus as an additive risk in HCV patients
mation can be obtained from the HCV treatment guidelines
for the development of PTDM.170 In addition, as mentioned
published by the American Association for the Study of
previously, in liver transplant recipients cyclosporine may
Liver Diseases (AASLD) accessible at https://www.hcvguid
have an anti-HCV effect and improve the probability of suc-
elines.org/.182
cessful HCV treatment.171,172 However, there are no data
regarding cyclosporine effects on HCV in kidney transplant Treatment of HCV in the Renal Transplant
recipients. Similarly, although corticosteroid boluses have Candidate and Recipient
been shown to increase HCV viral load dramatically and
decrease time to HCV recurrence in liver transplant recipi- The optimal timing of HCV treatment with new DAAs is yet
ents,173,174 there are no data in kidney transplant recipi- to be determined (see Table 32.5). Effective HCV therapy
ents. Finally, although poor outcomes have been reported is available for patients on dialysis and if cure is achieved,
with antibody induction in HCV-positive liver transplant relapse after renal transplant is unlikely. However, clearing
recipients, there are no data on kidney transplant recipients. HCV therapy on dialysis may deprive HCV-positive patients
In the absence of data, few recommendations can be of the chance of receiving an organ from a HCV-positive
made regarding immunosuppression strategy in HCV- donor with a shorter waiting time.
infected kidney transplant recipients. Given the permissive Posttransplant treatment of HCV with DAAs can work
effects of immunosuppression on HCV replication, a reason- rapidly, effectively, and safely and may be a better option
able goal is to provide the minimum dose of immunosup- rather than treating patients on the waiting list unless
pression to prevent rejection. there is concern about worsening liver disease on the
waiting list, usually in patients with advanced fibrosis or
early cirrhosis. The exact timing of posttransplant treat-
Hepatitis C Antiviral Therapy ment is not established but earlier treatment is preferred,
Eradication of HCV has several benefits (Table 32.5). HCV because it can prevent HCV-related worsening of kidney
is associated with worse patient and graft survival and function, liver disease, or other extrahepatic effects of
increased risk of PTDM and de novo glomerulopathy. Eradi- hepatitis C.
cation of HCV pretransplant might mitigate some of these
adverse outcomes.175–177 Treatment Before Renal Transplantation. Concerns
Posttransplant HCV treatment in kidney transplant with pretransplant treatment on dialysis include decreased
recipients was historically limited because of the use of options in patients with CKD. However recently therapies
interferon in all regimens until 2011. Interferon-based that are effective in ESRD have been approved.
therapy was long (48 weeks), poorly tolerated, with mul- Grazoprevir/elbasvir has a cure rate (SVR) of 94% in
tiple side effects including severe constitutional symptoms, stage 4 to 5 CKD (including dialysis) for patients with geno-
pancytopenia, and depression, and it had a poor response type 1 or 4 HCV infection.183 In another study the combina-
rate. Most importantly it carried the risk of precipitating tion of glecaprevir/pibrentasvir had a SVR rate of 98% in
renal transplant rejection and therefore was rarely used patients with CKD and ESRD.184
after renal transplantation.178,179 Sofosbuvir-based regimens are effective and safe in
The standard of care for HCV treatment has evolved over patients even with decompensated cirrhosis. Although
the years from standard interferon monotherapy, to stan- sofosbuvir is not approved for use if GFR is less than 30,
dard interferon plus the antiviral ribavirin, to pegylated there are numerous reports and cases of safety and toler-
interferon and ribavirin, to, most recently, the addition of ability when sofosbuvir is used in decompensated cirrhosis
direct-acting antiviral agents (DAAs). with GFR less than 30.185
The first DAAs (telaprevir and boceprevir) had similar
issues because they had to be used in interferon-contain- Treatment After Renal Transplantation. Hepatitis C
ing regimens and thus carried all the side effects and risks therapy with DAAs after renal transplant has been reported
of interferon.180 Since 2014, however, DAAs are avail- predominantly in real-world experiences from single-center
able that are used without interferon. The new generation and multicenter studies and in small clinical trials. The
of DAA treatments work directly on HCV replication and results demonstrate excellent SVR rates in excess of 95%
have revolutionized HCV therapy. These agents are potent, and therapy that is well tolerated.
highly effective in curing HCV infection, and well tolerated. Hepatitis C DAA treatment in kidney transplant recipi-
They inhibit the HCV NS5B polymerase, the HCV NS5A ents was analyzed in a real-world experience in 119 patients
protein, and the HCV NS3/4 protease, which are proteins from 15 hospitals in Spain. HCV was caused by genotype 1b
integral for viral replication. These antiviral agents are in 66.5%, genotype 1a in 13.4%, genotype 2 in 4.2%, and
available in all oral therapies and have virologic response genotype 3 in 7.5%. DAA treatment included a polymerase
TABLE 32.5 Selected Pretransplant and Posttransplant (Nonliver) Studies of Antiviral Therapy in HCV Patients
Duration of
Study Patient Population N Antiviral Therapy Therapy SVR Side Effects/ Discontinuation Outcome after Transplant
PRETRANSPLANT
Roth D, et al., The HCV genotype 1 with 235 Grazoprevir/elbasvir immediate 12 weeks 99% The frequencies of adverse events were
Lancet 2015270 stage 4–5 CKD includ- and deferred treatment arms comparable between the immediate
ing 76% on dialysis treatment and deferred treatment
groups (76% vs. 84%), most adverse
events were of mild or moderate
intensity in both treatment groups. The
most common adverse events (≥10%
frequency) were headache, nausea, and
fatigue and were comparable in the two
groups.
Gane E et al., NEJM HCV genotype 1–5 with 104 Glecaprevir/pibrentasvir 12 weeks 98% Adverse events that were reported in at HAI at 1 year after transplant
2017184 CKD and least 10% of the patients were pruritus, lower in group A (1.19 )
ESRD (85% on hemo- fatigue, and nausea. Serious adverse than group B (5.5).
dialysis) events were reported in 24% of the
patients. Four patients discontinued the
trial treatment prematurely because of
adverse events.
Pockros et al., Gastro- HCV genotype 1 20 Ombitasvir coformulated with 12 weeks 90% Four patients experienced serious adverse
enterology 2016271 patients with CKD paritaprevir and ritonavir, events; all were considered unrelated to
stage 4 and 5 administered with dasabuvir+ treatment. Ribavirin therapy was inter-
ribavirin for genotype 1a (13) rupted in 9 patients because of anemia
Desnoyer et al., J HCV genotype 12 Sofosbuvir, 400 mg once daily 12–24 weeks 83% Nine grade 1 adverse events possibly
Hepatol 2016272 1 (n = 11) or 2 (1) (n = 7) or 3 times a week (n = 5), related to treatment were reported in
after hemodialysis with either 7 patients (anemia, headache). One
simeprevir, daclatasvir, ledipasvir, grade-2 event of asthenia was reported.
or ribavirin One grade-3 event (Streptococcus sp.
sepsis) was reported and considered

32 • Liver Disease Among Renal Transplant Recipients


unrelated to treatment with SOF TIW
and DCV
POSTTRANSPLANT
Gentil et al., Transp Kidney transplant recipi- 119 91% had sofosbuvir-based therapy, 12–24 weeks 97.8% Side effects tolerable, 3 patients Renal function did not
Proc. 2016186 ents from 15 Spanish sofosbuvir with ledipasvir, or discontinued treatment because of change.
hospitals simeprevir or daclatasvir with or anemia, 1 because of tacrolimus
without ribavirin. neurotoxicity.
PrOD in 9%
Fernandez et al., Kidney transplant recipi- 101 57% had sofosbuvir with ledipasvir, 12–24 weeks 98% Side effects tolerable, grade 2–3 anemia Renal function did not
J Hepatol 2017187 ents from Spanish 17% sofosbuvir with daclatasvir. in 33% receiving ribavirin and in 15% change. Patients with
registry ribavirin adjuvant treatment in 41% without ribavirin. cirrhosis had higher
incidence of renal dys-
function; 55% required
immunosuppression
adjustment.

Continued

553
554
TABLE 32.5 Selected Pretransplant and Posttransplant (Nonliver) Studies of Antiviral Therapy in HCV Patients—cont’d
Duration of
Study Patient Population N Antiviral Therapy Therapy SVR Side Effects/ Discontinuation Outcome after Transplant

Kidney Transplantation: Principles and Practice


Lin MV et al., PLoS ONE Renal transplant 24 Sofosbuvir-based therapy in all. 12 weeks 91% 11 patients with adverse events, 3 serious. Renal function did not
2016188 recipients with HCV Combined with either simeprevir, 1 was related- cardiac arrhythmia change.
genotype 1 in 21/24 ledipasvir, paired with ribavirin, or (sofosbuvir + simeprevir)
and genotype 2 in ribavirin alone
3/24
Eisenberger et al., Renal transplant 15 Sofosbuvir and ledipasvir 8–12 weeks 100% Mild fatigue, nausea, headache, and No change in renal allograft
Transplantation recipients with HCV ­hypertension in 5/15 function; liver tests
2017189 genotype 1 or 4 improved; 55% required
increase in tacrolimus
dosing.
Kamar et al., American Renal transplant 25 Sofosbuvir plus ribavirin (n = 3); 12–24 weeks 100% 1 patient with anemia, otherwise well No change in renal graft
J Transplant, recipients with HCV sofosbuvir plus daclatasvir (n = 4); tolerated therapy function and no change in
2016190 genotype 1–4 sofosbuvir plus simeprevir, with immunosuppressive levels.
(n = 1) or without ribavirin (n = 6);
sofosbuvir plus ledipasvir, with
(n = 1) or without ribavirin (n =
9); and sofosbuvir plus pegylated-
interferon plus ribavirin (n = 1)
Sawinski et al., Am J Renal transplant 20 Sofosbuvir-based therapies with ledi- 12 weeks Anemia in 2 patients, one r­ equired Renal graft function remained
Transplant, 2016161 recipients with HCV pasvir, daclatasvir, simeprevir with ­transfusion rejection stable. Immunosuppression
genotype 1 (85%) and or without ribavirin dose adjusted in 45%.
genotype 2

CKD, chronic kidney disease; ESRD, end-stage renal disease; HAI, hepatic activity index; HCV, hepatitis C virus; PrOD, paritaprevir/ritonavir/ombitasvir plus dasabuvir; SVR, sustained virologic response, defined as
undetectable HCV RNA 12 weeks after the end of therapy.
32 • Liver Disease Among Renal Transplant Recipients 555

inhibitor in 108 (91%) patients, often in combination with In a real-world experience, 25 kidney transplant patients
the NS5a inhibitor ledipasvir in 65 cases, the NS3/4 pro- were treated with DAAs. Ten had advanced liver fibrosis (F3
tease inhibitor simeprevir in 17 cases, the NS5a inhibitor and F4 Metavir score) and the remaining 15 patients had
daclatasvir in 16 cases. In 20 patients ribavirin was used mild liver fibrosis (F1/F2 Metavir score) and either HCV-
as a coadjuvant therapy. Nine cases were treated with associated extrahepatic manifestations or a history of graft
3-D therapy (ombitasvir-dasabuvir-paritaprevir-ritonavir loss caused by HCV-associated glomerulonephritis. Most
combination). patients (20 of 25) were infected with HCV genotype 1.
SVR was seen in 97.8% of cases. Side effects were gen- Patients were given sofosbuvir plus ribavirin (n = 3);
erally limited and not attributable to DAA except in seven sofosbuvir plus daclatasvir (n = 4); sofosbuvir plus simepre-
cases where treatment had to be interrupted because of vir, with (n = 1) or without ribavirin (n = 6); sofosbuvir plus
anemia (three patients), neurotoxicity because of tacroli- ledipasvir, with (n = 1) or without ribavirin (n = 9); and
mus interaction with 3-D (one patient), and other (three sofosbuvir plus pegylated-interferon plus ribavirin (n = 1).
patients). Renal function and proteinuria did not change.186 Antiviral therapy was given for 12 (n = 19) or 24 weeks (n =
In the Hepa-C Spanish Registry, data were reported on 6). SVR at 12 weeks was 100%. Eight patients had impaired
103 HCV-positive kidney transplant recipients treated kidney function, but there were no acute rejection episodes
with DAAs. These patients were treated at a median and no graft losses. However, tacrolimus trough levels were
interval of 147 months after kidney transplant (range significantly decreased after HCV clearance.190
1–561 months). The majority (83%) were genotype 1, Sawinski et al. reported on 20 HCV-positive patients with
and 81% had previously failed interferon therapy. Cir- a kidney transplant and treated with a DAA-based ther-
rhosis was present in 35%. The most commonly used apy, without interferon alpha. Three of these patients also
DAA combinations were sofosbuvir/ledipasvir (n = 59, received ribavirin. Of the 20 patients, 88% were infected by
57%) and sofosbuvir/daclatasvir (n = 18, 17%). Ribavi- genotype 1 and 50% had biopsy-proven advanced hepatic
rin was used in 41% of patients. The SVR rate after 12 fibrosis. DAA therapy was initiated at a median of 888 days
weeks (SVR12) was 98%. Grade 2 or 3 anemia appeared after kidney transplantation. All 20 patients achieved SVR
in 14 (33%) of patients receiving ribavirin and in 9 (15%) at 12 weeks after completing therapy.161
without ribavirin (P = 0.03). There were three episodes In a randomized, phase 2, open-label study treatment-
of acute humoral graft rejection, no patient discontinued naïve or -experienced kidney transplant recipients with
therapy because of adverse events, and 57 (55%) patients chronic genotype 1 or 4 HCV infection with an eGFR of 40
required immunosuppression dose adjustment. Overall, mL/min or greater were randomly assigned 1:1 to receive
there were no significant differences in the mean level ledipasvir (90 mg) and sofosbuvir (400 mg) for 12 or 24
of serum creatinine, estimated GFR (eGFR), and protein- weeks. Of all participants, 91% had genotype 1 infection
uria before and after treatment. Nonetheless, 17 (16%) and 15% had compensated cirrhosis. One hundred per-
patients experienced renal dysfunction (increase in serum cent of patients (57 of 57) treated for 12 weeks (95% CI,
creatinine >25%) during antiviral therapy, of whom 65% 94%–100%) and 100% of those (57 of 57) treated for 24
were cirrhotic.187 weeks (95% CI, 94%–100%) achieved SVR. Serious adverse
A retrospective chart review with prospective clinical fol- events were reported in 13 patients (11%). Of these, three
low-up of postkidney transplant patients treated with DAA events—syncope, pulmonary embolism, and serum cre-
therapies in three city hospitals was reported. Twenty-four atinine increase—in three patients were determined to be
kidney recipients with HCV infection received all-oral DAA treatment related. Treatment with ledipasvir/sofosbuvir for
therapy posttransplant. The majority had HCV genotype 1a 12 or 24 weeks was well tolerated and seemed to have an
infection (58%). All patients received full-dose sofosbuvir; acceptable safety profile among kidney transplant recipients
it was paired with simeprevir (nine patients without and with HCV genotype 1 or 4 infection, all of whom achieved
three patients with ribavirin), ledipasvir (seven patients SVR12.191
without and one patient with ribavirin), or ribavirin alone Although some of these DAAs do not interact with cal-
(four patients). The overall SVR12 was 91% (21 out of 23 cineurin inhibitors or mTOR inhibitors, some such as
patients). Two treatment failures were successfully retreated ledipasvir, simeprevir, or ombitasvir can interact with
with alternative DAA regimens and achieved SVR. Both ini- immunosuppressive agents, most commonly through their
tial failures occurred in patients with advanced fibrosis or effect on cytochrome P-450. Also, as liver function improves
cirrhosis, with genotype 1a infection, and prior HCV treat- with use of any effective DAA, hepatic metabolism of immu-
ment failure.188 nosuppressive agents may improve, reducing trough levels
Fifteen renal allograft recipients with therapy-naïve HCV of antirejection agents. Vigilance of immunosuppression
genotype 1a, 1b, or 4 were treated with the combination levels and renal function is required in monitoring.
of sofosbuvir and ledipasvir without ribavirin for 8 or 12
weeks. Clinical data were retrospectively analyzed for viral HEPATITIS E
kinetics and for renal and liver function parameters. One
hundred percent of patients exhibited SVR at week 12 after Hepatitis E virus (HEV) is a nonenveloped, single-strand
the end of treatment. Clinical measures of liver function RNA virus of the family Hepaviridae.192 HEV is considered
improved substantially for all patients. Adverse events were endemic in developing countries and is spread via fecal-oral
scarce and the drugs well tolerated; renal transplant func- transmission, similarly to hepatitis A.193 In immunocom-
tion and proteinuria remained stable. Importantly, dose petent patients, HEV has a clinical course similar to HAV,
adjustments for tacrolimus were necessary for maintaining manifested by an acute, sometimes icteric, self-limited ill-
sufficient trough levels.189 ness without the potential for chronicity.
556 Kidney Transplantation: Principles and Practice

However, in recent years, zoonotic transmission (fre- fibrosis. These effects remained after adjustment for diabe-
quently from a swine vector) of HEV in industrialized tes status.200
countries has become increasingly recognized. In France, NAFLD itself may add to this renal dysfunction, possibly
anti-HEV antibodies are present in 16.6% of blood donors via associated chronic inflammation and oxidative stress.200
and 6% to 16% of renal transplant recipients.194 In addi- The role of inflammatory mediators, such as tumor necrosis
tion, chronic hepatitis resulting from HEV infection has factor (TNF) alpha, in NASH is of particular interest in the
been reported in immunocompromised patients, including pathophysiology of CKD.198,201
liver and kidney transplant recipients.195 Thus chronic HEV Not only is the risk of CKD in patients with NAFLD
infection should be considered in kidney transplant recipi- increased, the incidence of simultaneous liver-kidney trans-
ents with unexplained liver test abnormalities. plant (SLK) is increased in NAFLD compared with other
There is no effective vaccination available for HEV. Cur- causes of cirrhosis. In a UNOS database query of 38,533
rent recommendations for HEV prevention focus primarily liver transplants between 2002 to 2011 in the US, 5.6%
on proper hygiene and consumption of properly cooked received SLK with the proportions of SLKs being 8.7% in
food. Bloodborne transmission of HEV is felt to be rare. NAFLD; 5% in HCV, HBV, or liver cancer; and 6.2% in
There are no data on the natural history of HEV infec- alcohol-induced or cholestatic cirrhosis. Furthermore, the
tion on kidney transplant recipients. Similarly, there are incidence of SLK performed for NAFLD increased from 6.3%
very limited data on treatment for chronic HEV. Pegylated in 2002 to 2003 to 19.2% in 2010 to 2011.202
interferon-α has been used in liver transplant recipients,
but caution should be used in renal transplant recipients NAFLD POST RENAL TRANSPLANT
because of the known risk of allograft loss. Ribavirin has
also been used in kidney transplant recipients with chronic Given the high prevalence and incidence of metabolic syn-
HEV infection. Kamar et al.196 published a series of six drome in renal transplant recipients, NAFLD is expected
kidney transplant recipients with chronic HEV hepatitis to be commensurately frequent. The diagnosis of NAFLD
treated with ribavirin at a dose of 600 to 800 mg/day. All is traditionally made with liver biopsy, which is inva-
patients achieved serum viral clearance at 3 months. Four sive and not acceptable to many patients. Imaging tech-
patients achieved a durable response after cessation of riba- niques such as ultrasound, computed tomography (CT),
virin. Larger studies are needed to determine optimal dose or magnetic resonance imaging (MRI) have also been
and duration of ribavirin therapy for chronic HEV. used to make diagnosis of NAFLD after serologic exclusion
of other chronic liver conditions but suffer from reduced
sensitivity and specificity (ultrasound), and cost and lim-
Nonalcoholic Fatty Liver Disease ited availability (MRI). Consequently, few studies have
and Renal Transplant systematically investigated the frequency of NAFLD post–
renal transplant.
NAFLD is the most prevalent chronic liver disease in the Using a new technology (transient elastography, or TE)
US, affecting 30% of the general adult population and that utilizes noninvasive shear wave technology and liver
up to 60% to 70% of diabetic and obese patients. NAFLD attenuation (controlled attenuation parameter, or CAP)
encompasses a histologic spectrum ranging from simple to diagnose liver steatosis and stiffness, Mikolasevic et al.
steatosis to the more progressive form, nonalcoholic ste- investigated the prevalence of NAFLD in renal transplant
atohepatitis (NASH), which is often associated with fibro- recipients. When including presence of any steatosis inde-
sis and can lead to cirrhosis. NAFLD is intimately linked pendent of fibrosis as inclusion of NAFLD, a prevalence of
to metabolic syndrome, in particular the closely linked 57.5% was described in 73 renal transplant recipients fol-
epidemic of obesity, type 2 diabetes, hyperlipidemia, and lowed for more than 1-year post transplant. In the study,
hypertension.197 the severity of hepatic steatosis was independently associ-
Metabolic syndrome and its components are associated ated with increasing serum creatinine levels after transplant
with increased risk of CKD. Metabolic syndrome has been (correlation coefficient r = 0.66, P < 0.001). The liver fibro-
associated with azotemia, proteinuria, and glomerular sis severity in NAFLD also correlated with worsening serum
injury. Given the close association of NAFLD with meta- creatinine. Most importantly liver enzymes did not differ
bolic syndrome, it is not surprising that NAFLD is associated between those with and without NAFLD in the renal trans-
with CKD.198,199 plant population, suggesting that elevated liver enzymes are
However, there is significant evidence that NAFLD not an accurate way to detect NAFLD in this population.
increases the incidence of CKD, in excess of that expected Longer prospective studies in larger cohorts of renal
because of the associated metabolic risk factors such as dia- transplant recipients with biopsy-proven NAFLD are
betes and hypertension. In a meta-analysis of 20 studies, needed to confirm these findings and to determine whether
NAFLD was associated with an increased risk of prevalent improvement in NAFLD will prevent or delay the develop-
(odds ratio [OR] 2.12, 95% CI 1.69–2.66) and incident (HR ment and progression of CKD.203
1.79, 95% CI 1.65–1.95) CKD. The more progressive form
of NAFLD, NASH, was associated with a higher prevalence NAFLD AND OUTCOMES IN RENAL TRANSPLANT
(OR 2.53, 95% CI 1.58–4.05) and incidence (HR 2.12, 95%
RECIPIENTS
CI 1.42–3.17) of CKD than simple steatosis.
Advanced liver fibrosis was also associated with a higher Although 5-year patient and liver graft survival were simi-
prevalence (OR 5.20, 95% CI 3.14–8.61) and incidence lar in NAFLD and non-NAFLD SLK patients, in an audit of
(HR 3.29, 95% CI 2.30–4.71) of CKD than nonadvanced UNOS data, the 5-year kidney graft outcome was worse for
32 • Liver Disease Among Renal Transplant Recipients 557

NAFLD versus other causes (70% vs. 79%, P = 0.002) after and pioglitazone in nondiabetic adults with biopsy-proven
controlling for recipient characteristics.202 NASH.214
Cardiovascular diseases (CVDs) are the predominant Other treatments related to disordered cholesterol metab-
cause of mortality in renal transplant recipients and are olism and insulin resistance including statins, fibrates, met-
closely associated with the metabolic syndrome. Obesity, formin, and glucagon-like peptide (GLP-1) analogs have
hypertension, diabetes, and dyslipidemia are significant shown improvement in liver enzymes and weight loss with-
concerns post–renal transplant in 20% to 70% of recipi- out changes in histologic staging of the disease. Although
ents. These same risk factors are also closely associated with statins have not shown any benefit on NAFLD-related liver
NAFLD. In the general population the predominant cause disease there has been reduction in CVD-related events in
of mortality in patients with NAFLD is CVD.204 patients with NAFLD without any significant adverse effects
Whether NAFLD is independently associated with CVD on liver function.212
in renal transplant recipients was investigated in a large Guidelines recommend ACE inhibitors and angioten-
single-center study in the Netherlands. Of 602 renal trans- sin receptor blockers (ARBs) as preferred agents for dia-
plant recipients over a 2-year period, 388 had metabolic betic kidney disease and nondiabetic kidney diseases with
syndrome. NAFLD was presumed based on indirect mark- proteinuria. In these diseases, they lower blood pressure,
ers, primarily elevated liver enzymes, and was associated reduce proteinuria, slow the progression of kidney disease,
with the presence of metabolic syndrome. In regression and likely reduce CVD risk by mechanisms in addition to
analyses, both elevated gamma-glutamyltransferase (GGT) lowering blood pressure. In these types of CKD, ACE inhibi-
(HR 1.43(.21–1.69, P < 0.001) and alkaline phosphatase tors and ARBs are recommended even in the absence of
(HR = 1.34 (1.11–1.63), P = 0.003) were independently hypertension.145
associated with mortality. In patients with NAFLD who have CKD, renin-angioten-
Limitations of this study included that the definition of sin blockade using ACE inhibitors and ARBs have demon-
NAFLD as elevated liver enzymes is not necessarily sensi- strated benefits in liver and kidney disease. Limited data
tive or specific for NAFLD in the absence of either imaging from 223 patients in three randomized controlled trials
or liver biopsy to confirm the diagnosis.205 in NAFLD suggests that ARBs attenuate steatosis, insu-
In another single-center study, carotid atherosclerosis lin resistance, and inflammatory markers independent of
was assessed in 71 renal transplant recipients who had reduction in blood pressure.215
NAFLD diagnosed by TE. These individuals with NAFLD Telmisartan, which is an ARB with peroxisome prolifera-
showed more carotid atherosclerosis than renal transplant tor activated receptor gamma regulating activity, was com-
recipients without NAFLD.206 pared with the use of valsartan in the Fatty Liver Protection
Potential mechanisms whereby NAFLD is associated by Telmisartan (FANTASY Trial) and found to cause reduc-
with CVD and mortality include increased oxidative stress tion in necroinflammation, NAFLD activity score, fibrosis
and endothelial dysfunction seen in patients with NAFLD. stage in NASH, and microalbuminuria.216
Rajman et al. reported the presence of small low-density
lipoprotein (LDL) particles in uremic dyslipidemia, which
is persistent after renal transplantation. These may play a
Hepatocellular Carcinoma After
pathogenic role in NAFLD development in renal transplant Renal Transplantation
recipients, contribute to further insulin resistance, and pro-
mote atherosclerosis. Concentrations of plasminogen acti- In the setting of immunosuppression loss of tumor surveil-
vator inhibitor-1 (PAI-1) are also elevated in patients with lance can lead to higher risk for various malignancies. HCC is
NAFLD, which is another risk factor for CVD.207–211 more common after renal transplantation (incidence 1.4%–
4%) than in the general population (incidence 0.005%–
0.015%).217–220 This risk is particularly true for renal
TREATMENT OF NAFLD transplant recipients infected with chronic HBV or HCV.
The mainstay of treatment of NASH is weight loss through In areas endemic for HBV, HCC is the most common tumor
a combination of exercise and diet. Treatment of the under- after renal transplantation (20%–45%).221,222 Recently,
lying metabolic diseases including diabetes, hypertension, Hoffman et al.223 published data on the development of de
and hyperlipidemia is integral in preventing exacerbation novo HCC in transplant recipients. Using US registry data of
of the disease.212 more than 200,000 transplant recipients, the incidence of
Medical therapies for NAFLD include agents that can HCC in nonliver transplant recipients was 6.5 per 100,000
improve glycemic control, lipid metabolism, or those with person-years. HBV and HCV infection were independently
antioxidant effects. Use of thiazolidinediones (TZDs) such predictive of the development of de novo HCC. Estimated
as pioglitazone, which improve insulin resistance, have survival was worse than that expected for similar-stage
shown some benefit in randomized controlled trials and tumors in nontransplanted populations.219,222 Because
are associated with histologic improvements, and they can outcomes after HCC are poor, preventive measures are
be used in the treatment of NAFLD. Vitamin E, which is an important, including: vaccination of renal transplant wait-
antioxidant, has shown histologic benefits in randomized ing list patients for HBV, antiviral therapy for HCV and HBV
controlled trials of NAFLD as well.213 Although many of in the dialysis population, continued antiviral treatment for
these studies have a small cohort of patients and the his- HBV in the renal transplant recipient, exclusion of patients
tologic endpoints were not standardized, the AASLD has with ESRD and cirrhosis from isolated kidney transplanta-
published guidelines recommending the use of vitamin E tion and in select cases consideration of these patients for
combined liver transplantation.
558 Kidney Transplantation: Principles and Practice

Renal transplant recipients infected with HBV/HCV and is high.232 There is a marked male predominance and ame-
uncontrolled viral replication or with advanced fibrosis/ bic liver abscesses are usually solitary.233 Clinical signs and
cirrhosis should be entered in an HCC surveillance proto- symptoms and liver test abnormalities do not help distinguish
col. Current AASLD guidelines recommend surveillance amebic from pyogenic liver abscesses. Serology for antibod-
with ultrasound with or without alpha-fetoprotein every 6 ies to Entamoeba histolytica is useful to determine current or
months.224 past infection. After confirmation of an abscess on imaging,
if amebic rather than pyogenic liver abscess is suspected,
treatment with metronidazole for 10 days is necessary.
Systemic Infections Resulting in Renal transplant recipients traveling to areas endemic for
Hepatitis and Liver Disease amebiasis should be counseled to avoid ingestion of poten-
tially contaminated food and water, such as fresh produce
A number of systemic infections have hepatitis as part of the that cannot be adequately cooked.234 Boiling water before
clinical manifestation. Foremost among these are infections use is essential to destroy the cysts of E. histolytica, which are
caused by herpesviruses, which are major pathogens in not killed by low-dose iodine or chlorine tablets.
organ transplantation. Other infections primarily involving
the liver are also reviewed.
MYCOBACTERIAL INFECTION
LIVER ABSCESS Tuberculosis is an important cause of morbidity and mor-
tality among renal transplant recipients. The risk of active
Pyogenic liver abscess does not represent a specific liver dis- tuberculosis is at least 50-fold higher in renal transplant
ease but is a final common pathway of many pathologic pro- recipients compared with nontransplant patients; most
cesses. The incidence of pyogenic liver abscess ranges from reactivation disease has been reported to occur in the first
8 to 20 cases per 100,000 hospital admissions225; a popula- year after transplantation.235,236 Liver involvement with
tion-based study reported 2.3 cases per 100,000 persons per tuberculosis remains rare; when present, it is usually
year.226 A population-based study found no increased risk of associated with pulmonary disease237 or gastrointestinal
pyogenic liver abscess in renal transplant recipients.226 involvement. Three patterns of tuberculous liver involve-
Abscesses may be classified by presumed route of hepatic ment have been reported238: (1) diffuse involvement of the
invasion: (1) biliary tree, (2) portal vein, (3) hepatic artery, liver in association with tuberculosis at other body sites; (2)
(4) direct extension from contiguous focus of infection, and miliary involvement of the liver with no other known organ
(5) penetrating trauma.225 Approximately 50% of pyogenic involvement (granulomatous hepatitis); and (3) focal lesion
liver abscesses are cryptogenic.227 The microbiology of pyo- in the liver, either an abscess or a tuberculoma.239 Consti-
genic liver abscess is variable and depends on the route of tutional symptoms and fever are common but nonspecific.
infection. Most infections are polymicrobial, with enteric A modest degree of transaminase and alkaline phosphatase
facultative and anaerobic bacteria being the most common elevation is common. Imaging followed by tissue stain-
agents. Candida species should also be suspected as a patho- ing for acid-fast bacilli and culture for mycobacteria are
gen in pyogenic abscesses, accounting for 22% of abscesses required to confirm the diagnosis.
in one series.228
Although fever and constitutional symptoms are frequent, VIRAL INFECTIONS
only 1 in 10 patients presents with the classic triad of fever,
jaundice, and right upper quadrant tenderness. Although Herpesviruses
liver function tests are abnormal in most patients, the eleva- The herpesviruses include cytomegalovirus (CMV), Epstein–
tion is usually modest. Radiographic imaging using MRI, Barr virus (EBV), herpes simplex virus (HSV), human her-
CT, or ultrasonography is essential to making the diagnosis. pesvirus 6 and 7, and varicella-zoster virus (VZV). The
Microbiologic diagnosis rests on obtaining purulent mate- herpesvirus family is responsible for considerable morbidity
rial from the abscess cavity, which should be sent for Gram and mortality in transplant recipients. In particular, CMV
stain and culture. In general, treatment consists of antimi- remains a major health threat after solid-organ transplan-
crobial therapy for 3 to 4 weeks and drainage of the abscess. tation. All the herpesviruses have the ability to remain
Some investigators have reported success with treatment latent in tissues after acute infection. Liver involvement fre-
of small abscesses with antibiotic therapy alone; however, quently is a part of the clinical presentation of herpesvirus-
most patients will require some form of abscess drainage.229 related diseases.
Drainage may be achieved by percutaneous aspiration with
or without placement of a drainage catheter. Generally, Cytomegalovirus
abscesses larger than 5 cm require placement of a catheter.230 CMV is one of the most important pathogens in transplant
Endoscopic drainage via endoscopic retrograde chol- recipients.234,240 Unlike the other herpesviruses, such as
angiopancreatography has been reported to be successful HSV and VZV, which remain latent in highly restricted
in cases where the abscess communicates with the biliary areas of the body, once acquired, latent CMV can be found
tree.231 Finally, surgical drainage may be needed in cases in multiple body sites.
with multifocal abscesses, heavily loculated abscesses, or After transplantation, approximately 50% of transplant
unsuccessful percutaneous drainage. patients excrete CMV in body secretions (e.g., saliva and
Amebiasis is a far less common cause of liver abscess in urine) at some point.240 In addition, over 60% of patients
the US but one that must be considered in patients living in develop antigenemia within the first 100 days after
or traveling to countries where the prevalence of amebiasis transplantation.241
32 • Liver Disease Among Renal Transplant Recipients 559

Hepatitis is a major clinical manifestation of CMV dis- duration of symptoms.251 The virus establishes latency in
ease. In the immunocompetent patient, the disease is usu- ganglia and may reactivate. Immunocompromised patients
ally mild and self-limiting, although rare cases of fulminant have been found to have more severe primary infections
CMV hepatitis have been described.242 In the transplant and more frequent reactivations.252 In renal transplant
recipient, CMV hepatitis is a more severe illness, usually recipients, the incidence of HSV infection has been reported
with other organ involvement or disseminated disease, and to be 30% to 50% in the absence of prophylaxis.253,254 The
is not uncommon. In a series of 97 renal transplant recipi- risk of HSV reactivation is highest in the first 3 months
ents with CMV disease, half had evidence of CMV hepatitis; posttransplant because of the higher level of immunosup-
the severity of hepatitis was greater in primary disease than pression needed during this period. Oral acyclovir for pro-
in cases of reactivation.241 phylaxis against HSV has greatly reduced the incidence of
In an autopsy series of four immunocompromised HSV infection.253,254
patients with overwhelming CMV infection, Ten Napel et al. Hepatitis with HSV has been well described in the gen-
showed that liver cell damage was extensive but inflamma- eral population and the renal transplant population. Kusne
tory infiltration was less prominent than in immunocom- et al.255 reported a series of 12 cases of HSV hepatitis that
petent patients with CMV infection.243 Intracellular CMV developed a median of 18 days after solid-organ transplan-
inclusion bodies were found in the hepatocytes, vascular tation. The clinical features included fever, herpetic sto-
endothelium, and bile epithelium. Given the significant matitis, and abdominal pain, usually in association with
morbidity and mortality of CMV infection, including CMV disseminated disease. Clinical features associated with
hepatitis, in solid-organ transplant recipients, preven- mortality included bacteremia, hypotension, disseminated
tion and treatment of CMV infection posttransplant are of intravascular coagulation, and gastrointestinal bleeding.
utmost importance. The diagnosis and treatment of CMV HSV hepatitis was associated with 67% mortality in this
infection and strategies for CMV prophylaxis in kidney patient population.
transplant recipients are covered in another chapter. Conclusive diagnosis of HSV hepatitis rests on demon-
stration of viral involvement of liver tissue. Histologically,
Epstein–Barr Virus hepatocytes have enlarged “ground-glass” nuclei with
EBV, a member of the human gammaherpesvirus family, is chromatin margination. Because of the high mortality
a ubiquitous pathogen. More than 90% of the world’s pop- associated with HSV hepatitis, transplant recipients who
ulation is infected.244 The virus is shed intermittently into present with fever, progressive transaminase elevation, and
saliva245 and is believed to be transmitted through close abdominal symptoms with or without evidence of cutane-
contact with oral secretions. EBV infection may present as ous herpes simplex infection should prompt consideration
primary or secondary infection (reactivation). Childhood of HSV hepatitis and treatment with intravenous acyclovir.
disease is usually asymptomatic. Infection acquired in ado-
lescence or young adulthood frequently causes the clinical Varicella-Zoster Virus
syndrome of acute infectious mononucleosis, character- VZV is another herpesvirus that causes two distinct dis-
ized by fever, pharyngitis, and lymphadenopathy in 75% eases—varicella and herpes zoster. Primary infection with
of patients.246 A nonspecific hepatitis is common in acute VZV causes varicella in susceptible hosts. Children generally
infectious mononucleosis. Jaundice is apparent in 5% to develop mild disease compared with adults or immunosup-
9% of patients. Liver function test abnormalities peak with pressed patients. Whereas only 0.1% of varicella infections
acute illness and return to normal over 1 to 2 months. In develop in this population, 25% of varicella-related deaths
instances where liver biopsies have been obtained, mini- occur in this patient population.256
mal swelling and vacuolization of hepatocytes can be seen Hepatic involvement with varicella is uncommon but
accompanied by a lymphocytic or monocytic infiltrate in has been described in transplant recipients. A study assess-
portal regions.247 ing clinical features of liver transplant patients with vari-
EBV establishes latency248 and may reactivate later; the cella hepatitis showed that the most common presenting
risk of reactivation is especially high in immunosuppressed features were cutaneous vesicular lesions, fever, and acute
patients. Primary infection with EBV after transplantation abdominal or back pain. The rash may not be apparent at
may manifest as a febrile illness with constitutional signs the time of hepatic involvement, however, and the diag-
and symptoms. nosis of varicella hepatitis may be delayed. In case reports,
EBV has a central role in the pathogenesis of posttrans- high-dose acyclovir (10 mg/kg every 8 hours) has been
plant lymphoproliferative disorder (PTLD),249 although not shown to treat varicella hepatitis successfully.
all PTLD is caused by EBV. Like all herpesviruses, VZV establishes latency and may
The diagnosis and treatment of EBV and PTLD are dis- subsequently reactivate.257 Reactivation of latent VZV typi-
cussed in Chapter 31. cally results in a localized skin infection known as herpes
zoster, or shingles.258
Herpes Simplex Virus Disseminated zoster in transplant patients can be a
HSV is an alpha herpesvirus with a genome consisting of a severe, prolonged illness with hepatitis as a prominent
linear, double-stranded DNA molecule.250 The two types of manifestation. In a case series of four renal transplant
HSV, HSV-1 and HSV-2, have 50% sequence homology. In recipients that developed primary (one patient) or reactiva-
the US population, seroprevalence rates range from 56% to tion VZV infection (three patients), all four had multiorgan
60% for HSV-1 and from 15% to 18% for HSV-2. First epi- involvement and three of the four developed hepatitis.258
sodes of HSV, or primary infection, are frequently accom- In general, primary varicella infection is a more severe ill-
panied by systemic signs and symptoms and have a longer ness than reactivated disease. Fehr et al. reviewed all cases
560 Kidney Transplantation: Principles and Practice

of herpes zoster in renal transplant recipients and found 11. Ward CJ, Hogan MC, Rossetti S, et al. The gene mutated in autoso-
34 reported cases, most of which were primary infections. mal recessive polycystic kidney disease encodes a large, receptor-like
protein. Nat Genet 2002;30(3):259–69.
Analysis of these cases showed that disseminated intravas- 12. Kamath BM, Piccoli DA. Heritable disorders of the bile ducts. Gastro-
cular coagulation and hepatitis occurred in half of the cases enterol Clin North Am 2003;32(3):857–75.
and pneumonitis in 29% of patients. The overall mortality 13. Yonem O, Bayraktar Y. Clinical characteristics of Caroli’s syndrome.
was 34%, although it appears to have decreased over time World J Gastroenterol 2007;13(13):1934–7.
14. Andrade RJ, Camargo R, Lucena MI, Gonzalez-Grande R. Causality
from 53% to 22%.258 assessment in drug-induced hepatotoxicity. Expert Opin Drug Saf
Treatment of disseminated zoster in transplant patients 2004;3(4):329–44.
should be undertaken promptly with high-dose acyclovir. 15. Chalasani N, Fontana RJ, Bonkovsky HL, et al. Causes, clinical fea-
tures, and outcomes from a prospective study of drug-induced liver
Human Herpesvirus 6 and 7 injury in the United States. Gastroenterology 2008;135(6):1924–34.
16. Kaplowitz N. Drug-induced liver injury. Clin Infect Dis 2004;38
Human herpesvirus 6 (HHV-6) and 7 (HHV-7) are ubiqui- (Suppl. 2):S44–8.
tous lymphotropic herpesviruses and were initially isolated 17. Maddrey WC. Drug-induced hepatotoxicity: 2005. J Clin Gastroen-
from patients with lymphoproliferative disorders.259 Serop- terol 2005;39(4 Suppl. 2):S83–9.
revalence surveys have found that HHV-6 infection occurs 18. Navarro VJ, Senior JR. Drug-related hepatotoxicity. N Engl J Med
2006;354(7):731–9.
in the majority of children by age 3 years, and the preva- 19. Watkins PB, Seeff LB. Drug-induced liver injury: summary of a single
lence in adults is greater than 90%. topic clinical research conference. Hepatology 2006;43(3):618–31.
The major childhood clinical syndrome caused by 20. Kelly BD, Heneghan MA, Bennani F, Connolly CE, O’Gorman TA.
HHV-6 primary infection is exanthem subitum. Infection Nitrofurantoin-induced hepatotoxicity mediated by CD8+ T cells.
in immune-competent adults is usually benign, present- Am J Gastroenterol 1998;93(5):819–21.
21. Chitturi S, George J. Hepatotoxicity of commonly used drugs: non-
ing as fever with lymphadenopathy or an infectious mono- steroidal anti-inflammatory drugs, antihypertensives, antidiabetic
nucleosis-like syndrome. HHV-6 infection posttransplant agents, anticonvulsants, lipid-lowering agents, psychotropic drugs.
has been frequently reported in kidney transplant recipi- Semin Liver Dis 2002;22(2):169–83.
ents.260,261 Typically, HHV-6 reactivation is asymptomatic, 22. Eisenbach C, Goeggelmann C, Flechtenmacher C, Stremmel W,
Encke J. Severe cholestatic hepatitis caused by azathioprine. Immu-
even in kidney transplant patients. However, symptomatic nopharmacol Immunotoxicol 2005;27(1):77–83.
and even fatal HHV-6 infections have been reported in the 23. Loupy A, Anglicheau D, Mamzer-Bruneel MF, et al. Mycopheno-
kidney transplant population. Hepatitis has been rarely late sodium-induced hepatotoxicity: first report. Transplantation
reported in both the immune-competent and transplant 2006;82(4):581.
populations.262–264 There are no controlled trials of antivi- 24. Neff GW, Ruiz P, Madariaga JR, et al. Sirolimus-associated hepatotoxic-
ity in liver transplantation. Ann Pharmacother 2004;38(10):1593–6.
ral therapy for HHV-6 or HHV-7 infection. According to the 25. Bohan TP, Helton E, McDonald I, et al. Effect of L-carnitine treatment for
American Society of Transplantation’s Infectious Disease valproate-induced hepatotoxicity. Neurology 2001;56(10):1405–9.
Community of Practice guidelines, IV ganciclovir or foscar- 26. Polson J, Lee WM, American Association for the Study of Liver Dis-
net is first-line therapy for active disease.265 ease. AASLD position paper: the management of acute liver failure.
Hepatology 2005;41(5):1179–97.
27. de Boer NK, Mulder CJ, van Bodegraven AA. Nodular regenerative
hyperplasia and thiopurines: the case for level-dependent toxicity.
Liver Transpl 2005;11(10):1300–1.
References 28. Degott C, Rueff B, Kreis H, Duboust A, Potet F, Benhamou JP. Peliosis
1. Ioannou GN, Boyko EJ, Lee SP. The prevalence and predictors of hepatis in recipients of renal transplants. Gut 1978;19(8):748–53.
elevated serum aminotransferase activity in the United States in 29. Malekzadeh MH, Grushkin CM, Wright Jr HT, Fine RN. Hepatic
1999–2002. Am J Gastroenterol 2006;101(1):76–82. dysfunction after renal transplantation in children. J Pediatr
2. Fraser A, Longnecker MP, Lawlor DA. Prevalence of elevated ala- 1972;81(2):279–85.
nine aminotransferase among US adolescents and associated factors: 30. Marubbio AT, Danielson B. Hepatic veno-occlusive disease in a
NHANES 1999–2004. Gastroenterology 2007;133(6):1814–20. renal transplant patient receiving azathioprine. Gastroenterology
3. Leon DA, McCambridge J. Liver cirrhosis mortality rates in Britain, 1975;69(3):739–43.
1950 to 2002. Lancet 2006;367(9511):645. 31. McDonald GB, Sharma P, Matthews DE, Shulman HM, Thomas
4. Allison MC, Mowat A, McCruden EA, et al. The spectrum of ED. Venocclusive disease of the liver after bone marrow transplan-
chronic liver disease in renal transplant recipients. Q J Med tation: diagnosis, incidence, and predisposing factors. Hepatology
1992;83(301):355–67. 1984;4(1):116–22.
5. Everson GT, Taylor MR, Doctor RB. Polycystic disease of the liver. 32. Haboubi NY, Ali HH, Whitwell HL, Ackrill P. Role of endothelial cell
Hepatology 2004;40(4):774–82. injury in the spectrum of azathioprine-induced liver disease after
6. Reed B, McFann K, Kimberling WJ, et al. Presence of de novo muta- renal transplant: light microscopy and ultrastructural observations.
tions in autosomal dominant polycystic kidney disease patients Am J Gastroenterol 1988;83(3):256–61.
without family history. Am J Kidney Dis 2008;52(6):1042–50. 33. Arber N, Zajicek G, Nordenberg J, Sidi Y. Azathioprine treat-
7. Bae KT, Zhu F, Chapman AB, et al. Magnetic resonance imaging ment increases hepatocyte turnover. Gastroenterology
evaluation of hepatic cysts in early autosomal-dominant polycystic 1991;101(4):1083–6.
kidney disease: the consortium for radiologic imaging studies of poly- 34. Horsmans Y, Rahier J, Geubel AP. Reversible cholestasis with bile
cystic kidney disease cohort. Clin J Am Soc Nephrol 2006;1(1):64–9. duct injury following azathioprine therapy: a case report. Liver
8. Sherstha R, McKinley C, Russ P, et al. Postmenopausal estrogen 1991;11(2):89–93.
therapy selectively stimulates hepatic enlargement in women 35. Shaye OA, Yadegari M, Abreu MT, et al. Hepatotoxicity of 6-mercap-
with autosomal dominant polycystic kidney disease. Hepatology topurine (6-MP) and Azathioprine (AZA) in adult IBD patients. Am J
1997;26(5):1282–6. Gastroenterol 2007;102(11):2488–94.
9. Chauveau D, Fakhouri F, Grunfeld JP. Liver involvement in autoso- 36. Olsen TS, Fjeldborg O, Hansen HE. Portal hypertension with-
mal-dominant polycystic kidney disease: therapeutic dilemma. J Am out liver cirrhosis in renal transplant recipients. APMIS Suppl
Soc Nephrol 2000;11(9):1767–75. 1991;23:13–20.
10. Doshi SD, Bittermann T, Schiano TD, Goldberg DS. Waitlisted can- 37. Shulman HM, Gooley T, Dudley MD, et al. Utility of transvenous
didates with polycystic liver disease are more likely to be trans- liver biopsies and wedged hepatic venous pressure measure-
planted than those with chronic liver failure. Transplantation ments in sixty marrow transplant recipients. Transplantation
2017;101(8):1838–44. 1995;59(7):1015–22.
32 • Liver Disease Among Renal Transplant Recipients 561

38. Plessier A, Rautou PE, Valla DC. Management of hepatic vascular 62. Lavanchy D. Hepatitis B virus epidemiology, disease burden, treat-
diseases. J Hepatol 2012;56(Suppl. 1):S25–38. ment, and current and emerging prevention and control measures. J
39. Kohli HS, Jain D, Sud K, et al. Azathioprine-induced hepatic veno- Viral Hepat 2004;11(2):97–107.
occlusive disease in a renal transplant recipient: histological regres- 63. Allain JP. Epidemiology of hepatitis B virus and genotype. J Clin Virol
sion following azathioprine withdrawal. Nephrol Dial Transplant 2006;36(Suppl. 1):S12–7.
1996;11(8):1671–2. 64. Fornairon S, Pol S, Legendre C, et al. The long-term virologic and
40. Azoulay D, Castaing D, Lemoine A, et al. Successful treatment of pathologic impact of renal transplantation on chronic hepatitis B
severe azathioprine-induced hepatic veno-occlusive disease in a virus infection. Transplantation 1996;62(2):297–9.
kidney-transplanted patient with transjugular intrahepatic porto- 65. Bortolotti F, Guido M, Bartolacci S, et al. Chronic hepatitis B in chil-
systemic shunt. Clin Nephrol 1998;50(2):118–22. dren after e antigen seroclearance: final report of a 29-year longitu-
41. Buffet C, Cantarovitch M, Pelletier G, et al. Three cases of nodular dinal study. Hepatology 2006;43(3):556–62.
regenerative hyperplasia of the liver following renal transplantation. 66. Aroldi A, Lampertico P, Montagnino G, et al. Natural history of
Nephrol Dial Transplant 1988;3(3):327–30. hepatitis B and C in renal allograft recipients. Transplantation
42. Peters MG, Andersen J, Lynch P, et al. Randomized controlled study 2005;79(9):1132–6.
of tenofovir and adefovir in chronic hepatitis B virus and HIV infec- 67. Fabrizi F, Bunnapradist S, Lunghi G, Aucella F, Martin P. Epidemi-
tion: ACTG A5127. Hepatology 2006;44(5):1110–6. ology and clinical significance of hepatotropic infections in dialy-
43. Pol S, Cavalcanti R, Carnot F, et al. Azathioprine hepatitis in kidney sis patients. Recent evidence. Minerva Urol Nefrol 2004;56(3):
transplant recipients. A predisposing role of chronic viral hepatitis. 249–57.
Transplantation 1996;61(12):1774–6. 68. Tokars JI, Frank M, Alter MJ, Arduino MJ. National surveillance of
44. O’Grady JG, Burroughs A, Hardy P, Elbourne D, Truesdale A. dialysis-associated diseases in the United States, 2000. Semin Dial
Tacrolimus versus microemulsified ciclosporin in liver trans- 2002;15(3):162–71.
plantation: the TMC randomised controlled trial. Lancet 69. Wang C, Sun J, Zhu B, et al. Hepatitis B virus infection and related
2002;360(9340):1119–25. factors in hemodialysis patients in China: systematic review and
45. Busuttil RW, Lake JR. Role of tacrolimus in the evolution of liver meta-analysis. Ren Fail 2010;32(10):1255–64.
transplantation. Transplantation 2004;77(9 Suppl.):S44–51. 70. Finelli L, Miller JT, Tokars JI, Alter MJ, Arduino MJ. National sur-
46. Formea CM, Evans CG, Karlix JL. Altered cytochrome p450 metabo- veillance of dialysis-associated diseases in the United States, 2002.
lism of calcineurin inhibitors: case report and review of the litera- Semin Dial 2005;18(1):52–61.
ture. Pharmacotherapy 2005;25(7):1021–9. 71. Chandra M, Khaja MN, Hussain MM, et al. Prevalence of hepatitis B
47. Lorber MI, Van Buren CT, Flechner SM, Williams C, Kahan BD. and hepatitis C viral infections in Indian patients with chronic renal
Hepatobiliary complications of cyclosporine therapy following renal failure. Intervirology 2004;47(6):374–6.
transplantation. Transplant Proc 1987;19(1 Pt 2):1808–10. 72. Miller ER, Alter MJ, Tokars JI. Protective effect of hepatitis B
48. Pirsch JD, Miller J, Deierhoi MH, Vincenti F, Filo RS. A comparison of vaccine in chronic hemodialysis patients. Am J Kidney Dis
tacrolimus (FK506) and cyclosporine for immunosuppression after 1999;33(2):356–60.
cadaveric renal transplantation: FK506 kidney transplant study 73. Sezer S, Ozdemir FN, Guz G, et al. Factors influencing response to
group. Transplantation 1997;63(7):977–83. hepatitis B virus vaccination in hemodialysis patients. Transplant
49. Groth CG, Backman L, Morales JM, et al. Sirolimus (rapamycin)- Proc 2000;32(3):607–8.
based therapy in human renal transplantation: similar efficacy 74. Fabrizi F, Martin P, Dixit V, Bunnapradist S, Dulai G. Meta-anal-
and different toxicity compared with cyclosporine. Sirolimus Euro- ysis: the effect of age on immunological response to hepatitis
pean Renal Transplant Study Group Transplantation 1999;67(7): B vaccine in end-stage renal disease. Aliment Pharmacol Ther
1036–42. 2004;20(10):1053–62.
50. Moreno F, Morales JM, Colina F, et al. Influence of long-term cyclo- 75. DaRoza G, Loewen A, Djurdjev O, et al. Stage of chronic kidney dis-
sporine therapy on chronic liver disease after renal transplantation. ease predicts seroconversion after hepatitis B immunization: earlier
Transplant Proc 1990;22(5):2314–6. is better. Am J Kidney Dis 2003;42(6):1184–92.
51. Manzanares C, Moreno M, Castellanos F, et al. Influence of hepatitis 76. Aguilar P, Renoult E, Jarrosson L, et al. Anti-HBs cellular immune
C virus infection on FK 506 blood levels in renal transplant patients. response in kidney recipients before and 4 months after transplanta-
Transplant Proc 1998;30(4):1264–5. tion. Clin Diagn Lab Immunol 2003;10(6):1117–22.
52. Margreiter R. Efficacy and safety of tacrolimus compared with ciclo- 77. Fabrizi F, Dixit V, Messa P, Martin P. Intradermal vs intramuscular
sporin microemulsion in renal transplantation: a randomised multi- vaccine against hepatitis B infection in dialysis patients: a meta-
centre study. Lancet 2002;359(9308):741–6. analysis of randomized trials. J Viral Hepat 2011;18(10):730–7.
53. Hardwick LL, Batiuk TD. Severe prolonged tacrolimus overdose with 78. Rangel MC, Coronado VG, Euler GL, Strikas RA. Vaccine recommen-
minimal consequences. Pharmacotherapy 2002;22(8):1063–6. dations for patients on chronic dialysis. The Advisory Committee on
54. Jacques J, Dickson Z, Carrier P, et al. Severe sirolimus-induced Immunization Practices and the American Academy of Pediatrics.
acute hepatitis in a renal transplant recipient. Transpl Int Semin Dial 2000;13(2):101–7.
2010;23(9):967–70. 79. Josselson J, Kyser BA, Weir MR, Sadler JH. Hepatitis B surface anti-
55. Niemczyk M, Wyzgal J, Perkowska A, Porowski D, Paczek L. Siroli- genemia in a chronic hemodialysis program: lack of influence on
mus-associated hepatotoxicity in the kidney graft recipient. Transpl morbidity and mortality. Am J Kidney Dis 1987;9(6):456–61.
Int 2005;18(11):1302–3. 80. Cai YJ, Dong JJ, Wang XD, et al. A diagnostic algorithm for assess-
56. Panaro F, Piardi T, Gheza F, et al. Causes of sirolimus discontinuation in ment of liver fibrosis by liver stiffness measurement in patients with
97 liver transplant recipients. Transplant Proc 2011;43(4):1128–31. chronic hepatitis B. J Viral Hepat 2017;24(11):1005–15.
57. Marcos A, Eghtesad B, Fung JJ, et al. Use of alemtuzumab and 81. Terrault NA, Bzowej NH, Chang KM, et al. AASLD guidelines for
tacrolimus monotherapy for cadaveric liver transplantation: treatment of chronic hepatitis B. Hepatology 2016;63(1):261–83.
with particular reference to hepatitis C virus. Transplantation 82. Morales JM, Dominguez-Gil B, Sanz-Guajardo D, Fernandez J, Escuin
2004;78(7):966–71. F. The influence of hepatitis B and hepatitis C virus infection in the
58. Go MR, Bumgardner GL. OKT3 (muromonab-CD3) associ- recipient on late renal allograft failure. Nephrol Dial Transplant
ated hepatitis in a kidney transplant recipient. Transplantation 2004;19(Suppl. 3):iii72–6.
2002;73(12):1957–9. 83. Mathurin P, Mouquet C, Poynard T, et al. Impact of hepatitis
59. Ferrajolo C, Capuano A, Verhamme KM, et al. Drug-induced hepatic B and C virus on kidney transplantation outcome. Hepatology
injury in children: a case/non-case study of suspected adverse drug 1999;29(1):257–63.
reactions in VigiBase. Br J Clin Pharmacol 2010;70(5):721–8. 84. Fabrizi F, Martin P, Dixit V, Kanwal F, Dulai G. HBsAg seropositive
60. Seeger C, Mason WS. Hepatitis B virus biology. Microbiol Mol Biol status and survival after renal transplantation: meta-analysis of
Rev 2000;64(1):51–68. observational studies. Am J Transplant 2005;5(12):2913–21.
61. Yim HJ, Lok AS. Natural history of chronic hepatitis B virus infec- 85. Matos CA, Perez RM, Lemos LB, et al. Factors associated with the
tion: what we knew in 1981 and what we know in 2005. Hepatol- intensity of liver fibrosis in renal transplant patients with hepatitis B
ogy 2006;43(2 Suppl. 1):S173–81. virus infection. Eur J Gastroenterol Hepatol 2007;19(8):653–7.
562 Kidney Transplantation: Principles and Practice

86. Davies SE, Portmann BC, O’Grady JG, et al. Hepatic histological find- 108. Kamar N, Milioto O, Alric L, et al. Entecavir therapy for adefovir-
ings after transplantation for chronic hepatitis B virus infection, resistant hepatitis B virus infection in kidney and liver allograft
including a unique pattern of fibrosing cholestatic hepatitis. Hepa- recipients. Transplantation 2008;86(4):611–4.
tology 1991;13(1):150–7. 109. Colonno RJ, Rose R, Baldick CJ, et al. Entecavir resistance is
87. Yap DY, Tang CS, Yung S, Choy BY, Yuen MF, Chan TM. Long- rare in nucleoside naive patients with hepatitis B. Hepatology
term outcome of renal transplant recipients with chronic hepa- 2006;44(6):1656–65.
titis B infection-impact of antiviral treatments. Transplantation 110. Tenney DJ, Rose RE, Baldick CJ, et al. Long-term monitoring
2010;90(3):325–30. shows hepatitis B virus resistance to entecavir in nucleoside-
88. Fairley CK, Mijch A, Gust ID, Nichilson S, Dimitrakakis M, Lucas CR. naive patients is rare through 5 years of therapy. Hepatology
The increased risk of fatal liver disease in renal transplant patients 2009;49(5):1503–14.
who are hepatitis Be antigen and/or HBV DNA positive. Transplan- 111. Daude M, Rostaing L, Saune K, et al. Tenofovir therapy in hepatitis
tation 1991;52(3):497–500. B virus-positive solid-organ transplant recipients. Transplantation
89. Fong TL, Bunnapradist S, Jordan SC, Cho YW. Impact of hepatitis 2011;91(8):916–20.
B core antibody status on outcomes of cadaveric renal transplanta- 112. Durlik M, Gaciong Z, Rowinska D, et al. Long-term results of treat-
tion: analysis of United network of organ sharing database between ment of chronic hepatitis B, C and D with interferon-alpha in renal
1994 and 1999. Transplantation 2002;73(1):85–9. allograft recipients. Transpl Int 1998;11(Suppl. 1):S135–9.
90. De Feo TM, Grossi P, Poli F, et al. Kidney transplantation from anti- 113. Munoz de Bustillo E, Ibarrola C, Andres A, Colina F, Morales JM.
HBc+ donors: results from a retrospective Italian study. Transplan- Hepatitis-B-virus-related fibrosing cholestatic hepatitis after renal
tation 2006;81(1):76–80. transplantation with acute graft failure following interferon-alpha
91. Madayag RM, Johnson LB, Bartlett ST, et al. Use of renal allografts therapy. Nephrol Dial Transplant 1998;13(6):1574–6.
from donors positive for hepatitis B core antibody confers minimal 114. Kairaitis LK, Gottlieb T, George CR. Fatal hepatitis B virus infection
risk for subsequent development of clinical hepatitis B virus disease. with fibrosing cholestatic hepatitis following renal transplantation.
Transplantation 1997;64(12):1781–6. Nephrol Dial Transplant 1998;13(6):1571–3.
92. Akalin E, Ames S, Sehgal V, Murphy B, Bromberg JS. Safety of 115. Chan TM, Wu PC, Li FK, Lai CL, Cheng IK, Lai KN. Treatment of
using hepatitis B virus core antibody or surface antigen-positive fibrosing cholestatic hepatitis with lamivudine. Gastroenterology
donors in kidney or pancreas transplantation. Clin Transplant 1998;115(1):177–81.
2005;19(3):364–6. 116. Feinstone SM, Kapikian AZ, Purceli RH. Hepatitis A: detection by
93. Lee J, Park JY, Huh KH, et al. Rituximab and hepatitis B reactivation immune electron microscopy of a viruslike antigen associated with
in HBsAg-negative/anti-HBc-positive kidney transplant recipients. acute illness. Science 1973;182(116):1026–8.
Nephrol Dial Transplant 2017;32(5):906. 117. Choo QL, Kuo G, Weiner AJ, Overby LR, Bradley DW, Houghton M.
94. Han DJ, Kim TH, Park SK, et al. Results on preemptive or prophylac- Isolation of a cDNA clone derived from a blood-borne non-A, non-B
tic treatment of lamivudine in HBsAg (+) renal allograft recipients: viral hepatitis genome. Science 1989;244(4902):359–62.
comparison with salvage treatment after hepatic dysfunction with 118. Kuiken C, Simmonds P. Nomenclature and numbering of the hepati-
HBV recurrence. Transplantation 2001;71(3):387–94. tis C virus. Methods Mol Biol 2009;510:33–53.
95. Chan TM, Fang GX, Tang CS, Cheng IK, Lai KN, Ho SK. Preemptive 119. Bukh J, Purcell RH, Miller RH. At least 12 genotypes of hepatitis C
lamivudine therapy based on HBV DNA level in HBsAg-positive kid- virus predicted by sequence analysis of the putative E1 gene of isolates
ney allograft recipients. Hepatology 2002;36(5):1246–52. collected worldwide. Proc Natl Acad Sci U S A 1993;90(17):8234–8.
96. Fabrizi F, Dulai G, Dixit V, Bunnapradist S, Martin P. Lamivudine 120. Hissar SS, Goyal A, Kumar M, et al. Hepatitis C virus genotype 3
for the treatment of hepatitis B virus-related liver disease after renal predominates in North and Central India and is associated with
transplantation: meta-analysis of clinical trials. Transplantation significant histopathologic liver disease. J Med Virol 2006;78(4):
2004;77(6):859–64. 452–8.
97. Liaw YF, Chien RN. Case report: dramatic response to lamivudine 121. Kaba S, Dutta U, Byth K, et al. Molecular epidemiology of hepatitis C
therapy following corticosteroid priming in chronic hepatitis B. J in Australia. J Gastroenterol Hepatol 1998;13(9):914–20.
Gastroenterol Hepatol 1999;14(8):804–6. 122. Zein NN. Clinical significance of hepatitis C virus genotypes. Clin
98. Fabrizi F, Martin P, Bunnapradist S. Treatment of chronic viral Microbiol Rev 2000;13(2):223–35.
hepatitis in patients with renal disease. Gastroenterol Clin North Am 123. Amoroso P, Rapicetta M, Tosti ME, et al. Correlation between
2004;33(3):655–70, xi. virus genotype and chronicity rate in acute hepatitis C. J Hepatol
99. Kletzmayr J, Watschinger B, Muller C, et al. Twelve months of lami- 1998;28(6):939–44.
vudine treatment for chronic hepatitis B virus infection in renal 124. Thimme R, Oldach D, Chang KM, Steiger C, Ray SC, Chisari FV.
transplant recipients. Transplantation 2000;70(9):1404–7. Determinants of viral clearance and persistence during acute hepati-
100. Rostaing L, Henry S, Cisterne JM, Duffaut M, Icart J, Durand D. tis C virus infection. J Exp Med 2001;194(10):1395–406.
Efficacy and safety of lamivudine on replication of recurrent hepa- 125. Wasley A, Alter MJ. Epidemiology of hepatitis C: geographic differ-
titis B after cadaveric renal transplantation. Transplantation ences and temporal trends. Semin Liver Dis 2000;20(1):1–16.
1997;64(11):1624–7. 126. Farci P, Alter HJ, Shimoda A, et al. Hepatitis C virus-associated fulmi-
101. Hadziyannis SJ, Tassopoulos NC, Heathcote EJ, et al. Long-term ther- nant hepatic failure. N Engl J Med 1996;335(9):631–4.
apy with adefovir dipivoxil for HBeAg-negative chronic hepatitis B. 127. Lai ME, Mazzoleni AP, Argiolu F, et al. Hepatitis C virus in multiple
N Engl J Med 2005;352(26):2673–81. episodes of acute hepatitis in polytransfused thalassaemic children.
102. Marcellin P, Chang TT, Lim SG, et al. Adefovir dipivoxil for the treat- Lancet 1994;343(8894):388–90.
ment of hepatitis B e antigen-positive chronic hepatitis B. N Engl J 128. Bukh J, Wantzin P, Krogsgaard K, Knudsen F, Purcell RH, Miller RH.
Med 2003;348(9):808–16. High prevalence of hepatitis C virus (HCV) RNA in dialysis patients:
103. Fontaine H, Vallet-Pichard A, Chaix ML, et al. Efficacy and failure of commercially available antibody tests to identify a signifi-
safety of adefovir dipivoxil in kidney recipients, hemodialysis cant number of patients with HCV infection. Copenhagen Dialysis
patients, and patients with renal insufficiency. Transplantation HCV Study Group. J Infect Dis 1993;168(6):1343–8.
2005;80(8):1086–92. 129. Cacoub P, Renou C, Rosenthal E, et al. Extrahepatic manifestations
104. Peters MG, Hann Hw H, Martin P, et al. Adefovir dipivoxil alone or in associated with hepatitis C virus infection. A prospective multicenter
combination with lamivudine in patients with lamivudine-resistant study of 321 patients. The GERMIVIC. Groupe d’Etude et de Recher-
chronic hepatitis B. Gastroenterology 2004;126(1):91–101. che en Medecine Interne et Maladies Infectieuses sur le Virus de
105. Kamar N, Huart A, Tack I, Alric L, Izopet J, Rostaing L. Renal side l’Hepatite C. Medicine (Baltimore) 2000;79(1):47–56.
effects of adefovir in hepatitis B virus-(HBV) positive kidney allograft 130. Poynard T, Bedossa P, Opolon P. Natural history of liver fibro-
recipients. Clin Nephrol 2009;71(1):36–42. sis progression in patients with chronic hepatitis C. The OBS-
106. Chang TT, Gish RG, de Man R, et al. A comparison of entecavir and VIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet
lamivudine for HBeAg-positive chronic hepatitis B. N Engl J Med 1997;349(9055):825–32.
2006;354(10):1001–10. 131. Armstrong GL, Wasley A, Simard EP, McQuillan GM, Kuhnert
107. Hu TH, Tsai MC, Chien YS, et al. A novel experience of antiviral WL, Alter MJ. The prevalence of hepatitis C virus infection in the
therapy for chronic hepatitis B in renal transplant recipients. Antivir United States, 1999 through 2002. Ann Intern Med 2006;144(10):
Ther 2012;17(4):745–53. 705–14.
32 • Liver Disease Among Renal Transplant Recipients 563

132. Dodd RY, Notari EPT, Stramer SL. Current prevalence and inci- 155. Kamar N, Ribes D, Izopet J, Rostaing L. Treatment of hepatitis C virus
dence of infectious disease markers and estimated window-period infection (HCV) after renal transplantation: implications for HCV-
risk in the American Red Cross blood donor population. Transfusion positive dialysis patients awaiting a kidney transplant. Transplanta-
2002;42(8):975–9. tion 2006;82(7):853–6.
133. Fissell RB, Bragg-Gresham JL, Woods JD, et al. Patterns of hepatitis 156. Fabrizi F, Martin P, Dixit V, Bunnapradist S, Dulai G. Hepatitis C
C prevalence and seroconversion in hemodialysis units from three virus antibody status and survival after renal transplantation:
continents: the DOPPS. Kidney Int 2004;65(6):2335–42. meta-analysis of observational studies. Am J Transplant 2005;5(6):
134. Sivapalasingam S, Malak SF, Sullivan JF, Lorch J, Sepkowitz KA. 1452–61.
High prevalence of hepatitis C infection among patients receiving 157. Goodkin DA, Bieber B, Jadoul M, Martin P, Kanda E, Pisoni RL.
hemodialysis at an urban dialysis center. Infect Control Hosp Epide- Mortality, hospitalization, and quality of life among patients
miol 2002;23(6):319–24. with hepatitis C infection on hemodialysis. Clin J Am Soc Nephrol
135. Donahue JG, Munoz A, Ness PM, et al. The declining risk of post- 2017;12(2):287–97.
transfusion hepatitis C virus infection. N Engl J Med 1992;327(6): 158. Goodkin DA, Bragg-Gresham JL, Koenig KG, et al. Association of
369–73. comorbid conditions and mortality in hemodialysis patients in Europe,
136. Knudsen F, Wantzin P, Rasmussen K, et al. Hepatitis C in dialysis Japan, and the United States: the Dialysis Outcomes and Practice Pat-
patients: relationship to blood transfusions, dialysis and liver disease. terns Study (DOPPS). J Am Soc Nephrol 2003;14(12):3270–7.
Kidney Int 1993;43(6):1353–6. 159. Davis MI, Chute DF, Chung RT, Sise ME. When and how can nephrol-
137. Hinrichsen H, Leimenstoll G, Stegen G, Schrader H, Folsch UR, ogists treat hepatitis C virus infection in dialysis patients? Semin
Schmidt WE. Prevalence and risk factors of hepatitis C virus infec- Dial 2018 Jan;31(1):26–36. Available online at: https://doi.org/
tion in haemodialysis patients: a multicentre study in 2796 patients. 10.1111/sdi.12650.
Gut 2002;51(3):429–33. 160. Bloom RD, Sayer G, Fa K, Constantinescu S, Abt P, Reddy KR. Out-
138. De Vos JY, Elseviers M, Harrington M, et al. Infection control practice come of hepatitis C virus-infected kidney transplant candidates who
across Europe: results of the EPD. Edtna Erca J 2006;32(1):38–41. remain on the waiting list. Am J Transplant 2005;5(1):139–44.
139. Fabrizi F, Poordad FF, Martin P. Hepatitis C infection and the patient 161. Sawinski D, Kaur N, Ajeti A, et al. Successful treatment of hepatitis C
with end-stage renal disease. Hepatology 2002;36(1):3–10. in renal transplant recipients with direct-acting antiviral agents. Am
140. Abbott KC, Lentine KL, Bucci JR, Agodoa LY, Peters TG, Schnitzler J Transplant 2016;16(5):1588–95.
MA. The impact of transplantation with deceased donor hepati- 162. Liu CH, Liang CC, Huang KW, et al. Transient elastography to assess
tis c-positive kidneys on survival in wait-listed long-term dialysis hepatic fibrosis in hemodialysis chronic hepatitis C patients. Clin J
patients. Am J Transplant 2004;4(12):2032–7. Am Soc Nephrol 2011;6(5):1057–65.
141. Morales JM, Campistol JM, Castellano G, et al. Transplantation of 163. Delladetsima I, Psichogiou M, Sypsa V, et al. The course of hepatitis C
kidneys from donors with hepatitis C antibody into recipients with virus infection in pretransplantation anti-hepatitis C virus-negative
pre-transplantation anti-HCV. Kidney Int 1995;47(1):236–40. renal transplant recipients: a retrospective follow-up study. Am J
142. Mandal AK, Kraus ES, Samaniego M, et al. Shorter waiting times Kidney Dis 2006;47(2):309–16.
for hepatitis C virus seropositive recipients of cadaveric renal 164. Fabrizi F, Martin P, Dixit V, Bunnapradist S, Kanwal F, Dulai G. Post-
allografts from hepatitis C virus seropositive donors. Clin Transplant transplant diabetes mellitus and HCV seropositive status after renal
2000;14(4 Pt 2):391–6. transplantation: meta-analysis of clinical studies. Am J Transplant
143. Ali MK, Light JA, Barhyte DY, et al. Donor hepatitis C virus status 2005;5(10):2433–40.
does not adversely affect short-term outcomes in HCV+ recipients in 165. Yildiz A, Tutuncu Y, Yazici H, et al. Association between hepa-
renal transplantation. Transplantation 1998;66(12):1694–7. titis C virus infection and development of posttransplantation
144. Goldberg DS, Abt PL, Blumberg EA, et al. Trial of transplantation diabetes mellitus in renal transplant recipients. Transplantation
of HCV-infected kidneys into uninfected recipients. N Engl J Med 2002;74(8):1109–13.
2017;376(24):2394–5. 166. Bloom RD, Rao V, Weng F, Grossman RA, Cohen D, Mange KC. Asso-
145. Stevens PE, Levin A. Kidney disease: improving global outcomes ciation of hepatitis C with posttransplant diabetes in renal transplant
chronic kidney disease guideline development work group M. Evalu- patients on tacrolimus. J Am Soc Nephrol 2002;13(5):1374–80.
ation and management of chronic kidney disease: synopsis of the 167. Morales JM, Fabrizi F. Hepatitis C and its impact on renal transplan-
kidney disease: improving global outcomes 2012 clinical practice tation. Nat Rev Nephrol 2015;11(3):172–82.
guideline. Ann Intern Med 2013;158(11):825–30. 168. Meyers CM, Seeff LB, Stehman-Breen CO, Hoofnagle JH. Hepatitis C
146. Goldberg DS, Abt PL, Reese PP, Investigators TT. Transplant- and renal disease: an update. Am J Kidney Dis 2003;42(4):631–57.
ing HCV-infected kidneys into uninfected recipients. N Engl J Med 169. Cruzado JM, Carrera M, Torras J, Grinyo JM. Hepatitis C virus infec-
2017;377(11):1105. tion and de novo glomerular lesions in renal allografts. Am J Trans-
147. Orloff SL, Stempel CA, Wright TL, et al. Long-term outcome in kidney plant 2001;1(2):171–8.
transplant patients with hepatitis C (HCV) infection. Clin Transplant 170. van Duijnhoven EM, Christiaans MH, Boots JM, Nieman FH,
1995;9(2):119–24. Wolffenbuttel BH, van Hooff JP. Glucose metabolism in the first 3
148. Stempel CA, Lake J, Kuo G, Vincenti F. Hepatitis C—its prevalence years after renal transplantation in patients receiving tacrolimus
in end-stage renal failure patients and clinical course after kidney versus cyclosporine-based immunosuppression. J Am Soc Nephrol
transplantation. Transplantation 1993;55(2):273–6. 2002;13(1):213–20.
149. Alric L, Di-Martino V, Selves J, et al. Long-term impact of renal trans- 171. Inoue K, Yoshiba M. Interferon combined with cyclosporine treat-
plantation on liver fibrosis during hepatitis C virus infection. Gastro- ment as an effective countermeasure against hepatitis C virus recur-
enterology 2002;123(5):1494–9. rence in liver transplant patients with end-stage hepatitis C virus
150. Lee WC, Shu KH, Cheng CH, Wu MJ, Chen CH, Lian JC. Long-term related disease. Transplant Proc 2005;37(2):1233–4.
impact of hepatitis B, C virus infection on renal transplantation. Am 172. Cescon M, Grazi GL, Cucchetti A, et al. Predictors of sustained viro-
J Nephrol 2001;21(4):300–6. logical response after antiviral treatment for hepatitis C recurrence
151. Breitenfeldt MK, Rasenack J, Berthold H, et al. Impact of hepatitis B following liver transplantation. Liver Transpl 2009;15(7):782–9.
and C on graft loss and mortality of patients after kidney transplanta- 173. McCaughan GW, Zekry A. Impact of immunosuppression on immu-
tion. Clin Transplant 2002;16(2):130–6. nopathogenesis of liver damage in hepatitis C virus-infected recipients
152. Legendre C, Garrigue V, Le Bihan C, et al. Harmful long-term impact following liver transplantation. Liver Transpl 2003;9(11):S21–7.
of hepatitis C virus infection in kidney transplant recipients. Trans- 174. Charlton M, Seaberg E. Impact of immunosuppression and acute
plantation 1998;65(5):667–70. rejection on recurrence of hepatitis C: results of the National Insti-
153. Hanafusa T, Ichikawa Y, Kishikawa H, et al. Retrospective study on tute of Diabetes and Digestive and Kidney Diseases Liver Transplan-
the impact of hepatitis C virus infection on kidney transplant patients tation Database. Liver Transpl Surg 1999;5(4 Suppl. 1):S107–14.
over 20 years. Transplantation 1998;66(4):471–6. 175. Huraib S, Iqbal A, Tanimu D, Abdullah A. Sustained virological and
154. Periera BJ, Wright TL, Schmid CH, Levey AS. The impact of pretrans- histological response with pretransplant interferon therapy in renal
plantation hepatitis C infection on the outcome of renal transplanta- transplant patients with chronic viral hepatitis C. Am J Nephrol
tion. Transplantation 1995;60(8):799–805. 2001;21(6):435–40.
564 Kidney Transplantation: Principles and Practice

176. Kamar N, Toupance O, Buchler M, et al. Evidence that clearance 199. Yasui K, Sumida Y, Mori Y, et al. Nonalcoholic steatohepati-
of hepatitis C virus RNA after alpha-interferon therapy in dialysis tis and increased risk of chronic kidney disease. Metabolism
patients is sustained after renal transplantation. J Am Soc Nephrol 2011;60(5):735–9.
2003;14(8):2092–8. 200. Musso G, Gambino R, Tabibian JH, et al. Association of non-alcoholic
177. Cruzado JM, Casanovas-Taltavull T, Torras J, Baliellas C, Gil-Vernet fatty liver disease with chronic kidney disease: a systematic review
S, Grinyo JM. Pretransplant interferon prevents hepatitis C virus- and meta-analysis. PLoS Med 2014;11(7): e1001680.
associated glomerulonephritis in renal allografts by HCV-RNA clear- 201. Musso G, De Michieli F, Bongiovanni D, et al. New pharmaco-
ance. Am J Transplant 2003;3(3):357–60. logic agents that target inflammation and fibrosis in nonalcoholic
178. Baid S, Tolkoff-Rubin N, Saidman S, et al. Acute humoral rejection steatohepatitis-related kidney disease. Clin Gastroenterol Hepatol
in hepatitis C-infected renal transplant recipients receiving antiviral 2017;15(7):972–85.
therapy. Am J Transplant 2003;3(1):74–8. 202. Singal AK, Hasanin M, Kaif M, Wiesner R, Kuo YF. Nonalcoholic ste-
179. Rostaing L, Izopet J, Baron E, Duffaut M, Puel J, Durand D. Treatment atohepatitis is the most rapidly growing indication for simultaneous
of chronic hepatitis C with recombinant interferon alpha in kidney liver kidney transplantation in the United States. Transplantation
transplant recipients. Transplantation 1995;59(10):1426–31. 2016;100(3):607–12.
180. Shiffman ML, Esteban R. Triple therapy for HCV genotype 1 infec- 203. Mikolasevic I, Racki S, Lukenda V, Milic S, Pavletic-Persic M, Orlic
tion: telaprevir or boceprevir? Liver Int 2012;32(Suppl. 1):54–60. L. Nonalcoholic fatty liver disease in renal transplant recipients
181. Soriano V, Fernandez-Montero JV, de Mendoza C, et al. Treatment of proven by transient elastography. Transplant Proc 2014;46(5):
hepatitis C with new fixed dose combinations. Expert Opin Pharma- 1347–52.
cother 2017;18(12):1235–42. 204. Ekstedt M, Hagstrom H, Nasr P, et al. Fibrosis stage is the strongest
182. American Association for the Study of Liver Diseases and the Infec- predictor for disease-specific mortality in NAFLD after up to 33 years
tious Diseases Society of America. Guidelines AHP. Available online of follow-up. Hepatology 2015;61(5):1547–54.
at: https://www.hcvguidelines.org/ 205. Zelle DM, Corpeleijn E, van Ree RM, et al. Markers of the hepatic
183. Bruchfeld A, Roth D, Martin P, et al. Elbasvir plus grazoprevir in component of the metabolic syndrome as predictors of mortality in
patients with hepatitis C virus infection and stage 4-5 chronic renal transplant recipients. Am J Transplant 2010;10(1):106–14.
kidney disease: clinical, virological, and health-related quality- 206. Mikolasevic I, Racki S, Zaputovic L, Lukenda V, Sladoje-Martinovic
of-life outcomes from a phase 3, multicentre, randomised, dou- B, Orlic L. Nonalcoholic fatty liver disease (NAFLD) and cardiovas-
ble-blind, placebo-controlled trial. Lancet Gastroenterol Hepatol cular risk in renal transplant recipients. Kidney Blood Press Res
2017;2(8):585–94. 2014;39(4):308–14.
184. Gane E, Lawitz E, Pugatch D, et al. Glecaprevir and pibrentasvir 207. Targher G, Marra F, Marchesini G. Increased risk of cardiovascular
in patients with HCV and Severe renal impairment. N Engl J Med disease in non-alcoholic fatty liver disease: causal effect or epiphe-
2017;377(15):1448–55. nomenon? Diabetologia 2008;51(11):1947–53.
185. Li T, Qu Y, Guo Y, Wang Y, Wang L. Efficacy and safety of direct-act- 208. Rajman I, Harper L, McPake D, Kendall MJ, Wheeler DC. Low-den-
ing antivirals-based antiviral therapies for hepatitis C virus patients sity lipoprotein subfraction profiles in chronic renal failure. Nephrol
with stage 4-5 chronic kidney disease: a meta-analysis. Liver Int Dial Transplant 1998;13(9):2281–7.
2017;37(7):974–81. 209. Emdin M, Pompella A, Paolicchi A. Gamma-glutamyltransferase,
186. Gentil MA, Gonzalez-Corvillo C, Perello M, et al. Hepatitis C treatment atherosclerosis, and cardiovascular disease: triggering oxidative
with direct-acting antivirals in kidney transplant: preliminary results stress within the plaque. Circulation 2005;112(14):2078–80.
from a multicenter study. Transplant Proc 2016;48(9):2944–6. 210. Bakker SJ, RG IJ, Teerlink T, Westerhoff HV, Gans RO, Heine RJ.
187. Fernandez I, Munoz-Gomez R, Pascasio JM, et al. Efficacy and toler- Cytosolic triglycerides and oxidative stress in central obesity: the
ability of interferon-free antiviral therapy in kidney transplant recip- missing link between excessive atherosclerosis, endothelial dysfunc-
ients with chronic hepatitis C. J Hepatol 2017;66(4):718–23. tion, and beta-cell failure? Atherosclerosis 2000;148(1):17–21.
188. Lin MV, Sise ME, Pavlakis M, et al. Efficacy and safety of direct act- 211. Mikolasevic I, Racki S, Lukenda V, Pavletic-Persic M, Milic S, Orlic
ing antivirals in kidney transplant recipients with chronic hepatitis L. Non-alcoholic fatty liver disease; a part of the metabolic syndrome
C virus infection. PLoS One 2016;11(7): e0158431. in the renal transplant recipient and possible cause of an allograft
189. Eisenberger U, Guberina H, Willuweit K, et al. Successful treatment dysfunction. Med Hypotheses 2014;82(1):36–9.
of chronic hepatitis C virus infection with sofosbuvir and ledipasvir 212. Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and man-
in renal transplant recipients. Transplantation 2017;101(5):980–6. agement of nonalcoholic fatty liver disease: practice guidance from
190. Kamar N, Marion O, Rostaing L, et al. Efficacy and safety of sofosbu- the American Association for the Study of Liver Diseases. Hepatology
vir-based antiviral therapy to treat hepatitis C virus infection after 2018;67(1):328–57. Available online at: https://doi.org/10.1002/
kidney transplantation. Am J Transplant 2016;16(5):1474–9. hep.29367.
191. Colombo M, Aghemo A, Liu H, et al. Treatment with ledipasvir-sofos- 213. Said A, Akhter A. Meta-analysis of randomized controlled trials of
buvir for 12 or 24 weeks in kidney transplant recipients with chronic pharmacologic agents in non-alcoholic steatohepatitis. Ann Hepatol
hepatitis C virus genotype 1 or 4 infection: a randomized trial. Ann 2017;16(4):538–47.
Intern Med 2017;166(2):109–17. 214. Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and man-
192. Guu TS, Liu Z, Ye Q, et al. Structure of the hepatitis E virus-like par- agement of non-alcoholic fatty liver disease: practice Guideline by
ticle suggests mechanisms for virus assembly and receptor binding. the American Association for the Study of Liver Diseases, American
Proc Natl Acad Sci U S A 2009;106(31):12992–7. College of Gastroenterology, and the American Gastroenterological
193. Balayan MS, Andjaparidze AG, Savinskaya SS, et al. Evidence for a Association. Hepatology 2012;55(6):2005–23.
virus in non-A, non-B hepatitis transmitted via the fecal-oral route. 215. Musso G, Cassader M, Cohney S, Pinach S, Saba F, Gambino R.
Intervirology 1983;20(1):23–31. Emerging liver-kidney interactions in nonalcoholic fatty liver dis-
194. Kamar N, Weclawiak H, Guilbeau-Frugier C, et al. Hepatitis E virus ease. Trends Mol Med 2015;21(10):645–62.
and the kidney in solid-organ transplant patients. Transplantation 216. Hirata T, Tomita K, Kawai T, et al. Effect of telmisartan or losartan
2012;93(6):617–23. for treatment of nonalcoholic fatty liver disease: Fatty Liver Protec-
195. Kamar N, Mansuy JM, Cointault O, et al. Hepatitis E virus-related tion Trial by Telmisartan or Losartan Study (FANTASY). Int J Endo-
cirrhosis in kidney- and kidney-pancreas-transplant recipients. Am J crinol 2013;2013:587140.
Transplant 2008;8(8):1744–8. 217. Jeng LB, Huang CC, Lai MK, Chu SH. Hepatocellular carcinoma after
196. Kamar N, Izopet J, Tripon S, et al. Ribavirin for chronic hepa- kidney transplantation. Transplant Proc 1999;31(1-2):1273–4.
titis E virus infection in transplant recipients. N Engl J Med 218. Oldakowska-Jedynak M, Durlik M, Paczek L, et al. Hepatocellular
2014;370(12):1111–20. carcinoma development in renal allograft recipients. Transplant
197. Kim CH, Younossi ZM. Nonalcoholic fatty liver disease: a manifesta- Proc 2000;32(6):1363–4.
tion of the metabolic syndrome. Cleve Clin J Med 2008;75(10):721–8. 219. Ridruejo E, Mando OG, Davalos M, Diaz C, Vilches A. Hepatocel-
198. Targher G, Bertolini L, Rodella S, Lippi G, Zoppini G, Chonchol M. lular carcinoma in renal transplant patients. Transplant Proc
Relationship between kidney function and liver histology in sub- 2005;37(5):2086–8.
jects with nonalcoholic steatohepatitis. Clin J Am Soc Nephrol 220. Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet
2010;5(12):2166–71. 2003;362(9399):1907–17.
32 • Liver Disease Among Renal Transplant Recipients 565

221. Chok KS, Lam CM, Li FK, et al. Management of hepatocellular car- 248. Babcock GJ, Decker LL, Volk M, Thorley-Lawson DA. EBV persistence
cinoma in renal transplant recipients. J Surg Oncol 2004;87(3): in memory B cells in vivo. Immunity 1998;9(3):395–404.
139–42. 249. Walker RC, Marshall WF, Strickler JG, et al. Pretransplantation
222. Chiang YJ, Chen CH, Wu CT, et al. De novo cancer occurrence assessment of the risk of lymphoproliferative disorder. Clin Infect Dis
after renal transplantation: a medical center experience in Taiwan. 1995;20(5):1346–53.
Transplant Proc 2004;36(7):2150–1. 250. Whitley RJ, Kimberlin DW, Roizman B. Herpes simplex viruses. Clin
223. Hoffmann CJ, Subramanian AK, Cameron AM, Engels EA. Incidence Infect Dis 1998;26(3):541–53.
and risk factors for hepatocellular carcinoma after solid organ trans- 251. Amir J. Clinical aspects and antiviral therapy in primary herpetic
plantation. Transplantation 2008;86(6):784–90. gingivostomatitis. Paediatr Drugs 2001;3(8):593–7.
224. Bruix J, Sherman M, American Association for the Study of Liver Dis- 252. Walker DP, Longson M, Mallick NP, Johnson RW. A prospective
ease. Management of hepatocellular carcinoma: an update. Hepatol- study of cytomegalovirus and herpes simplex virus disease in renal
ogy 2011;53(3):1020–2. transplant recipients. J Clin Pathol 1982;35(11):1190–3.
225. Johannsen EC, Sifri CD, Madoff LC. Pyogenic liver abscesses. Infect 253. Seale L, Jones CJ, Kathpalia S, et al. Prevention of herpesvirus infec-
Dis Clin North Am 2000;14(3):547–63, vii. tions in renal allograft recipients by low-dose oral acyclovir. JAMA
226. Kaplan GG, Gregson DB, Laupland KB. Population-based study of the 1985;254(24):3435–8.
epidemiology of and the risk factors for pyogenic liver abscess. Clin 254. Jirasiritham S, Sumethkul V, Chiewsilp P, Gojaseni P. Prevention of
Gastroenterol Hepatol 2004;2(11):1032–8. recurrent herpes infection after renal transplantation by low-dose
227. Rahimian J, Wilson T, Oram V, Holzman RS. Pyogenic liver oral acyclovir. Transplant Proc 1994;26(4):2125–6.
abscess: recent trends in etiology and mortality. Clin Infect Dis 255. Kusne S, Schwartz M, Breinig MK, et al. Herpes simplex virus
2004;39(11):1654–9. hepatitis after solid organ transplantation in adults. J Infect Dis
228. Huang CJ, Pitt HA, Lipsett PA, et al. Pyogenic hepatic abscess. 1991;163(5):1001–7.
Changing trends over 42 years. Ann Surg 1996;223(5):600–7; dis- 256. Gnann Jr JW, Whitley RJ. Clinical practice. Herpes zoster. N Engl J
cussion 607-9. Med 2002;347(5):340–6.
229. Calvo-Romero JM, Lima-Rodriguez EM. Favourable outcome of mul- 257. Cohen JI, Brunell PA, Straus SE, Krause PR. Recent advances in vari-
tiple pyogenic liver abscesses with conservative treatment. Scand J cella-zoster virus infection. Ann Intern Med 1999;130(11):922–32.
Infect Dis 2005;37(2):141–2. 258. Fehr T, Bossart W, Wahl C, Binswanger U. Disseminated varicella
230. Zerem E, Hadzic A. Sonographically guided percutaneous catheter infection in adult renal allograft recipients: four cases and a review
drainage versus needle aspiration in the management of pyogenic of the literature. Transplantation 2002;73(4):608–11.
liver abscess. Am J Roentgenol 2007;189(3):W138–42. 259. Campadelli-Fiume G, Mirandola P, Menotti L. Human herpesvirus 6:
231. Serste T, Bourgeois N, Vanden Eynden F, Coppens E, Deviere J, Le An emerging pathogen. Emerg Infect Dis 1999;5(3):353–66.
Moine O. Endoscopic drainage of pyogenic liver abscesses with sus- 260. Lautenschlager I, Razonable RR. Human herpesvirus-6 infections in
pected biliary origin. Am J Gastroenterol 2007;102(6):1209–15. kidney, liver, lung, and heart transplantation: review. Transpl Int
232. Hughes MA, Petri Jr WA. Amebic liver abscess. Infect Dis Clin North 2012;25(5):493–502.
Am 2000;14(3):565–82, viii. 261. Morris DJ, Littler E, Arrand JR, Jordan D, Mallick NP, Johnson RW.
233. Stanley Jr SL. Amoebiasis. Lancet. 2003;361(9362):1025–34. Human herpesvirus 6 infection in renal-transplant recipients. N
234. Kotton CN, Ryan ET, Fishman JA. Prevention of infection in adult Engl J Med 1989;320(23):1560–1.
travelers after solid organ transplantation. Am J Transplant 262. Clark DA. Human herpesvirus 6 and human herpesvirus 7: emerg-
2005;5(1):8–14. ing pathogens in transplant patients. Int J Hematol 2002;76(Suppl.
235. Agarwal SK, Gupta S, Dash SC, Bhowmik D, Tiwari SC. Prospective 2):246–52.
randomised trial of isoniazid prophylaxis in renal transplant recipi- 263. Harma M, Hockerstedt K, Lautenschlager I. Human herpesvirus-6
ent. Int Urol Nephrol 2004;36(3):425–31. and acute liver failure. Transplantation 2003;76(3):536–9.
236. Sayiner A, Ece T, Duman S, et al. Tuberculosis in renal transplant 264. Pilmore H, Collins J, Dittmer I, et al. Fatal human herpesvirus-6
recipients. Transplantation 1999;68(9):1268–71. infection after renal transplantation. Transplantation 2009;88(6):
237. Apaydin S, Altiparmak MR, Serdengecti K, Ataman R, Ozturk R, 762–5.
Erek E. Mycobacterium tuberculosis infections after renal transplan- 265. Razonable RR, Zerr DM, AST Infectious Diseases Community of
tation. Scand J Infect Dis 2000;32(5):501–5. Practice. HHV-6, HHV-7 and HHV-8 in solid organ transplant recipi-
238. Alvarez SZ. Hepatobiliary tuberculosis. J Gastroenterol Hepatol ents. Am J Transplant 2009;9(Suppl. 4):S97–100.
1998;13(8):833–9. 266. Fontaine H, Thiers V, Chretien Y, et al. HBV genotypic resistance to
239. Subramanyam SG, Kilpadi AB, Correa M, Pai M. Hepatic TB: four lamivudine in kidney recipients and hemodialyzed patients. Trans-
cases and a review of the literature. Trop Doct 2006;36(2):121–2. plantation 2000;69(10):2090–4.
240. Kotton CN, Fishman JA. Viral infection in the renal transplant recipi- 267. Duarte R, Huraib S, Said R, et al. Interferon-alpha facilitates renal
ent. J Am Soc Nephrol 2005;16(6):1758–74. transplantation in hemodialysis patients with chronic viral hepati-
241. Sagedal S, Hartmann A, Nordal KP, et al. Impact of early cytomega- tis. Am J Kidney Dis 1995;25(1):40–5.
lovirus infection and disease on long-term recipient and kidney graft 268. Thabut D, Thibault V, Bernard-Chabert B, et al. Long-term therapy
survival. Kidney Int 2004;66(1):329–37. with lamivudine in renal transplant recipients with chronic hepatitis
242. Serna-Higuera C, Gonzalez-Garcia M, Milicua JM, Munoz V. B. Eur J Gastroenterol Hepatol 2004;16(12):1367–73.
Acute cholestatic hepatitis by cytomegalovirus in an immuno- 269. Chan TM, Tse KC, Tang CS, Lai KN, Ho SK. Prospective study on
competent patient resolved with ganciclovir. J Clin Gastroenterol lamivudine-resistant hepatitis B in renal allograft recipients. Am J
1999;29(3):276–7. Transplant 2004;4(7):1103–9.
243. Ten Napel CH, Houthoff HJ, The TH. Cytomegalovirus hepatitis in 270. Roth D, Nelson DR, Bruchfeld A, et al. Grazoprevir plus elbasvir in
normal and immune compromised hosts. Liver 1984;4(3):184–94. treatment-naive and treatment-experienced patients with hepati-
244. Biggar RJ, Henle G, Bocker J, Lennette ET, Fleisher G, Henle W. Pri- tis C virus genotype 1 infection and stage 4–5 chronic kidney dis-
mary Epstein-Barr virus infections in African infants. II. Clinical ease (the C-SURFER study): a combination phase 3 study. Lancet
and serological observations during seroconversion. Int J Cancer 2015;386(10003):1537–45.
1978;22(3):244–50. 271. Pockros PJ, Reddy KR, Mantry PS, et al. Efficacy of direct-acting
245. Yao QY, Rickinson AB, Epstein MA. Oropharyngeal shedding of antiviral combination for patients with hepatitis C virus genotype
infectious Epstein-Barr virus in healthy virus-immune donors. A 1 infection and severe renal impairment or end-stage renal dis-
prospective study. Chin Med J (Engl). 1985;98(3):191–6. ease. Gastroenterology 2016;150(7):1590–8.
246. Ebell MH. Epstein-Barr virus infectious mononucleosis. Am Fam 272. Desnoyer A, Pospai D, Le MP, et al. Pharmacokinetics, safety and
Physician 2004;70(7):1279–87. efficacy of a full dose sofosbuvir-based regimen given daily in
247. Drebber U, Kasper HU, Krupacz J, et al. The role of Epstein-Barr virus hemodialysis patients with chronic hepatitis C. J Hepatol 2016;65
in acute and chronic hepatitis. J Hepatol 2006;44(5):879–85. (1):40–7.
33 Neurologic Complications
after Kidney Transplantation
CASEY VICTORIA FARIN and MATTHEW WILLIAM LUEDKE

CHAPTER OUTLINE Neurologic Complications of Chronic Mononeuropathies


Kidney Disease Myopathy
Central Nervous System Neurologic Complications After Kidney
Uremic Encephalopathy Trans­plantation
Movement Disorders Associated With Acute Neurologic Complications (Less Than
Uremia 1 Month)
Seizures Central Nervous System
Pharmacologic Neurotoxicity Peripheral Nervous System
Cerebrovascular Disease Subacute Neurologic Complications
Subdural Hematomas (1–6 Months)
Dialysis Disequilibrium Syndrome Central Nervous System
Hemodialysis-Related Headache Peripheral Nervous System
Dialysis Dementia Chronic Neurologic Complications (More
Sleep Disorders Than 6 Months)
Wernicke’s Encephalopathy Opportunistic Infections
Peripheral Nervous System Posttransplant Lymphoproliferative Disease
Polyneuropathy Cognitive Impairment
Summary

Neurologic complications occur frequently in the setting


CENTRAL NERVOUS SYSTEM
of kidney disease and transplantation, with an incidence
reported at 10% to 21% in the posttransplant setting.1 Neu- When renal failure results in insufficient clearance of nitrogen
rologic complications may arise immediately after trans- waste products, buildup of these products can result in ure-
plantation or can occur months to years after transplant. mia. Patients will present with an elevated blood urea nitro-
Renal disease can also lead to neurologic conditions, some gen (BUN) level; however, the absolute value of the BUN or
of which can be improved or resolved by kidney transplan- BUN to creatinine ratio does not always correlate to the clini-
tation. In this chapter, we will first discuss the neurologic cal presentation.3,4 Acute kidney failure can often result in
complications of chronic kidney disease in terms of cen- more dramatic neurologic clinical presentations than chronic
tral and peripheral neurologic complications. We will also kidney failure.4 The majority of the neurologic manifesta-
discuss the neurologic complications of renal transplanta- tions of uremia affect the central nervous system, but uremic
tion by the timing of the onset of symptoms and by clinical polyneuropathy and uremic myopathy can also be seen (as
localization. Encephalopathy and seizures can occur due to below). In addition to uremia, patients with CKD may exhibit
a variety of etiologies, and are some of the more common neurovascular disease, including subdural hematomas.
symptoms of the neurologic complications detailed in this Dialysis presents a unique challenge in that it can improve
chapter. Tables 33.1 and 33.2 list the most common etiolo- or stabilize many central nervous system sequelae of CKD,
gies of encephalopathy and seizures, respectively, in kidney but it can also lead to new neurologic symptoms and syn-
disease and transplantation by the timing of onset. dromes. Dialysis disequilibrium syndrome, hemodialysis-
related headaches, dialysis dementia, and sleep disorders
may be seen after initiation of renal replacement therapy.
Neurologic Complications of
Chronic Kidney Disease Uremic Encephalopathy
Uremic encephalopathy exists on a spectrum and can
Chronic kidney disease (CKD) affects 12% of adults in the range from fatigue and confusion to coma.3 Initial present-
US.2 Renal failure and its management with hemodialysis ing symptoms may be subtle, consisting of fatigue, apa-
are associated with both central and peripheral nervous thy, clumsiness, and poor concentration.4 Symptoms can
system complications. progress to emotional lability, sluggishness, irritability,
566
33 • Neurologic Complications after Kidney Transplantation 567

TABLE 33.1 Differential Diagnosis of Encephalopathy by Timing of Symptom Onset


DIFFERENTIAL DIAGNOSIS OF ENCEPHALOPATHY
Pretransplant Acute Subacute Chronic
Uremic encephalopathy Allograft dysfunction Allograft dysfunction Allograft dysfunction
Electrolyte disturbance Neurotoxicity from Neurotoxicity from Neurotoxicity from
immunosuppressants immunosuppressants immunosuppressants
Dialysis disequilibrium syndrome PRES PRES PTLD
Wernicke encephalopathy Donor-derived opportunistic Opportunistic infections Opportunistic infections
infections
Dialysis dementia Residual effect of anesthesia Cognitive decline
Subdural hematoma Cerebrovascular complications
Metabolic derangements Metabolic derangements

PRES, posterior reversible encephalopathy syndrome; PTLD, posttransplant lymphoproliferative disease.

TABLE 33.2 Differential Diagnosis of Seizure by Timing of Symptom Onset


DIFFERENTIAL DIAGNOSIS OF SEIZURES
Pretransplant Acute Subacute Chronic
CNI side effect CNI side effect CNI side effect CNI side effect
PRES PRES PRES Metastatic cancer
Uremia Opportunistic infection Opportunistic infection
Electrolyte disturbance Electrolyte disturbance
Hemorrhagic or ischemic stroke
Dialysis disequilibrium syndrome

CNI, calcineurin inhibitor; PRES, posterior reversible encephalopathy syndrome.

and impaired abstract thinking; later symptoms include uremic neurotoxins, inflammatory response in the acute
delirium with visual hallucinations, disorientation, and setting leading to breakdown of the blood-brain barrier,
agitation, before progressing to coma and death. Uremia hormonal disturbances, alterations in intermediary metab-
was usually fatal before the advent of renal replacement olism, and alterations in relative neurotransmitter con-
therapy.3,4 Chronic uremia may cause progressive subtle centrations.3,6 There is no single metabolite that has been
cognitive changes, including psychomotor slowing person- identified as the cause of uremia.7
ality changes, forgetfulness, and symptoms similar to atten-
tion deficit disorder.5 Patients with acute kidney injury or Movement Disorders Associated With Uremia
acutely decompensated chronic kidney disease are more Uremia may present with involuntary movements, includ-
susceptible to severe uremic symptoms due to the rapid rise ing asterixis (sudden loss of tone causing flapping move-
in BUN.3,6 ments), myoclonus (sudden brief muscle jerking, which can
Neuroimaging is typically not necessary to make a diag- be seen in a single limb or generalized), and coarse postural
nosis of uremic encephalopathy in patients with CKD who and kinetic tremor4; these abnormal movements are some-
have altered mental status in the setting of elevated BUN times seen with fasciculations and are referred to as the
or BUN to creatinine ratio or in dialysis patients who have twitch-convulsive syndrome.7 EEG is sometimes performed
missed dialysis; however, imaging may help exclude other to differentiate involuntary movements, which are typically
causes of encephalopathy.3 Patients with chronic uremic asynchronous but may become rhythmic at times, from
encephalopathy may develop reversible magnetic reso- seizures.3 Chorea or parkinsonism occur less frequently,
nance imaging (MRI) changes (low signal intensity on T1 but can be associated with radiologic changes in the basal
imaging and high signal intensity on T2 imaging) in the ganglia.3,8
basal ganglia, periventricular white matter, and internal
capsule, which resolve with dialysis.4 Electroencephalo- Seizures
gram (EEG) may show diffuse slowing, generalized periodic Patients with renal failure may present with seizures in
discharges with triphasic morphology, frontal intermittent the setting of uremia alone or secondary to other struc-
rhythmic theta, or paroxysmal bilateral diffuse high-volt- tural or metabolic complications.3 Up to one-third of
age theta.3,5 EEG may become slower with higher creati- patients with uremic encephalopathy develop seizures,
nine values, with a correlation between the percentage of which may be focal or generalized, convulsive or non-
frequencies below 7 Hz and increase in serum creatinine.4 convulsive.3,5 Convulsive or nonconvulsive status epi-
EEG during sleep may show increased vertex waves, a lack lepticus is common.5 Treatment with antibiotics, such
of spindles in stage 2 sleep, and prolonged high-voltage, as beta-lactam penicillins, cephalosporins, carbapenems,
slow bursts on awakening.5 and quinolones, can cause neurotoxicity leading to con-
The pathology of uremic encephalopathy has been vulsive or nonconvulsive status epilepticus.5 Fluctua-
debated, but leading theories include the accumulation of tions in blood pressure can cause patients with CKD to
568 Kidney Transplantation: Principles and Practice

Fig. 33.1 Electroencephalogram (EEG) from a 70-year-old woman with a history of lung transplant and chronic kidney disease stage 3 who developed
altered mental status in the setting of pneumonia treated with cefepime. Encephalopathy improved with discontinuation of cefepime and change to
ceftazidime. EEG shows generalized sharp waves and diffuse slowing.

develop posterior reversible encephalopathy syndrome Atherosclerosis and thromboembolic disease are the
(PRES), which is associated with seizures in 60% to 75% most common causes of ischemic strokes in patients with
of patients.3 CKD, although hypotensive episodes during dialysis may
Patients with CKD who require antiepileptic drugs also cause watershed infarcts.3 Patients with CKD have
(AEDs) for seizure management may require renal dos- advanced carotid atherosclerosis compared with patients
ing of their medications. Renally cleared AEDs, such as without renal disease, and a component of atherosclerotic
levetiracetam, topiramate, or gabapentin, require supple- disease may be due to uremia itself.11 Anemia, which is a
mental dosing after dialysis sessions,3 and may require common sequela of CKD, is an independent risk factor for
lower doses in patients who do not require dialysis. Highly stroke3; the combination of CKD and anemia produces a
protein-bound AEDs, such as phenytoin, valproic acid, marked increase in stroke risk.12
and carbamazepine, are less susceptible to dialysis and The most likely etiology of hemorrhagic stroke in patients
are generally preferred in these patients,3 although dos- with CKD is hypertension, and the most likely locations
ing adjustments may still be necessary for renally excreted for hypertensive hemorrhages are the basal ganglia, cer-
metabolites. ebellum, and brainstem.3 Cerebral microhemorrhages are
observed on brain imaging of patients with CKD, and the
Pharmacologic Neurotoxicity extent of microhemorrhages inversely correlates with glo-
Patients with CKD are at increased risk of side effects and merular filtration rate (GFR).10 Uremia can lead to platelet
toxicity from medications. Cefepime, in particular, can dysfunction, which, along with anticoagulation used dur-
cause encephalopathy (Fig. 33.1) and myoclonus, and less ing hemodialysis, can contribute to risk of hemorrhagic
commonly seizures including nonconvulsive status epi- stroke.3,11 There is also an association between CKD and
lepticus.9 Cefepime and other medications must be renally retinal artery disease, with microvascular changes that are
adjusted to avoid neurotoxicity. independent of diabetes history.10
Cerebrovascular Disease Subdural Hematomas
Chronic kidney disease predisposes patients to both hemor- Patients undergoing both hemodialysis and peritoneal dial-
rhagic and ischemic stroke and progressive microvascular ysis are at increased risk of developing subdural hematomas,
disease,2,10 and CKD patients requiring dialysis have an which most commonly occur after trauma.3 This increased
even higher risk of stroke.10 Impaired regulation of cerebral risk may be due to uremia-related coagulation disturbances
blood flow is seen in patients with CKD and may contrib- or platelet dysfunction, use of anticoagulants during dialy-
ute to elevated risk of stroke and white matter injury.2 Out- sis, and use of rapid filtration and hypertonic dialysate.3,5
comes in patients with CKD who are hospitalized with acute Patients who sustain subdural hematomas or are thought
ischemic or hemorrhagic stroke are worse than in patients to be at especially high risk may benefit from heparin-free
without renal disease.10 dialysis and fall prevention education.3 Symptoms may be
33 • Neurologic Complications after Kidney Transplantation 569

focal, such as hemiparesis, or may consist of more general- excessive daytime sleepiness, obstructive sleep apnea (OSA),
ized symptoms such as encephalopathy and ataxia.4 and restless legs syndrome (RLS).3 Nightmares, sleepwalk-
ing, rapid eye movement (REM) behavior disorder, and nar-
Dialysis Disequilibrium Syndrome colepsy have also been reported.5
Dialysis disequilibrium syndrome describes a pattern Patients with CKD lack the evening melatonin surge that
of symptoms including headache, irritability, restless- controls the circadian sleep-wake cycle.19 Melatonin supple-
ness, blurred vision, nausea, muscle cramps, myoclonus, mentation, along with good sleep hygiene and sleep-focused
encephalopathy, and seizures, which develops toward cognitive behavioral therapy, may be helpful for patients with
the end of the dialysis session and typically resolves over insomnia.19 Acidemia increases ventilatory effort in patients
hours.3–5 Symptoms may include focal deficits, creating a with CKD and may contribute to OSA.5 Dialysis patients may
stroke-like presentation,13 and can last for days in severe be predisposed to RLS and periodic limb movements due to
presentations.3 The risk of developing dialysis disequilib- cerebral iron deficiency, accumulation of uremic toxins,
rium syndrome is highest with initiation of dialysis or in central dopamine deficiency, or uremic polyneuropathy.3,19
the setting of missing dialysis sessions.3 Dialysis disequilib- Cool dialysate, early morning dialysis sessions, short daily
rium syndrome has become uncommon with preventive hemodialysis, dopaminergic medications (levodopa, rop-
measures, including adding osmotically active solutes to inirole, pramipexole, and rotigotine), and gabapentinoids
dialysate, and should be treated as a diagnosis of exclusion, (gabapentin or pregabalin) can improve RLS symptoms, and
particularly when presenting with symptoms that are pro- transplantation can cause lasting relief.5,19
longed or focal.3,13
Several theories as to the etiology of dialysis disequilib- Wernicke’s Encephalopathy
rium syndrome have been posited. The reverse urea hypoth- Patients on hemodialysis or peritoneal dialysis are at risk
esis suggests that slower cerebral urea clearance causes of developing thiamine deficiency leading to Wernicke’s
osmotic shifts in the brain leading to cerebral edema.3,14 encephalopathy because of a combination of reduced oral
The idiogenic osmole hypothesis offers the idea that newly intake and increased loss of thiamine.5,7 Thiamine defi-
formed osmoles within the brain create an osmotic gradi- ciency is most likely to develop in patients on chronic dialy-
ent between the brain and the periphery.3,14 Another the- sis, but can also occur at the initiation of dialysis.3 Patients
ory proposes edema is caused by increased production of typically present with ataxia, ophthalmoplegia, and altered
organic acids, causing cortical intracellular acidosis.3 mental status or memory loss. Presentation can be atypical
in patients on chronic dialysis, and Wernicke’s encepha-
Hemodialysis-Related Headache lopathy may be underdiagnosed in this patient population.4
The International Headache Society defines hemodialysis-
related headaches as headaches that develop during at least PERIPHERAL NERVOUS SYSTEM
half of hemodialysis sessions, occur at least three times, and
resolve within 72 hours of dialysis.15 These headaches are Peripheral nervous system complications of CKD include
thought to be the result of water and electrolyte shifts during polyneuropathy, mononeuropathies, and uremic myopa-
dialysis,3 but hypoxemia occurring at the beginning of ses- thy. Mononeuropathies, especially median neuropathy, are
sions, changes in serotonin levels, hyponatremia, urea dis- common in patients with CKD, and may result from uremia.
turbances, or a mild dialysis disequilibrium syndrome have
also been proposed as etiologies.16 Hemodialysis-related Polyneuropathy
headaches typically resolve with kidney transplantation.3 Polyneuropathy occurs in 60% to 100% of patients with
end-stage renal disease.3 Neuropathy can stem from ure-
Dialysis Dementia mia alone, but it can also be attributed to diseases that cause
Dialysis dementia is a subacute neurodegenerative condition renal failure, such as diabetes mellitus and vasculitis.3
consisting of apathy, personality changes, dysarthria, dys- Uremic polyneuropathy can be painless or present with
phagia, ataxia, myoclonus, seizures, and dementia, progress- numbness, tingling, burning, or other abnormal sensations
ing to immobilization and mutism followed by death within in the distal extremities.3 There is a male predominance, and
6 months.3–5 This condition was more common before 1980, the course is variable.4 On examination, patients typically
when patients were dialyzed with an aluminum-containing have loss of pinprick, temperature, proprioception, and vibra-
dialysate used to bind dietary phosphate.3 Dialysis demen- tion sensation in the distal extremities, with lower extremi-
tia has become less common with the use of aluminum-free ties being affected before upper extremities, followed by loss
phosphate binders and low concentration of aluminum in of Achilles reflexes.3 Later, weakness and muscle wasting of
modern dialysate; however, dementia in dialysis patients, the lower before upper extremities occurs, and autonomic
particularly vascular dementia, still occurs at rates higher involvement has been reported.3 Uremic polyneuropathy
than age-matched healthy controls.3,10,17 Cognitive impair- is classically a length-dependent, distal, axonal, sensorimo-
ment can be seen in 50% to 87% of dialysis patients.18 Trans- tor, large-fiber neuropathy, but cases of pure motor and pure
plantation may improve memory and processing speeds, but sensory neuropathy have also been reported.3 Treatment
not attention and executive functioning.17 includes erythropoietin, biotin, pyridoxine, cobalamine, and
thiamine.5 Uremic neuropathy may stabilize or improve with
Sleep Disorders initiation of dialysis3 and typically resolves with transplan-
About 80% of patients on dialysis report sleep complaints, tation,4 but a few patients have reported worsening of their
and the most common sleep disorders include insomnia, neuropathy with initiation of dialysis.4
570 Kidney Transplantation: Principles and Practice

Mononeuropathies Central Nervous System


Isolated mononeuropathies can be an effect of uremia or Perioperative Vascular Insults. Patients with CKD are
may be due to increased susceptibility of patients with CKD at high risk of cerebrovascular disease, and this risk is still
to compression or ischemia of peripheral nerves.3 The most present posttransplantation.24 The perioperative state is
common nerves affected are the ulnar, median, and femo- a high-risk period due to volume and blood pressure shifts
ral nerves; optic, facial, trigeminal, and vestibulocochlear intraoperatively, but the mean timing of strokes is 49 months
nerves may also be affected.3 posttransplantation.24 Major surgery in the last 14 days is
Carpal tunnel syndrome is a compressive neuropathy a relative contraindication to thrombolytic administration
affecting the median nerve at the wrist. Symptoms include for acute stroke, but thrombolysis should be considered if
numbness, paresthesia, and pain in the thumb and first two strokes occur outside of this time period. Acute large vessel
or three fingers, followed by weakness and atrophy of the occlusions should be considered for mechanical thrombec-
thenar muscles in severe cases. Carpal tunnel syndrome tomy if patients present within the treatment window.26
can be confirmed by nerve conduction studies, electromy- Spinal cord infarctions are possible during renal transplant
ography (EMG), and ultrasound.20 Patients undergoing surgery. Normally, the caudal spinal cord is supplied by the
hemodialysis are at increased risk of developing carpal tun- intercostal arteries; however, a normal variant may occur
nel syndrome, especially in the upper extremity ipsilateral with branches of the internal iliac arteries supplying the cau-
to the arteriovenous fistula.20 The frequency of diagnosis dal spinal cord.4 When the iliac artery is manipulated in the
of carpal tunnel syndrome increases with the duration of setting of this variant, spinal cord ischemia can occur, causing
hemodialysis.21 Mechanisms include local deposition of conus medullaris syndrome, a constellation of lower extremity
amyloid, increased venous pressures distal to the fistula, pain and sensory loss, sphincter disturbance, and mixed upper
direct uremic toxic effect, and nerve ischemia. 20 and lower motor neuron signs.4 Caudal spinal cord ischemia
Ulnar neuropathy at the wrist due to uremic tumoral is irreversible and is therefore not a neurosurgical emergency.
calcinosis in Guyon canal results in weakness of the
intrinsic hand muscles and sensory loss in an ulnar nerve Postsurgical Encephalopathy. Patients with early graft
distribution.5 failure may have more difficulty clearing anesthetics or par-
The vestibulocochlear nerve can be affected by uremia, alytics and may present with encephalopathy or paralysis
leading to hearing loss and deafness; these symptoms can in the postoperative period due to medication effect.27 Peri-
reverse with dialysis or renal transplantation.4 operative stroke should be ruled out with head imaging,
Ischemic optic neuropathy can result in sudden vision especially if patients have focal neurologic deficits.
loss due to vascular compromise to the anterior or poste-
rior portion of the optic nerve.5,22 In anterior ischemic optic Uremic and Metabolic Encephalopathy. Patients who
neuropathy, funduscopic examination reveals pale optic undergo kidney transplantation are at high risk of metabolic
discs with blurred margins and retinal hemorrhage.5,22 derangements.24 Allograft dysfunction may cause recur-
Posterior ischemic optic neuropathy, on the other hand, rence of uremia, with patients developing encephalopathy,
initially has a normal funduscopic examination, but pallor seizures, myoclonus, and asterixis.24 Separate from uremia,
appears within 4 to 8 weeks.5,22 rejection encephalopathy is seen in transplant patients
with allograft dysfunction and is thought to be due to cyto-
Myopathy kine release.27 Hyponatremia and hypocalcemia may also
Uremic myopathy consists of proximal limb weakness, mus- develop and present with encephalopathy.27 Polypharmacy
cle wasting, easy fatigability, limited exercise tolerance, and and hospital delirium may also play a role in altered mental
pain.3,23 Up to 50% of patients on hemodialysis have some status in the immediate postoperative phase.27
functional alteration of skeletal muscle,23 and cardiomy-
opathy is sometimes present as well.5 Neurologic examina- Side Effects of Immunosuppressant Medications.
tion and serum creatine kinase are typically normal, but Immunosuppressant medications are used as antirejec-
a myopathic pattern may be revealed on EMG.4,23 Uremic tion agents after transplantation. Side effects of calcineu-
myopathy often worsens as renal failure progresses but rin inhibitors (CNI), such as tacrolimus and cyclosporine,
improves with renal transplantation.3,23 Possible mecha- include tremor, hyperreflexia, insomnia, headache, and
nisms include direct toxic effect from uremic toxins, altered mood disturbances; less commonly, akinetic mutism, hear-
vitamin D metabolism, carnitine deficiency, lactic acidosis, ing loss, optic neuropathy, pseudotumor cerebri, tumefac-
insulin resistance, ischemia, and malnutrition.3,23 tive demyelination, leukoencephalopathy, and seizures
can occur.28,29 CNI can also lead to hippocampal sclerosis
and development of refractory epilepsy.25 CNI, through an
Neurologic Complications After unknown pathophysiology, can cause a chronic pain syn-
Kidney Trans­plantation drome referred to as CNI-induced pain syndrome, or CIPS.24
Medication side effects and toxicity can occur at any time
ACUTE NEUROLOGIC COMPLICATIONS (LESS and at any drug level, including within the therapeutic
range.29 If side effects are addressed by dose adjustment or
THAN 1 MONTH)
discontinuation, immunosuppressants may be changed or
Neurologic symptoms may present quickly posttransplanta- restarted at a lower dose once symptoms improve.27
tion or may develop months or years later. In this section, we
will explore the central and peripheral nervous system com- Seizures. Seizures occur in 5% to 10% of transplant recipi-
plications that arise within the first month of transplantation. ents, most often in the immediate posttransplant period.
33 • Neurologic Complications after Kidney Transplantation 571

CNI side effect or toxicity is the most common cause of sei- Treatment involves aggressive blood pressure control,
zures, and CNI levels may be normal or elevated at time of removal of the offending agent when possible, and treat-
seizure.27 Hypomagnesemia due to cyclosporine-induced ment of seizures. In the posttransplant patient, leveti-
renal wasting can cause seizures, and other metabolic racetam and lacosamide are first-line agents, although
abnormalities such as hyponatremia, hypocalcemia, and intravenous lorazepam and fosphenytoin are favored in
hypoglycemia should also be included in the differential of a status epilepticus. Unprovoked seizures after resolution of
posttransplant patient who develops new onset seizures.25 PRES are uncommon but can occur, so AEDs can usually be
Seizures can be seen as part of PRES or can occur as a symp- weaned after resolution of symptoms and imaging findings,
tom of a central nervous system (CNS) infection or stroke. typically around 3 months after presentation.36 If PRES is
The aim of treating seizures in posttransplant patients is untreated, areas of ischemia can be seen on MRI, indicat-
seizure control without significant side effects or medica- ing irreversible damage and infarction. The syndrome can
tion interactions.25 Patients who present in status epilepti- progress to permanent injury or death.32 Clinical symp-
cus, with seizures lasting longer than 5 minutes or multiple toms, MRI findings, and the underlying pathology are often
seizures without return to baseline, should be treated with reversible and typically resolve in days to weeks.32 PRES is
intravenous lorazepam followed by intravenous fosphe- recurrent in 5% to 10% of cases.31
nytoin, although intravenous valproate and levetiracetam
are also supported by guidelines.30 Patients who develop Opportunistic Infections. Opportunistic infections are
recurrent seizures or who have a single seizure with an rare in the first 30 days posttransplantation; in this period,
epileptogenic focus on brain imaging or EEG should be infections are most likely due to donor-derived infections,
started on an AED.25 Seizures are typically provoked, and pretransplant colonization, or nosocomial infections24,25
therefore only short-term treatment (1–3 months) is gen- (see Chapter 31).
erally needed.25 Phenytoin, phenobarbital, and carbam- Donor-Derived Encephalitis. Fatal cases of encephali-
azepine induce cytochrome P450 enzymes and interact tis due to West Nile virus (WNV), rabies, and lymphocytic
with CNI and corticosteroids, so they are not typically rec- choriomeningitis virus (LCMV) have been seen in clusters
ommended for transplant patients, except for treatment of of patients who received infected transplants from the same
status epilepticus.25,27 Newer agents such as levetiracetam deceased donor.24 Encephalitis can be fatal if it is not detected
and lacosamide are preferred, because they can be loaded and treated quickly, and infectious signs and symptoms can
intravenously, have few medication interactions, and are be masked in patients who are immunosuppressed.
not highly protein bound.27 Gabapentin and pregabalin WNV encephalitis presents with fever and altered mental
are safe for use as adjunctive therapy in transplant patients status with or without focal neurologic deficits. WNV is typ-
with focal-onset seizures, but cannot be given intrave- ically transmitted through a mosquito vector. Around 75%
nously, are not approved for use in generalized seizures, of WNV infections in immunocompetent adults are asymp-
and are not highly effective first-line agents.25 tomatic, with <1% of infections resulting in encephalitis,
meningitis, or a polio-like acute flaccid paralysis.37 Donor-
Posterior Reversible Encephalopathy Syndrome. derived WNV is associated with a higher incidence of severe
PRES is commonly associated with CNI use and occurs neuroinvasive disease.37 Between 2002 and 2013, six clus-
most often in the first 3 months posttransplantation, but ters of WNV infections were reported to the Centers for Dis-
may present at any point.28 Risk factors include hyperten- ease Control and Prevention (CDC); among 16 transplant
sion, large blood pressure fluctuations, CNI administra- recipients, there were 12 WNV infections including nine
tion, vascular endothelial growth factor (VEGF) inhibitors cases of WNV encephalitis, which resulted in four deaths.38
such as bevacizumab, sepsis, and renal failure, so both Intravenous immunoglobulin (IVIG) may have some ben-
patients with CKD and patients who have received a kid- efit in treating WNV encephalitis.39
ney transplant are at risk.31 In patients who are treated Rabies virus is typically transmitted from an infected
with CNI, most cases of PRES occur within 2 weeks of animal to a person via bites or scratches and results in a
medication initiation or dose increase, but 20% of cases nonspecific prodrome of malaise, fever, and headache, and
present months to years after starting the offending progresses to confusion, followed by hydrophobia, coma,
agent.32 Patients treated with CNI who develop PRES can and death.38 In 2004 there were two clusters of donor-
have onset of symptoms at relatively low blood pressures, derived rabies infections, one in the US and one in Germany;
so PRES should not be ruled out because of normotension all of the transplant recipients died except for one patient
on presentation.32 who had previously been vaccinated against rabies.38 In
Patients with PRES typically present with hours to days 2013 a patient who had received a deceased donor kidney
of altered mental status, seizures, headache, and vision contracted donor-derived rabies after a 17-month incuba-
changes.31 Imaging reveals patchy or confluent cortical and tion period, but three patients who also received organs
subcortical vasogenic edema, most commonly in the bilateral from the same patient were treated with postexposure pro-
occipital lobes extending anteriorly to the parietal lobes in a phylaxis and did not develop rabies.40
gyriform pattern, but sometimes involving the frontal and LCMV infections are typically transmitted by rodents
temporal lobes as well25,32 (Fig. 33.2). PRES has been reported and are asymptomatic or result in a mild self-limited flu-
in the spinal cord33 and in the brainstem and cerebellum, like illness in immunocompetent patients; immunosup-
which can cause reversible obstructive hydrocephalus.34,35 pressed patients can develop severe meningoencephalitis.38
Etiology is thought to be secondary to autoregulatory failure, Between 2002 and 2013, there have been four reported
direct effect of cytokines, or compromised endothelial integrity, clusters of donor-derived LCMV meningoencephalitis in the
causing capillary leakage into the parenchyma.25,31,32 US and Australia, resulting in 12 deaths.38
572 Kidney Transplantation: Principles and Practice

Fig. 33.2 Fluid-attenuated inversion recovery (FLAIR) sequences of brain MRI from a 23-year-old woman with end-stage renal disease in the setting
of failed kidney transplant who presented with hypertension (blood pressure 242/90 mm Hg), headache, and altered mental status. T2 hyperintensi-
ties throughout the posterior white matter (top right) extending anteriorly (bottom left and right) and into the pons (top left) are typical for posterior
reversible encephalopathy syndrome (PRES).

Immune Reconstitution Inflammatory Syndrome. corticosteroids, are at high risk of developing polyneuropa-
Sudden reduction of immunosuppression can lead to a thy or myopathy, which both typically present with gen-
proinflammatory response that can cause deterioration of eralized weakness and difficulty weaning off ventilation.27
clinical status.29 Termed immune reconstitution inflammatory EMG reveals evidence of polyneuropathy and/or myopathy.
syndrome, or IRIS, this presentation was initially seen in the Patients with critical illness polyneuropathy or myopathy
HIV population once highly active antiretroviral therapy have longer hospital stays, longer utilization of ventilators,
(HAART) was introduced.29 IRIS is a diagnosis of exclusion and are less likely to be discharged home, although they do
and should be considered in a patient undergoing immuno- not have higher mortality rates.41
suppressant reduction for PRES or an opportunistic infection
who has new or worsening neurologic symptoms or signs, SUBACUTE NEUROLOGIC COMPLICATIONS
worsening neuroimaging findings, and a negative or improv- (1–6 MONTHS)
ing infectious workup.29 Treatment involves use of cortico-
steroids, and there is a high likelihood of allograft rejection.29 In this section, we will explore the central and peripheral
nervous system complications that arise 1 to 6 months
Peripheral Nervous System posttransplantation.
Perioperative Compressive Mononeuropathies. Intra-
operative compression by retractors can cause peripheral Central Nervous System
nerve injuries during renal transplantation, with the Opportunistic Infections. Because this time course
most common nerves affected being the femoral nerve includes the most intensive immunosuppression, opportu-
and the lateral femoral cutaneous nerve.4 Acute femoral nistic infections are most common between 1 and 6 months
neuropathy presents after 2% of renal transplants with posttransplantation.24
quadriceps weakness and pain or sensory deficit on the Cytomegalovirus. Cytomegalovirus (CMV) infection
thigh and lateral calf.5,7 Lumbosacral plexopathy can is the most common opportunistic infection after kidney
also be seen.24 Prognosis for recovery is generally good.4 transplantation.7 In the immunocompetent host, CMV is
asymptomatic, but in immunocompromised patients, it can
Critical Illness Polyneuropathy and Myopathy. present as meningitis, encephalitis, myelitis, and radiculi-
Patients who are critically ill, especially those receiving tis.7 Risk factors for CNS CMV infection include recurrent
33 • Neurologic Complications after Kidney Transplantation 573

CMV viremia, prolonged T cell depletion, and infection with cerebellar ataxia, and encephalopathy with MRI find-
ganciclovir-resistant strains.39 CMV encephalitis presents ings, including increased T2 signal intensity in the pons,
with acute onset of cognitive decline, visual disturbances, medulla, and cerebellum.45 Workup includes CSF poly-
and progressive encephalopathy.39 Brain MRI can reveal merase chain reaction (PCR) testing for HHV-6 DNA, as
evidence of viral ependymitis.39 Cerebrospinal fluid (CSF) CSF pleocytosis can be absent.27 HHV-6 can be treated
evaluation may be normal or consist of a lymphocytic pleo- with foscarnet, ganciclovir, or cidofovir and 25% to 40%
cytosis with elevated protein and normal glucose.39 Treat- of infected patients fully recover.39
ment comprises a combination of foscarnet, ganciclovir, Nocardia Species. Nocardia infection after solid-organ
and cidofovir, but mortality is high.39 transplantation occurs in up to 3.5% of patients and is
Varicella-Zoster Virus. Varicella-zoster virus (VZV) acquired via inhalation and dissemination to CNS, bone,
reactivation is common in immunosuppressed patients, and skin via the bloodstream.39 CNS is involved in up
occurring in 8.6% of patients who have received a solid- to one-third of patients with systemic infection.24 Brain
organ transplant.39 The most common manifestation is abscess is the most common CNS manifestation and pres-
herpes zoster, which presents as a vesicular rash in the dis- ents with focal neurologic symptoms, headaches, seizures,
tribution of one to three dermatomes, associated with severe and fevers.39 Biopsy and culture are sometimes necessary
radicular pain, sensitivity to touch, and unpleasant tingling for diagnosis.39 Treatment typically consists of a combi-
or itching.42 VZV can affect cranial nerves, causing cranial nation of multiple antibiotics such as trimethoprim-sulfa-
neuropathies.42 Treatment with famciclovir or valacyclovir methoxazole, imipenem, a third-generation cephalosporin,
is not required, except in the case of trigeminal zoster, but and amikacin, followed by narrowing of therapy based on
speeds healing of the rash.43 Postherpetic neuralgia occurs antibiotic sensitivities.39 Surgical drainage is sometimes
when pain along the affected dermatome persists for longer necessary.24
than 3 months and can be treated with gabapentin, pregab- Listeria Monocytogenes. Listeria monocytogenes is a food-
alin, tricyclic antidepressants, and topical lidocaine patches borne pathogen transmitted by ingestion of contaminated
or capsaicin cream.43 foods such as unpasteurized milk or undercooked meat, and
VZV vasculopathy is caused by infection of the cerebral, infections are rare in the posttransplant population, affect-
spinal cord, or temporal arteries, resulting in pathologic ing 0.2% of patients.39,46 Listeria can present as meningitis
vessel remodeling and leading to ischemic strokes or tempo- with fever, headache, and nuchal rigidity, or as encephalitis
ral arteritis.43 MRA reveals focal arterial stenosis and occlu- with fever, headache, seizures, and altered mental status.39
sion in the majority of patients.42 Patients are treated with Listeria rhombencephalitis presents with a prodrome similar
prednisone in addition to intravenous acyclovir.43 to that seen in meningitis, followed by cranial neuropathies,
VZV can reactivate in the meninges, brain paren- cerebellar signs, paralysis, sensory loss, and respiratory
chyma, and nerve roots, causing meningoencephalitis, failure; CSF evaluation may not readily identify Listeria.46
meningoradiculitis, and cerebellitis.43 VZV myelopathy Listeria abscesses can occur in the basal ganglia, thalamus,
occurs via direct infection of the spinal cord or indirectly medulla, and rarely in the spinal cord.39 Diagnosis is made
by inducing vasculopathy within the spinal cord arter- by positive CSF and/or blood cultures.39 Rhombencephalitis
ies.43 Patients present with weakness and sensory loss of and abscesses also show focal abnormalities on MRI imag-
lower and/or upper extremities, and MRI reveals longitu- ing.39,46 Empiric treatment with parenteral ampicillin or
dinal serpiginous enhancing lesions.43 In suspected cases penicillin is recommended, with gentamicin initially added
of meningoencephalitis, meningoradiculitis, cerebellitis, for synergy.39
or myelitis, lumbar puncture should be performed, and Mycobacterium Tuberculosis. Mycobacterium tubercu-
VZV deoxyribonucleic acid (DNA) or anti-VZV antibod- losis (TB) is rare in the posttransplant population but still
ies can be seen in the CSF.43 Intravenous acyclovir is the occurs much more frequently than in the general population;
treatment.43 infections generally represent reactivation of latent infection
Herpes Simplex Virus. Herpes simplex virus (HSV) and typically occur in the first year posttransplant.39 Most
encephalitis is rare after transplantation and presents with posttransplant TB infections do not affect the CNS, with only
altered mental status, fevers, headache, and seizures.39 EEG 3% of infections occurring as meningitis and 1% occurring
is more likely to show periodic discharges and focal slowing as abscesses or tuberculomas.39 TB meningitis presents with
in the frontotemporal and occipital areas than in patients fever and altered mental status more often than neck stiffness,
with encephalitis due to other causes.44 CSF reveals lym- and some patients can present with hydrocephalus or cranial
phocytic pleocytosis and HSV DNA.39 Treatment consists of neuropathies as well.24,39 MRI typically shows basilar men-
intravenous acyclovir.39 ingeal enhancement, and CSF findings include lymphocytic
Human Herpesvirus 6. Human herpesvirus 6 (HHV-6) pleocytosis, elevated protein, and low glucose.39 Tuberculo-
is a beta herpesvirus that most people contract early in mas can be found throughout the brain and spinal cord and
life.39 Reactivation in the setting of immunosuppression present with fever, altered mental status, headache, seizures,
can cause limbic encephalitis, which presents with sei- and focal deficits.39 MRI reveals ring-enhancing lesions with
zures, hallucinations, altered mental status, and amne- surrounding edema. Treatment for both TB meningitis and
sia.27,39 Neuroimaging reveals characteristic mesial tuberculomas uses a four-drug therapy including isoniazid,
temporal lobe involvement.24 HHV-6 can also manifest as rifabutin, ethambutol, and pyrazinamide for 9 to 12 months,
myelitis, which presents with lower extremity weakness with adjunctive dexamethasone for meningitis and reduc-
and/or numbness and signs of spinal cord inflammation tion of immunosuppression.39 Mortality is 40% or higher.39
on cord imaging.39 There have been a few cases of rhomb- Candida Albicans. Candidal infections tend to occur in
encephalitis which presents with cranial neuropathies, the first 3 to 6 months posttransplantation, and are the
574 Kidney Transplantation: Principles and Practice

most common invasive fungal infections in renal trans- rejection or infection with CMV.24 GBS classically pres-
plant patients.47 Candidemia can cause CNS involvement ents with ascending weakness and hyporeflexia or are-
through hematogenous spread and typically presents with flexia, but a pure sensory variant, Miller-Fisher syndrome,
multiple small abscesses at the gray-white junction or in and a pharyngeal-cervical-brachial variant also can be
regions supplied by perforating arteries.47 seen.48 CSF evaluation reveals albuminocytologic dis-
Aspergillus Species. Aspergillus is acquired through inha- sociation. Nerve conduction study typically reveals evi-
lation, and the CNS is involved most commonly via hematog- dence of demyelinating polyneuropathy with early loss
enous spread and infrequently by direct extension through of H-reflexes and F-waves but can be challenging in the
the cribriform plate.39 The CNS is the most common target setting of preexisting neuropathy. In immunosuppressed
of hematogenous spread, and CNS aspergillosis is seen in patients, GBS can be associated with CMV infection in the
50% of invasive aspergillosis infections.39 Patients present first 6 months posttransplant, and these patients should be
with fever, altered mental status, seizures, strokes, and focal treated with intravenous ganciclovir in addition to IVIG.49
deficits, and symptoms can progress rapidly.39 MRI reveals
ring-enhancing or hemorrhagic lesions.39 CSF evaluation Side Effects of Immunosuppressant Medications. In
typically reveals elevated protein, but fungal cultures are addition to the CNS side effects listed previously, peripheral
often negative; serum or CSF galactomannan antigen testing nervous system side effects of CNIs include chronic inflam-
or DNA PCR can be helpful in these cases, and biopsy is some- matory demyelinating polyneuropathy (CIDP) and brachial
times necessary.39 Treatment includes voriconazole and plexopathy.28 Polymyositis has also been seen as a result of
reduction of immunosuppression as first-line therapy and tacrolimus treatment.27
liposomal amphotericin B, posaconazole, and itraconazole
as second-line treatments.39 Mortality approaches 100%, CHRONIC NEUROLOGIC COMPLICATIONS
although recent reports have indicated good outcomes with (MORE THAN 6 MONTHS)
use of voriconazole.39
Mucor Species. Mucor species are transmitted via inhala- In this section, we will explore the central and periph-
tion of fungal spores and can result in invasive infections.39 eral nervous system complications that arise more than 6
Invasive mucormycosis is more commonly seen in trans- months after transplantation.
plant patients who have a history of diabetes or renal fail-
ure, although it remains rare, affecting 2% of solid-organ Opportunistic Infections
transplant recipients.39,47 Sinus infections with Mucor spe- After the first 6 months posttransplantation, opportunistic
cies can directly extend through the cribriform plate into infections tend to include community-acquired infections
the frontal lobe, orbits, and optic nerves, presenting with that affect immunosuppressed patients more severely than
headaches, fever, facial pain, sinus congestion, orbital the general population.24
swelling, vision loss, proptosis, and ophthalmoparesis.39,47
Cavernous sinus thrombosis can be seen, and strokes and Cryptococcus Neoformans. Cryptococcal infections are a late
seizures may result.39 Mucormycosis should be suspected if complication of transplantation, occurring 1.5 to 5 years
a black mucosal or cutaneous eschar is observed, and diag- after transplantation.47 Cryptococcal meningitis occurs in
nosis is made with endoscopic sinus evaluation and culture 60% of solid-organ transplant recipients with cryptococ-
of infected tissue.39,47 Treatment involves emergent surgi- cosis and in 2.8% of solid-organ transplant patients over-
cal debridement, liposomal amphotericin B, and reversal of all.28,39 It typically presents with headache, fevers, night
immunosuppression, but mortality is nearly universal.39 sweats, weight loss, nausea, vomiting, and altered mental
Toxoplasmosis. Toxoplasmosis is a protozoal infection status; cranial neuropathies can be present as well.39 Neck
caused by reactivation of latent infection or as a primary stiffness is often absent, and symptoms can be subacute to
infection after ingesting contaminated foods.24,39 CNS toxo- chronic.24,27 Some patients with advanced disease may
plasmosis presents as meningoencephalitis, myelitis, or ring- present with intracerebral cryptococcomas, so head imag-
enhancing discrete parenchymal lesions, typically in the first ing should be performed before lumbar puncture.47 Lum-
3 months after transplant.39 A patient with toxoplasmosis bar puncture typically shows elevated opening pressure,
may present with fevers, altered mental status, seizures, or elevated protein, low glucose, and pleocytosis, which may
focal neurologic deficits.39 Brain MRI shows multiple ring- be neutrophilic or lymphocytic.39 CSF cryptococcal antigen
enhancing lesions with a predilection for the basal ganglia assay is sensitive and specific.47 Patients are treated with
and cerebrum, which may have surrounding edema or associ- at least 2 weeks of induction therapy involving liposomal
ated hemorrhage.39 Toxoplasma IgG or DNA may be present amphotericin B and flucytosine, followed by consolidation
in the CSF, but histopathology is the gold standard for diag- and maintenance therapy with fluconazole.39 Classically,
nosis.39 Empiric treatment with sulfadiazine, pyrimethamine, patients with cryptococcal meningitis have very high intra-
and leucovorin should be started based on clinical suspicion, cranial pressures, requiring serial high-volume lumbar
seropositivity, and imaging findings, and patients will need punctures and sometimes ventriculoperitoneal shunts.39
chronic suppressive therapy after completion of initial treat- Mortality is 30% to 50%.39
ment.39 Mortality of CNS toxoplasmosis is around 13%.24 Cryptococcomas are diagnosed with neuroimaging and
CSF studies and typically require longer duration of induc-
Peripheral Nervous System tion, consolidation, maintenance therapies, and corticoste-
Guillain-Barre Syndrome. Guillain-Barre syndrome roids for edema causing mass effect.39 Surgical intervention
(GBS) is an uncommon complication after kidney trans- is recommended for cryptococcomas larger than 3 cm or
plantation, but can occur in the setting of allograft those causing significant mass effect.39
33 • Neurologic Complications after Kidney Transplantation 575

Endemic Fungal Infections. Coccidioides and Histoplasma 56%.50 Treatment involves reducing immunosuppression.
can rarely cause meningitis in the immunosuppressed No other treatment has been efficacious, although case
patient, typically as a late posttransplant complication.47 reports exist of treatment with mirtazapine and cidofovir.50
Patients in locations where these fungal infections are Survivors of PML often have significant neurologic sequelae,
common present with fever, headache, and altered men- although there are case reports of patients recovering with
tal status; CSF reveals a lymphocytic pleocytosis.47 Coc- little or no neurologic deficits after early withdrawal of
cidioides, Histoplasma, and Blastomyces can uncommonly immunosuppression.51,52
cause abscesses as well.47 The presence of Coccidioides Epstein-Barr Virus. Epstein-Barr virus (EBV) reacti-
antibodies in the CSF is diagnostic, and Histoplasma diag- vation is most commonly associated with development
nosis typically requires urine antigen or serum antibody of posttransplantation lymphoproliferative disorder,
testing.47 described in detail later. Rarely, EBV can cause enceph-
Progressive Multifocal Leukoencephalopathy. Immu- alitis, which can present months to several years after
nosuppressed patients are at risk of developing progressive transplantation.53 CSF analysis may reveal a lymphocytic
multifocal leukoencephalopathy (PML), a CNS demyelinat- pleocytosis with EBV DNA.39 Treatment includes foscar-
ing disease caused by reactivation of the John Cunningham net, ganciclovir, or cidofovir along with discontinuation
(JC) virus.50 The median time to developing PML in transplant of immunosuppression.39 In a case report, EBV encepha-
patients is 17 months but ranges from less than 1 month to litis was followed by parkinsonism and basal ganglia
more than 20 years.50 PML has been associated with many necrosis.54
different immunosuppressant or immunomodulatory agents,
with prednisone, cyclosporine, rituximab, and azathioprine Posttransplant Lymphoproliferative Disease
being the most common in transplantation.50 The most Posttransplant lymphoproliferative disease (PTLD) is a
common symptoms of PML are cognitive complaints, weak- hematologic malignancy that usually presents late after
ness, cerebellar symptoms, personality change, and seizures; transplantation, and can occur more than 10 years post-
although dizziness, hallucinations, falls, lethargy, and head- transplantation.24,55 PTLD occurs in about 2% of renal
aches are also seen.50 Neuroimaging reveals multiple or con- transplant patients.56 The primary locations involved are
fluent white matter lesions without contrast enhancement24 the abdomen, thorax, or allograft, with CNS involvement
(Fig. 33.3). The mortality rate in transplant patients with in a minority of affected patients29; however, kidney trans-
PML is around 84%, with 1-year survival rates around plant patients are at highest risk of developing primary

Fig. 33.3 Fluid-attenuated inversion recovery (FLAIR) (left) and postcontrast T1 sequences (left) sequences of brain MRI from a 65-year-old man on
hydroxychloroquine for dermatologic sarcoidosis who presented with 6 months of progressive face, upper extremity, and lower extremity weakness
and cognitive dysfunction. Brain biopsy was consistent with progressive multifocal leukoencephalopathy (PML). Large confluent subcortical T2 hyperin-
tensities are seen in bilateral frontoparietal regions (left). These lesions are T1 hypointense and do not enhance with gadolinium administration (right).
576 Kidney Transplantation: Principles and Practice

CNS PTLD.24 Immunosuppression compromises the func- 9. Fugate JE, Kalimullah EA, Clark SL, Wijdicks EF, Rabinstein AA.
tion of T cells, which can lead to unregulated proliferation Cefepime neurotoxicity in the intensive care unit: a cause of severe,
underappreciated encephalopathy. Crit Care 2013;17(6):R264.
of B cells, especially B cells that have been transformed by 10. Toyoda K. Cerebral small vessel disease and chronic kidney disease.
EBV.55 The majority of PTLD is B cell lymphoma, with only J Stroke 2015;17(1):31–7.
about 5% being T cell or T/NK lymphoma.55 The risk of 11. Seliger SL, Gillen DL, Longstreth WT, Kestenbaum B, Stehman-Breen
developing PTLD increases when an EBV-negative patient CO. Elevated risk of stroke among patients with end-stage renal dis-
ease. Kidney Int 2003;64(2):603–9.
receives an EBV-positive donor organ, and certain HLA 12. Abramson JL, Jurkovitz CT, Vaccarino V, Weintraub WS, McCelllan W.
and cytokine gene polymorphisms can predispose patients Chronic kidney disease, anemia, and incident stroke in a middle-
to developing PTLD.55 CNS PTLD should be suspected in aged, community-based population: the ARIC Study. Kidney Int
a patient receiving immunosuppression for renal trans- 2003;64(2):610–5.
plant who presents with subacute focal neurologic deficits 13. Attur RP, Kandavar R, Kadavigere R, Baig WW. Dialysis disequilib-
rium syndrome presenting as a focal neurological deficit. Hemodial
and brain imaging showing areas of vasogenic edema and Int 2008;12(3):313–5.
ring-enhancing lesions on computed tomography (CT) or 14. Silver SM, Sterns RH, Halperin ML. Brain swelling after dialysis: old
MRI; lumbar puncture showing elevated nucleated cells urea or new osmoles? Am J Kidney Dis 1996;28(1):1–13.
with a lymphocytic predominance and elevated protein 15. Antoniazzi AL, Corrado AP. Dialysis headache. Curr Pain Headache
Rep 2007;11(4):297–303.
should also raise concern, and flow cytometry may reveal 16. Antoniazzi AL, Bigal ME, Bordini CA, Speciali JG. Headache associated
a monoclonal population of B or less commonly T cells.56 with dialysis: the International Headache Society criteria revisited.
Treatment typically consists of reduction of immunosup- Cephalalgia 2003;23(2):146–9.
pression with cytotoxic chemotherapy; adding rituximab 17. Dixon BS, VanBuren JM, Rodrigue JR, Lockridge RS, Lindsay R, Chan C,
and/or high-dose methotrexate has been shown to increase et al. Cognitive changes associated with switching to frequent noctur-
nal hemodialysis or renal transplantation. BMC Nephrol 2016;17:12.
progression-free survival.55,57 Outcomes are typically poor 18. Gupta A, Mahnken JD, Johnson DK, et al. Prevalence and correlates
in CNS PTLD.57 of cognitive impairment in kidney transplant recipients. BMC Nephrol
2017;18(1):158.
Cognitive Impairment 19. Scherer JS, Combs SA, Brennan F. Sleep disorders, restless legs
syndrome, and uremic pruritis: diagnosis and treatment of com-
As described previously, dialysis patients have a high rate mon symptoms in dialysis patients. Am J Kidney Dis 2017;69(1):
of cognitive impairment, which tends to improve after 117–28.
transplantation. However, cognitive impairment is not 20. Gousheh J, Iranpour A. Association between carpel tunnel syndrome
entirely reversible and is associated with increased mortal- and arteriovenous fistula in hemodialysis patients. Plast Reconstr
ity in posttransplant patients.18 A contributing factor to Surg 2005;116(2):508–13.
21. Gejyo F, Narita I. Current clinical and pathogenic understanding of
the increased mortality may be a higher rate of medication B2-m amyloidosis in long-term haemodialysis patients. Nephrology
nonadherence in transplant patients with cognitive impair- (Carlton). 2003;8(Suppl):S45–9.
ment and dementia.18 22. Buono LM, Foroozan R, Savino PJ, Danesh-Meyer HV, Stanescu D.
Posterior ischemic optic neuropathy after hemodialysis. Ophthalmol-
ogy 2003;110(6):1216–8.
23. Campistol JM. Uremic myopathy. Kidney Int 2002;62(5):1901–13.
Summary 24. Avila JD, Zivkovic S. The neurology of solid organ transplantation.
Curr Neurol Neurosci Rep 2015;15(7):38.
Neurologic complications occur both before and after kid- 25. Shepard PW, St Louis EK. Seizure treatment in transplant patients.
ney transplantation, and the diagnosis is often dependent Curr Treat Options Neurol 2012;14(4):332–47.
26. Molina CA, Chamorro A, Rovira A, et al. REVASCAT: a randomized
on determination of the timing of onset and localization of trial of revascularization with SOLITAIRE FR device vs. best medical
signs and symptoms. Neurologic complications after kidney therapy in the treatment of acute stroke due to anterior circulation
transplantation are varied and typically represent a unique large vessel occlusion presenting within eight-hours of symptom
challenge to both the nephrologist and neurologist alike. onset. Int J Stroke 2015;10(4):619–26.
27. Dhar R. Neurologic complications of transplantation. Neurocrit Care
Patients often require cooperation from multiple teams to 2017;141:545–72.
ensure the best outcomes. 28. Pruitt AA. Neurologic complications of transplantation. Continuum
(Minneap Minn) 2017;23(3):802–21.
References 29. Pruitt AA, Graus F, Rosenfeld MR. Neurological complications of solid
1. Potluri K, Holt D, Hou S. Neurologic complications in renal transplan- organ transplantation. Neurohospitalist 2013;3(3):152–66.
tation. Handb Clin Neurol 2014;121:1245–55. 30. Glauser T, Shinnar S, Gloss D, et al. Evidence-based guideline: treat-
2. Tamura MK, Pajewski NM, Bryan RN, et al. Chronic kidney disease, ment of convulsive status epilepticus in children and adults: report of
cerebral blood flow, and white matter volume in hypertensive adults. the guideline committee of the American Epilepsy Society. Epilepsy
Neurology 2016;86(13):1208–16. Curr 2016;16(1):48–61.
3. Hocker SE. Renal disease and neurology. Continuum (Minneap Minn) 31. Fugate JE, Rabinstein AA. Posterior reversible encephalopathy syn-
2017;23(3):722–43. drome: clinical and radiological manifestations, pathophysiology, and
4. Burn DJ, Bates D. Neurology and the kidney. J Neurol Neurosurg Psy- outstanding questions. Lancet Neurol 2015;14(9):914–25.
chiatry 1998;65(6):810–21. 32. Burnett MM, Hess CP, Roberts JP, Bass NM, Douglas VC, Joseohson
5. Lacerda G, Krummel T, Hirsch E. Neurologic presentations of renal SA. Presentation of reversible posterior leukoencephalopathy syn-
diseases. Neurol Clin 2010;28(1):45–59. drome in patients on calcineurin inhibitors. Clin Neurol Neurosurg
6. Nongnuch A, Panorchan K, Davenport A. Brain-kidney crosstalk. Crit 2010;112(10):886–91.
Care 2014;18(3):225. 33. de Havenon A, Joos Z, Longenecker L, Shah L, Ansari S, Digre K. Pos-
7. Brouns R, De Deyn PP. Neurological complications in renal failure: a terior reversible encephalopathy syndrome with spinal cord involve-
review. Clin Neurol Neurosurg 2004;107(1):1–16. ment. Neurology 2014;83(22):2002–6.
8. Yoon JE, Kim JS, Park JH, Lee KB, Roh H, Park ST. Uremic parkinson- 34. Kumar A, Keyrouz SG, Willie JT, Dhar R. Reversible obstructive
ism with atypical phenotypes and radiologic features. Metab Brain Dis hydrocephalus from hypertensive encephalopathy. Neurocrit Care
2016;31(2):481–4. 2012;16(3):433–9.
33 • Neurologic Complications after Kidney Transplantation 577

35. Grossbach AJ, Abel TJ, Hodis B, Wassef SN, Greenlee JDW. Hyper- 47. Anesi JA, Baddley JW. Approach to the solid organ transplant
tensive posterior reversible encephalopathy syndrome causing pos- patient with suspected fungal infection. Infect Dis Clin North Am
terior fossa edema and hydrocephalus. J Clin Neurosci 2014;21(2): 2016;30(1):277–96.
207–11. 48. Dimachkie MM, Barohn RJ. Guillain-Barré syndrome and variants.
36. Datar S, Singh T, Rabinstein AA, Fugate JE, Hocker S. Long-term risk Neurol Clin 2013;31(2):491–510.
of seizures and epilepsy in patients with posterior reversible encepha- 49. Shaban E, Gohh R, Knoll BM. Late-onset cytomegalovirus infec-
lopathy syndrome. Epilepsia 2015;56(4):564–8. tion complicated by Guillain-Barre syndrome in a kidney trans-
37. Winston DJ, Vikram HR, Rabe IB, et al. Donor-derived West Nile virus plant recipient: case report and review of the literature. Infection
infection in solid organ transplant recipients: report of four additional 2016;44(2):255–8.
cases and review of clinical, diagnostic, and therapeutic features. 50. Mateen FJ, Muralidharan R, Carone M, et al. Progressive multifocal
Transplantation 2014;97(9):881–9. leukoencephalopathy in transplant recipients. Ann Neurol 2011;
38. Basavaraju SV, Kuehnert MJ, Zaki SR, Sejvar JJ. Encephalitis caused 70(2):305–22.
by pathogens transmitted through organ transplants, United States, 51. Ohara H, Kataoka H, Nakamichi K, Saijo M, Ueno S. Favorable out-
2002–2013. Emerg Infect Dis 2014;20(9):1443–51. come after withdrawal of immunosuppressant therapy in progressive
39. Cohen BA, Stosor V. Opportunistic infections of the central ner- multifocal leukoencephalopathy after renal transplantation: case
vous system in the transplant patient. Curr Neurol Neurosci Rep report and literature review. J Neurol Sci 2014;341(1–2):144–6.
2013;13(9):376. 52. Sauer R, Golitz P, Jacobi J, Schwab S, Linker RA, Lee DH. Good
40. Vora NM, Orciari LA, Niezgoda M, et al. Clinical management and outcome of brain stem progressive multifocal leukoencepha-
humoral immune responses to rabies post-exposure prophylaxis lopathy in an immunosuppressed renal transplant patient:
among three patients who received solid organs from a donor with importance of early detection and rapid immune reconstitution.
rabies. Transpl Infect Dis 2015;17(3):389–95. J Neurol Sci 2017;375:76–9.
41. Kelmenson DA, Held N, Allen RR, Quan D, Burnham EL, Clark BJ. 53. Lau JSY, Low ZM, Abbott I, et al. Epstein-Barr virus encephalitis in
Outcomes of ICU patients with a discharge diagnosis of critical illness solid organ transplantation. New Microbiol 2017;40(3):212–7.
polyneuromyopathy: a propensity-matched analysis. Crit Care Med 54. Espay AJ, Henderson KK. Postencephalitic parkinsonism and basal
2017;45(12):2055–60. ganglia necrosis due to Epstein-Barr virus infection. Neurology 2011;
42. Nagel MA, Gilden D. Complications of Varicella zoster virus reactiva- 76(17):1529–30.
tion. Curr Treat Options Neurol 2013;15(4):439–53. 55. Jagadeesh D, Woda BA, Draper J, Evens AM. Post transplant lympho-
43. Nagel MA, Gilden D. Neurological complications of Varicella zoster proliferative disorders: risk, classification, and therapeutic recommen-
virus reactivation. Curr Opin Neurol 2014;27(3):356–60. dations. Curr Treat Options Oncol 2012;13(1):122–36.
44. Sutter R, Kaplan PW, Cervenka MC, et al. Electroencephalography 56. Honig LS. Subacute imbalance in a renal transplant patient. JAMA
for diagnosis and prognosis of acute encephalitis. Clin Neurophysiol Neurol 2015;72(11):1367–8.
2015;126(8):1524–31. 57. Evens AM, Choquet S, Kross-Desosiers AR, et al. Primary CNS post-
45. Jubelt B, Mihai C, Li TM, Veerapeneni P. Rhombencephalitis/brain- transplant lymphoproliferative disease (PTLD): an international report
stem encephalitis. Curr Neurol Neurosci Rep 2011;11(6):543–52. of 84 cases in the modern era. Am J Transplant 2013;13(6):1512–22.
46. Mathews BK, Chuang C, Rawal A, Gertner E. Listeria rhomboencepha-
litis in a patient on a tumor necrosis factor alpha inhibitor (etaner-
cept). J Clin Rheumatol 2014;20(6):325–7.
34 Cutaneous Disease in Kidney
Transplantation Patients
ANNE LOUISE MARANO, JOANNA HOOTEN, and
SARAH A. MYERS

CHAPTER OUTLINE Introduction Corticosteroids


Cutaneous Malignancy in Renal Azathioprine
Transplant Patients Cyclosporine
Nonmelanoma Skin Cancer Mycophenolate Mofetil
Epidemiology and Pathogenesis Tacrolimus
Melanoma Sirolimus and Everolimus
Epidemiology and Pathogenesis Premalignant and Benign Cutaneous
Merkel Cell Carcinoma Tumors in Renal Transplant Patients
Epidemiology and Pathogenesis Actinic Keratosis
Rare Cutaneous Malignancies Seborrheic Keratosis
Atypical Fibroxanthoma and Porokeratosis
Undifferentiated Pleomorphic Sarcoma Infections and Inflammatory Cutaneous
Kaposi Sarcoma Findings in OTRs
Immunosuppressive Drug Considerations Cutaneous Infections
Sirolimus and Everolimus Viral Infections
Voriconazole Fungal Infections
Conclusion on Cutaneous Malignancy in Common Inflammatory Disorders
OTRs Conclusion
Immunosuppressive Drug Cutaneous Side
Effects

Introduction skin cancer is vital to the care of these patients for targeted
screening and prevention. The International Transplant
Organ transplant recipients (OTRs), which include renal Skin Cancer Collaborative (ITSCC) and the Transplant Skin
transplant patients, are significantly affected by cutaneous Cancer Network (TSCN) are valuable resources for further
disease as a result of a prolonged immunosuppressed state. information on this topic.2
Renal transplant recipients have a 5-year survival rate In addition, OTRs often demonstrate increased rates of
of 85%.1 Therefore with promising odds for survival, the premalignant and benign cutaneous tumors, and infec-
chronic nature of skin disease may become challenging to tious and inflammatory cutaneous conditions. These will
manage for these patients. Because the skin is visible, cuta- be discussed in this chapter; however, the main focus of this
neous disease may significantly affect patient quality of life. chapter is to inform the transplant physician on the press-
Renal transplantation is also unique in that the return to ing matter of cutaneous malignancy in OTRs.
hemodialysis is an option, so the risks and benefits of side
effects of transplantation and the immunosuppression regi-
men must be heavily weighed if the skin cancer burden is
extreme or life threatening.
Cutaneous Malignancy in Renal
In terms of cutaneous disease, the most significant and Transplant Patients
burdensome complication of organ transplantation is skin
cancer.1 The transplant physician should be aware of the NONMELANOMA SKIN CANCER
importance of evaluation for cutaneous malignancy in the
pretransplant and posttransplant periods. This chapter will Epidemiology and Pathogenesis
discuss the approach to OTRs with cutaneous malignancy. OTRs are at an increased risk of malignancy compared
In these patients, the transplant physician and derma- with the general population because of immunosuppres-
tologist should work closely together for the benefit of the sion. Skin cancer is the most frequently reported posttrans-
patient. Understanding the risk factors in posttransplant plant malignancy. In particular, nonmelanoma skin cancer
578
34 • Cutaneous Disease in Kidney Transplantation Patients 579

TABLE 34.1 Risk Factors for Development of Skin accumulate and lead to carcinogenesis.4 This is supported
Cancer in Organ Transplant Patients by the fact that NMSC in OTRs is associated with latitude
and that Australian OTRs have a higher risk of NMSC than
Fitzpatrick skin type I to III patients in England.4 Ultraviolet light causes a charac-
Increasing age at transplantation
Duration and level of immunosuppression teristic type of mutation, resulting in cytosine to thymine
Type of organ transplant (heart/lung > kidney > liver) transitions through the formation of cyclobutane dimers,
Previous transplant which are commonly found in NMSC p53 mutations.4 The
Squamous cell carcinoma before transplant immune system normally functions to provide surveil-
History of lymphoma pre-/posttransplant
Pretransplant end organ disease (e.g., rheumatoid arthritis, systemic
lance and eliminate precancer.4 This is evidenced by the
lupus erythematosus, or autoimmune hepatitis) effectiveness of immunomodulatory medications, such as
Liver transplant recipients with psoriasis on previous biologic therapy/ imiquimod, in treating premalignant actinic keratosis. In
psoralen plus ultraviolet A light phototherapy addition, the immunosuppressive medications themselves
may be directly carcinogenic, the most prominent exam-
From Zwald F, Brown M. Skin cancer in solid organ transplant recipients:
advances in therapy and management: Part 1. Epidemiology of skin cancer ple of which is azathioprine.4 Cyclosporine may also be
in solid organ transplant recipients. J Am Acad Dermatol 2011;65:253–61. carcinogenic.4
More recently, the role of the human papillomavirus
(HPV) infection in OTRs in NMSC pathogenesis has become
(NMSC) represents 95% of posttransplant skin cancer.1 The more widely appreciated.5 OTRs have greater quantities of
incidence of posttransplant skin cancer has been reported at HPV deoxyribonucleic acid (DNA) found in SCC samples.1
1427 per 100,000 person-years.2 Interestingly, in the gen- HPV viral proteins E6 and E7 are oncoproteins involved
eral population, basal cell carcinoma (BCC) is the most com- in regulating cell cycle checkpoints.6 This may also be
mon type of NMSC and squamous cell carcinoma (SCC) is observed clinically in the warty appearance of many SCC in
the second most common type of NMSC, whereas the ratio OTRs. Of consequence, a recent study found that NMSC in
in the OTR population is reversed. Organ transplantation nonwhite OTRs was more likely to occur in nonsun-exposed
increases the risk of cutaneous SCC by 65-fold and BCC by areas, in particular, on the genitals.7 This is suggestive of
10-fold.1 Furthermore, SCC is more likely to be aggressive the role of sexually transmitted HPV in NMSC in nonwhite
in OTRs. Specifically, in the general population, the risk OTRs.7 The value of pretransplant or posttransplant HPV
of metastasis of SCC is 0.5% but increases to about 8% for vaccination for preventing NMSC has yet to be studied and
OTRs.1 warrants further investigation.8
The factors associated with risk of NMSC posttrans- In summary, NMSC incidence is increased in OTRs
plant include the following: Fitzpatrick skin type I or II because of immunosuppression, ultraviolet carcinogenesis,
(fair skin), increasing age at transplantation, duration drug carcinogenicity, and possibly because of HPV infec-
and level of immunosuppression, type of organ transplant tion, particularly in nonwhite OTRs.
(heart and lung > kidney > liver), previous transplant, his-
tory of SCC pretransplant, history of lymphoma, pretrans- Clinical Presentation. SCCs may vary in clinical appear-
plant end-organ disease (rheumatoid arthritis, systemic ance, but as previously noted, they are the most prevalent
lupus erythematosus, autoimmune hepatitis), and liver skin cancer in OTRs; therefore any suspicious growing
transplant recipients with a history of psoriasis on previ- lesion should be biopsied. In general, both BCC and SCC
ous biologic therapy or psoralen plus ultraviolent A light commonly arise on sun-exposed areas of the body. SCCs are
(Table 34.1). To simplify risk stratification, a recent study often erythematous, keratotic plaques that may also ulcer-
from the TSCN from 26 centers with more than 10,000 ate (Fig. 34.1). The subtype of keratoacanthoma is a cra-
patients identified the following statistically significant teriform appearing SCC that is rapidly growing. SCCs are
risk factors for posttransplant skin cancer: pretransplant often tender. BCCs are classically described as “pink, pearly
skin cancer, male sex, white race, and age at transplant papules” but may also become ulcerated.
50 years or older.2 Field cancerization is a term that describes a heavy bur-
An additional predictive index was developed for a com- den of cancer in an area or “field” of skin. It commonly
prehensive and cost effective targeted surveillance strategy refers to extensive areas of sun damage with multiple kera-
for skin cancer in renal transplant patients so that those at totic neoplasms including actinic keratosis, SCC in situ, and
high risk can be easily recognized early on posttransplant.3 SCC. Both “transplant hands” (Fig. 34.2) and “transplant
Patient age, outdoor exposure, ultraviolet light exposure, scalp” are common descriptors of field cancerization in the
pretransplant skin cancer, childhood sunburn, and skin OTR population.
type were selected as predictors.3 A score >7 was designated In general, physical examination should be performed
as high risk for NMSC within 2 years, and of these patients, of the draining lymph node basins in transplant patients
the chance of being SCC-free at 2 years was 43%, and at 5 with a high-risk SCC. SCCs can be aggressive, metastasizing
years was 17%. In contrast, with a score <4, 99% were free to the lymph nodes, and can cause death. The risk of local
from SCC at 6 months, 95% at 2 years, and 89% at 5 years.3 recurrence of high-risk SCC is 13.4%, usually within the
The direct association of duration and degree of immu- first 6 months after excision.9 The risk of metastasis of SCC
nosuppression with the risk of NMSC in OTRs suggests in the general population is 0.5 to 5%. The risk increases to
that immunosuppression is key to the pathogenesis of 8% for OTRs.10 This is a poor prognostic sign, with a 3-year
NMSC in OTRs. The pathogenesis of NMSC is multifacto- overall survival for OTRs with metastatic SCC of 48%.4 In
rial. Ultraviolet light causes genetic mutations, in particu- contrast, BCCs rarely metastasize but can be locally aggres-
lar in the p53 gene in SCC, and these mutations eventually sive and cause physical destruction.
580 Kidney Transplantation: Principles and Practice

TABLE 34.2 High-Risk Features of Squamous Cell


Carcinoma in Organ Transplant Patients
Large size (>2 cm)
Multiple squamous cell carcinomas
High-risk location (ear, lips, over parotid gland, scalp, or temple)
In-transit metastatic lesion
Recurrence
Histology
Poorly differentiated
Perineural invasion
Deep invasion

From Zwald F, Brown M. Skin cancer in solid organ transplant recipients:


advances in therapy and management: Part 2. Management of skin cancer
in solid organ transplant recipients. J Am Acad Dermatol 2011;65:263–79.

also involve chemoprevention with medication, which


will be discussed later. The treatment of NMSC in OTRs is
often complicated in that the risks and benefits of various
medical and surgical procedures must be weighed. Sur-
veillance is particularly important for SCC, which has a
higher rate of metastasis in OTRs compared with the gen-
eral population.10
Fig. 34.1 Squamous cell carcinoma on the left temple.
Prevention. Chemoprevention may be used in OTRs with
numerous NMSCs. The options are discussed later. Reti-
noid derivatives, such as acitretin, have been shown to be
effective in the suppression of SCC development, includ-
ing reduction in actinic keratoses and SCCs.12 Indications
for the initiation of systemic retinoids in organ transplant
patients are the following: development of multiple SCCs
per year (5–10/year); development of multiple SCCs in
high-risk locations (head/neck); patients with a history of
lymphoma/leukemia and SCCs; a single SCC with high met-
astatic risk; metastatic SCC; explosive SCC development;
and eruptive keratoacanthomas.13 Low-dose acitretin (10
mg) therapy should be started to minimize side effects with
slow titration by 10 mg increments at 2- to 4-week inter-
vals to the target dose of 20 to 25 mg daily. Side effects are
dose related, with dry eyes and mouth being the most com-
mon; dry skin and pruritus are less common. Acitretin is a
known teratogen (pregnancy category X) and its use should
be carefully considered in childbearing women who wish
to conceive within 3 years. Severe hyperlipidemia that is
refractory to standard treatment is a contraindication. Lab-
oratory monitoring must be performed frequently, includ-
ing a baseline pregnancy test, complete blood count, fasting
lipid panel, liver panel, and serum creatinine. Chemopre-
Fig. 34.2 Field cancerization on the hands, also known as transplant vention with oral retinoids is a lifelong treatment. When
hands. the medication is discontinued, a rebound effect occurs that
is difficult to control. Patients can experience the eruption
of multiple aggressive SCCs over a relatively short period
In SCC, high-risk features include the following: large size of time. Thus reduction of the dosage of acitretin is usually
>2 cm, high-risk location (ear, lip, scalp, temple), tumor successful at maintaining patient compliance while still
depth >2 mm, recurrent, poor differentiation, perineural benefiting from chemoprevention.14
invasion, and lymphovascular invasion1,11 (Table 34.2). Capecitabine, a prodrug of 5-deoxy-5-fluorouridine, is
metabolized by the liver to 5-FU and is less commonly used
Management. The management of NMSC in OTRs for NMSC chemoprevention. Initially used for the treatment
involves prevention, treatment, and surveillance. Of these, of metastatic breast and colon cancer, the use of systemic
prevention is of the utmost importance and should begin 5-FU has been shown to be beneficial in reducing the devel-
in the pretransplant evaluation. Prevention also includes opment of precancerous and cancerous lesions in organ
continued education on sun protective measures and on transplant patients.14 In a retrospective review performed
self-skin examination. In severe cases, prevention may at the University of Minnesota,15 organ transplant patients
34 • Cutaneous Disease in Kidney Transplantation Patients 581

who were given low-dose capecitabine had lower rates of Wide local excision or Mohs surgery are the preferred
developing NMSC and actinic keratoses compared with the modalities for treatment of infiltrative BCC and SCC.4,14
period before capecitabine treatment. However, grade 3 Wide local excision is often performed with 4-mm margins
and 4 toxicities were common, causing discontinuation.15 for NMSC and a surgical pathologist subsequently evalu-
Prospective studies are needed to determine the long-term ates all of the tissue margins postoperatively. Mohs surgery
efficacy and safety of lower doses. entails removing one thin layer of tissue at a time; each layer
More recently, a randomized controlled trial demon- is evaluated through frozen section histology for the pres-
strated efficacy of nicotinamide in preventing NMSC.16 This ence of remaining tumor. If the circumferential and deep
offers an appealing alternative to acitretin or capecitabine margins are clear, the surgery is ended. If not, another layer
because of a limited side effect profile for nicotinamide. This is removed from the margin where tumor was noted, and
study was not performed in OTRs but included patients with the procedure is repeated until all margins of the final tissue
at least two NMSC in the previous 5 years. Patients received sample are clear of cancer. This offers better margin control,
nicotinamide 500 mg twice daily or placebo for 12 months. minimal tissue defect, and has higher cure rates than wide
At 12 months, the rate of new NMSC was lowered by 23% local excision. Mohs surgery is preferred for NMSC on the
in the nicotinamide group compared with the rate of new face, large lesions, and recurrent NMSC.4,14
NMSC in the placebo group.16 The efficacy of nicotinamide The most concerning outcome in SCC in OTRs is metas-
in reducing NMSC in OTRs requires further study. tasis, either to lymph nodes or systemically because of the
Overall, patient education on sun protective behaviors worsened prognosis, as mentioned earlier. The role of imag-
and early signs of skin cancer is very important in the ing in cutaneous SCC is somewhat controversial. Patients
OTR population. Specifically, patients should be taught with palpable lymph nodes should undergo sentinel lymph
to use daily broad-spectrum sunscreen, protective cloth- node biopsy and dissection.4,14 Otherwise, a sentinel lymph
ing, and avoidance of sun tanning and tanning beds. A node biopsy should be considered for OTR patients with
helpful resource for patients and clinicians is the ITSCC high-risk SCC or lymph nodes found on imaging performed
patient education brochure “After Transplantation - in the appropriate patient. A recent consensus article sug-
Reduce Incidence of Skin Cancer” (AT-RISC Alliance: gested consideration of imaging in patients with possible
http://at-risc.org/). bony invasion, possible orbital invasion, for assessment of
extent of tumor invasion in soft tissue, for staging evalua-
Treatment. Topical therapies are used for precancer- tion in high-risk SCC, for evaluation of potential perineu-
ous actinic keratosis, BCC, and SCC. In particular, topical ral spread, and for postoperative surveillance for recurrent
therapies may be appropriate for the superficial variant of disease.11
BCC and SCC in-situ. 5-fluorouracil 5% cream is a topical For metastatic SCC, medical, surgical, and radiation ther-
chemotherapy cream and imiquimod is a topical immuno- apies should be combined.4,14 Currently, there are limited
modulatory agent that targets toll-like receptor 7. These medical therapies for metastatic SCC and most of these have
are effective for the treatment of actinic keratosis, super- been studied in immunocompetent patients. Radiotherapy
ficial BCC, and SCC in-situ and for chemoprevention.14 may also be used adjunctively.4,14 The most important
These therapies cause an inflammatory and crusted reac- medical intervention is to decrease immunosuppression
tion, which may be intolerable to patients. Some patients, if possible. Chemotherapy traditionally included cispla-
however, prefer this treatment to surgery because it does tin and 5-flourouracil. More recently, epidermal growth
not require a doctor’s visit and is often nonscarring. Many factor receptor inhibitors such as gefitinib, erlotinib, and
male transplant patients with hair loss benefit from regu- cetuximab have been used to treat metastatic SCC.4,14,17,18
lar use of 5-flourouracil cream on the scalp to prevent SCC Furthermore, programmed cell death receptor 1 (PD1) or
formation.4 its ligand (PDL1) inhibitors such as pembrolizumab and
Photodynamic therapy (PDT) is another treatment nivolumab have been used in single patients with meta-
modality for actinic keratosis and superficial NMSC, partic- static SCC but there have been no randomized controlled
ularly if there is a large area such as the scalp involved. PDT studies.19 Pharmaceutical companies Regeneron and
involves the use of an exogenously administered precursor Sanofi are planning trials for cemiplimab for cutaneous
of photosensitizer protoporphyrin IX synthesis (usually SCC.20 Importantly, the effect of using an immune check-
either aminolevulinic acid or methyl aminolevulinate) that point inhibitor on a transplant patient’s graft function is
is activated by light, producing reactive oxygen species and unknown and may be problematic.
destroying tumor cells. PDT has been effective in the treat- For metastatic BCC in OTRs, there is a Food and Drug
ment of actinic keratosis and superficial NMSC in OTRs.4,14 Administration–approved medication, vismodegib. Vis-
PDT must be administered under physician supervision in a modegib is a smoothened inhibitor in the sonic hedgehog
clinic setting. Common side effects are pain and erythema. pathway.21 Efficacy is approximately 30% with many sys-
The simplest surgical procedure for treatment of NMSC temic side effects including electrolyte abnormalities, alope-
is electrodessication and curettage.4,14 This is similar to a cia, taste loss, and muscle cramps.21 Additionally, a recent
biopsy but with the addition of three passes of electrodessi- study showed an increased risk of SCC with vismodegib
cation and curettage to the base of the lesion. This is a sim- treatment, which may be of particular concern in OTRs
ple procedure that may be performed at the time of biopsy, with NMSC.22 Interestingly, a subsequent study did not
which is preferable for many OTRs with frequent doctors’ show an increased risk of SCC in patients on vismodegib.23
visits. The downsides are a slightly lower cure rate than The use of vismodegib and risk of SCC remains unclear and
excision and often a large circular hypopigmented scar in requires further study and careful application in the trans-
contrast to a linear scar from an excision. plant population.
582 Kidney Transplantation: Principles and Practice

Surveillance. Patients with a history of NMSC and organ melanoma, and Merkel cell carcinoma.24 As stated previ-
transplantation should be followed-up with every 3 to 6 ously, one of the major risk factors for posttransplant skin
months, and examination should include a lymph node cancer is pretransplant skin cancer. For renal transplant
examination for high-risk SCC (Table 34.3). For patients patients with a history of pretransplant SCC, there is a
with high-risk SCC with concerning features on follow-up 40% to 80% chance of posttransplant SCC.24 The ITSCC
physical examination (i.e., lymphadenopathy, neurologic guidelines are based on the principle that a patient should
signs) should undergo a positron emission tomography have at least a 5-year survival from malignancy of 60%
(PET)/computed tomography (CT) for evaluation for recur- to ethically transplant an organ.24 For SCC, the consensus
rent disease.11 The frequency of long-term follow-up may panel recommends the following: no delay in transplanta-
be stratified by risk3 (see Table 34.3). tion for stage T1 and T2b, a 2-year delay after clearance of
tumor for high-risk SCC (large size >2 cm, high-risk loca-
Pretransplant Delay. From the perspective of the tion [ear, lip, scalp, temple], tumor depth >2 mm, recur-
transplant physician, a previous diagnosis of a cutane- rent, poor differentiation), and a 2- to 3-year delay after
ous malignancy may affect the decision to proceed with clearance for tumor for high-risk SCC with perineural
solid-organ transplantation. A consensus guideline from invasion24 (Table 34.4).
the ITSCC was developed for recommendations regard-
ing solid-organ transplantation with a pretransplant SCC, MELANOMA
Epidemiology and Pathogenesis
TABLE 34.3 Follow-Up Intervals for Total Body Skin
Examination for Solid-Organ Transplantation Melanoma is an immunogenic cutaneous malignancy
with higher risk of metastasis, and its incidence continues
Interval for Total Body Skin to increase. The relationship between melanoma risk and
Patient Risk Factor Examination (No. of Months) organ transplantation is less clear than that for NMSC.
No skin cancer/field disease 12 The transplant physician should be aware of a history of
Field disease 3–6 pretransplant melanoma and screen for melanoma in the
One nonmelanoma skin cancer 3–6 posttransplant period.25 For melanoma, there are reports
Multiple nonmelanoma skin cancers 3
High-risk squamous cell carcinoma 3 ranging from a 3- to 5-fold increased risk in OTRs com-
or melanoma pared with the general population.1,26 In renal transplant
Metastatic squamous cell carcinoma 1–3 recipients, a large study found a 3.6-fold increased risk of
or melanoma melanoma in renal transplant recipients.27 African Ameri-
From Zwald F, Brown M. Skin cancer in solid organ transplant recipients:
can OTRs have a 1.72-fold increased risk for melanoma.1
advances in therapy and management: Part 2. Management of skin cancer The mean duration from transplantation to posttransplant
in solid organ transplant recipients. J Am Acad Dermatol 2011;65:263–79. melanoma is 5 years.1

TABLE 34.4 Recommended Wait Times Pretransplantation for Patients With a History of Skin Cancer Before Transplantation
Wait Time Before
Skin Malignancy Appropriate Treatment Pretransplantation Transplantation After Treatment
SQUAMOUS CELL CARCINOMA
No history of SCC but high risk for SCC Treatment of field disease No delay
Low-risk SCC Surgical excision with clear margins of Mohs micrographic No delay
surgery
High-risk SCCa other than Surgical excision with clear margins of Mohs micrographic 2 years
perineural invasion surgery
High-risk SCCa with perineural invasion or ≥2 Surgical excision with clear margins of Mohs micrographic 2–3 years
high-risk features surgery +/− radiotherapy
High-risk SCC with local nodal metastasis Surgical excision with clear margins of Mohs micrographic 5 years
surgery + radiotherapy
Distant metastasis Refer for oncology opinion Not eligible for transplantation
MERKEL CELL CARCINOMA
Local with negative sentinel lymph node biopsy Wide local excision +/− radiotherapy 2 years
Local with nodal metastasis Wide local excision, lymph node dissection + radiotherapy 3–5 years
Distant metastasis Refer for oncology opinion Not eligible for transplantation
MELANOMA
In situ melanoma Wide local excision No delay, follow-up posttransplan-
tation 3 months
Stage Ib melanoma Wide local excision 2 years
Stage Ib/IIa melanoma Wide local excision +/− sentinel lymph node biopsy 2–5 years
Stage IIb/IIc melanoma Wide local excision + sentinel lymph node biopsy 5 years
Stage III or IV melanoma Refer for oncology opinion Not eligible for transplantation
alargesize >2 cm, high-risk location (ear, lip, scalp, temple), tumor depth <2 mm, recurrence, poor differentiation, perineural invasion.
From Zwald F, Leitenberger J, Zeitouni N, et al. Recommendations for solid organ transplantation for transplant candidates with a pretransplant diagnosis of
cutaneous squamous cell carcinoma, Merkel cell carcinoma and melanoma: a consensus opinion from the International Transplant Skin Cancer Collaborative
(ITSCC). Am J Transplant 2016;16(2):407–13.
34 • Cutaneous Disease in Kidney Transplantation Patients 583

Risk factors for developing melanoma specifically in renal For metastatic melanoma, several emerging therapies
transplant patients have recently been outlined in a cohort are rapidly advancing the field. BRAF inhibitors such as
study of a large national data registry and include older age, vemurafenib and dabrafenib are available for metastatic
male sex, recipient white race, less than four human leuko- melanoma.34 In addition, immunotherapies such as ipilim-
cyte antigens (HLA) mismatches, living donors, and siroli- umab and PD-1 inhibitors are being widely used in mela-
mus and cyclosporine therapy.28 Transplant clinicians can noma for the possibility of a durable response.35 The effects
identify these risks factors and close skin surveillance should of these therapies on graft function are unknown. In life-
be provided for patients with higher risk characteristics. threatening melanoma, immunosuppression changes and/
Immunosuppression also appears to deleteriously affect or reduction should also be considered.
the course of melanoma in transplant patients. In the larg-
est study of melanoma in the OTR population, patients who Surveillance. Patients with a history of melanoma and
developed melanoma demonstrated worse overall survival organ transplantation should be followed-up with every
compared with expected survival in control subjects.29 In 3 months, and examination should include a lymph node
addition, patients with thicker melanomas appear to have examination and examination for in-transit metastases (see
increased risk of dying of metastatic melanoma.29 Large Table 34.3).
multicenter studies are needed to provide better under-
standing of the role of immunosuppression on the behavior Pretransplant Delay. As mentioned in the NMSC section,
of melanoma in OTRs. a consensus guideline was developed for management of
With the advent of immunotherapy in treating melanoma, the pretransplant SCC, melanoma, and Merkel cell carci-
it is known that the immune system plays a role in mela- noma.24 For melanoma, case control studies have shown
noma pathogenesis. The less defined relationship between that the immunosuppression has a differential effect on
melanoma and OTR is likely due to the multifactorial nature melanoma in the context of transplantation, such that a
of melanoma pathogenesis. Melanoma risk is also increased thin melanoma (Breslow depth 1.5–2 mm) may behave
with ultraviolet light exposure, in particular with tanning bed comparably to melanoma in immunocompetent individu-
use, and genetic factors such as mutations in the p16 gene.1,26 als, whereas thick melanomas (Breslow depth >2 mm) may
An illustrative example of the role of immunity in melanoma behave more aggressively and be associated with increased
pathogenesis is the case report of a renal transplant patient mortality risk.24 Patients with melanoma in-situ, stage
who developed metastatic melanoma from a donor kidney.30 Ia, Ib, IIa, IIb, and IIIa are considered organ transplant
The donor did not have clinical symptoms of metastatic mela- candidates, but stages IIc, IIIb, IIIc, and IV should not be
noma, suggesting that the donor immune system was keep- considered organ transplant candidates.24 The consensus
ing the melanoma in check. However, when the transplant guideline recommends the following wait time for pretrans-
allograft was placed in a host on immunosuppressive medica- plant melanoma: no delay for melanoma in-situ, a 2-year
tion, metastatic melanoma developed.30 Therefore melanoma delay after wide local excision for stage Ia melanoma, a
risk is likely related to immunosuppression but appears to be 2-year delay after wide local excision +/− sentinel lymph
less clearly linked than NMSC is to immunosuppression. node biopsy for stage Ib/IIa melanoma, a 2- to 5-year delay
after wide local excision +/− sentinel lymph node biopsy for
Clinical Presentation. In general, melanomas are pig- stage IIb/IIc melanoma, and that stage III and IV melanoma
mented lesions, with the exception of the rare variant of patients are not candidates for organ transplantation24 (see
amelanotic melanoma. The subtypes of melanoma are the Table 34.4).
following: superficial spreading, lentigo maligna, nodular,
and acral lentiginous. The “ABCDE” acronym summarizes MERKEL CELL CARCINOMA
the clinical features of melanoma: asymmetry, border irreg-
ularity, color variation, diameter more than 6 mm, and Epidemiology and Pathogenesis
evolution.31 Loss of color should also raise suspicion. Any Merkel cell carcinoma (MCC) is a rare cutaneous malig-
suspicious lesion should be biopsied, and treatment is based nancy of neuroendocrine origin, but is particularly aggres-
on the pathology (Breslow depth ± ulceration or mitoses) sive and concerning in organ transplant patients. Therefore
and lymph node involvement of melanoma.31 Therefore a reduction of immunosuppression may be recommended in
lymph node examination for the draining lymph nodes in OTRs who develop MCC. There is a 10-fold increased risk
an OTR with melanoma is imperative.31 of MCC in OTRs compared with the general population.1,36
MCC tends to develop 7 to 8 years posttransplant.1,36 The
Management. Melanoma should be managed surgically pathogenesis of MCC involves ultraviolet light, demon-
with wide local excision with consideration of a sentinel strated by the increased incidence of MCC on the head and
lymph node biopsy based on the severity of the tumor. The neck, and also infection with the human polyoma virus
following surgical margins are appropriate: 5 mm for mela- in the majority but not all patients. Logically, the more
noma in-situ, 1 cm for melanoma <1 mm Breslow depth, aggressive nature of MCC in OTRs makes sense given its fre-
and 2 cm for melanoma >2 mm Breslow depth. Sentinel quent viral association. Overall, MCC in OTRs metastasize
lymph node biopsy should be considered for melanomas to lymph nodes in 68% of cases and cause death in 56% of
with Breslow depth >1 mm and some argue >0.75 mm.32 cases.1,36
A large prospective trial showed there was no survival
benefit to complete lymph node dissection in node-positive Clinical Presentation. MCCs do not have a distinct
melanoma, and this may eventually change the surgical appearance and are often biopsied to rule out NMSC. Many
management of these patients, particularly in the era of of these lesions present on the head and neck. Lesions pres-
improved medical therapies.33 ent as dome-shaped pinkish-red to bluish-brown papules or
584 Kidney Transplantation: Principles and Practice

nodules and are frequently ulcerated. A full skin examina-


tion for satellite lesions, a sign of in-transit spread, and a
Immunosuppressive Drug
lymph node examination of draining lymph node basins are Considerations
necessary.
The degree of immunosuppression, but also the specific
Management and Treatment. All MCCs require surgical medications used, affect the risk of cutaneous malignancy
treatment with wide local excision or Mohs surgery.14,37 in OTRs. In general, it is preferable to minimize the number
Given the aggressive nature of MCC, all MCC in OTRs need of immunosuppressive drugs in patients with a high cuta-
a sentinel lymph node biopsy performed.14,37 Management neous malignancy disease burden or a high-risk cutaneous
includes surgery with a wide local excision with at least tumor. Several studies have shown that patients receiving
2.5- to 3-cm margins or Mohs surgery and radiotherapy triple immunosuppression (cyclosporine, prednisone, aza-
to the site and regional lymph nodes.14,37 Immunosup- thioprine or sirolimus) were at increased risk for skin cancer
pression may need to be reduced with MCC given the high development compared with those on dual therapy (predni-
malignancy mortality rate of over 50%.14,37 sone and azathioprine or sirolimus).1 Both azathioprine and
For patients with advanced MCC, treatment is challeng- cyclosporine have been shown to be carcinogenic.1,4 There-
ing. A recent phase II study demonstrated that for 26 patients fore maintenance therapy with tacrolimus over cyclosporine
with unresectable MCC, pembrolizumab (anti-PD1) ther- is recommended as first-line calcineurin inhibitor in combi-
apy was used and resulted in a 56% objective response rate, nation with mycophenolate mofetil rather than azathioprine
with four patients with a complete response.38 Responses as an antiproliferative agent. Azathioprine has mutagenic
were seen in virus-positive and virus-negative tumors.38 and photosensitizing properties and sensitizes to DNA dam-
These patients were not OTRs. Nevertheless, this may be age by ultraviolet A via its metabolite 6-thioguanine.1,4
a promising new therapy for advanced MCC in which few Cyclosporine is also thought to be carcinogenic independent
treatment options exist. of immunosuppression but the mechanism is unclear.1,4
Details of cutaneous side effects of specific transplant
Surveillance. Patients with a MCC and organ transplan- medications will be discussed in the drug side effect section.
tation should be followed-up with every 3 months and
examination should include a lymph node examination SIROLIMUS AND EVEROLIMUS
and examination for in-transit metastases (see Table 34.3).
Emerging evidence suggests that sirolimus is the preferred
Pretransplant Delay. As mentioned in the NMSC and immunosuppressive agent used in OTRs with high cuta-
melanoma sections, a consensus guideline was developed neous malignancy burden.1 The concern with sirolimus is
for management of the pretransplant SCC, melanoma, and that it may affect wound healing and may not be a potent
MCC.24 For MCC, the consensus guidelines recommend the enough immunosuppressant early posttransplant.1 A ret-
following: at least a 2-year delay for patients with stage IIb rospective study showed an 11.6% reduction in skin cancer
or less MCC (local disease, tumor size <2 cm, negative senti- in the sirolimus-treated versus nonsirolimus-treated group
nel lymph node biopsy), and that patients with stage III and overall. There was no difference in mortality or rejection.41
higher (regional lymph node disease or metastasis) are not The cumulative incidence of skin cancer at 1, 3, and 5 years
organ transplantation candidates24 (see Table 34.4). after the index posttransplant cancer were 9.3%, 20.6%,
and 24.7%, respectively in the sirolimus-treated group, ver-
RARE CUTANEOUS MALIGNANCIES sus 17.7%, 31%, and 35.8%, respectively in the nonsiroli-
mus-treated group, thus demonstrating a lower risk for skin
Atypical Fibroxanthoma and Undifferentiated cancer with sirolimus treatment.41 In renal transplant spe-
Pleomorphic Sarcoma cifically, a retrospective study showed that, compared with
Atypical fibroxanthoma (AFX) is a rare spindle cell neo- mycophenolate and standard cyclosporine, everolimus
plasm that presents as a solitary pink to red nodule, usually with reduced-dose cyclosporine resulted in hazard ratios
arising on the head and neck, often biopsied to evaluate for of 0.28, 0.39, and 0.41, respectively for NMSC, nonskin
NMSC. AFX is considered to be an indeterminate lesion, a cancer, and any cancer.42 There were no associations with
less aggressive superficial variant of undifferentiated pleo- rejection, graft loss, or death.42 These data require further
morphic sarcoma. In OTRs, both AFX and undifferentiated prospective study. Additionally, there was a retrospective
pleomorphic sarcoma have a higher rate of local recurrence study of SCC in OTRs that did not find statistical significance
and metastasis than in the general population.39 Treatment for decreased SCC risk in patients taking sirolimus.43
should be aggressive, with Mohs surgery or wide local exci-
sion with 2-cm margins, followed by adjuvant radiation VORICONAZOLE
therapy.39
Voriconazole is a triazole antifungal medication approved
Kaposi Sarcoma for the treatment of aspergillosis in transplant patients and is
Classically, Kaposi sarcoma (KS) is associated with HIV/ most often used in lung transplant patients. One of the well-
AIDS but may present similarly in immunosuppressed OTR established side effects of voriconazole is photosensitivity.
patients. KS in OTRs is very rare.40 KS presents with viola- Voriconazole-induced photosensitivity results in a sunburn-
ceous plaques, most often on the extremities. KS is mediated like erythema on sun-exposed sites.44 Voriconazole-induced
by the herpesvirus 8.40 KS may be managed with destruc- photosensitivity accelerates the risk of skin cancer post-
tion of lesions, chemotherapy, radiation therapy, but most transplantation, but the mechanism is not understood and
importantly in OTRs, by reducing immunosuppression.40 may involve the interaction of voriconazole metabolites
34 • Cutaneous Disease in Kidney Transplantation Patients 585

with ultraviolet light.45 A large retrospective study found


an increase in cutaneous SCC in lung transplant patients
treated with voriconazole with an absolute risk at 5 years
after transplant of 28%.46 An additional large retrospective
study found that voriconazole exposure was associated with
73% increased risk of cutaneous SCC with each additional
30-day exposure at the standard dose, thereby increasing
the risk by 3%.47 The duration of voriconazole exposure has
been shown to be an independent risk factor for skin cancer
in lung transplant patients.48 This is an important consid-
eration because prolonged therapy with voriconazole may
be necessary given the chronic nature of invasive fungal
infections.45 In addition, there is concern that voriconazole-
associated cutaneous SCC in an immunosuppressed patient
may be especially aggressive, and there are reports of patients
dying from cutaneous SCC metastatic disease.49 Therefore it Fig. 34.3 Steroid acne with monomorphic inflamed lesions and few
is important to note voriconazole exposure as a risk factor for comedones.
developing SCC in transplant patients.
is responsible for the appearance of hirsutism and steroid
acne. Steroid acne resembles acne vulgaris, affecting only
Conclusion on Cutaneous androgen-dependent areas of skin bearing sebaceous glands
Malignancy in OTRs (i.e., face, chest, back, and upper arms), but with predomi-
nant monomorphic papulopustules and a paucity of open
Based upon the information just discussed, the importance of comedones (blackheads) (Fig. 34.3). Steroid-induced acne
the cooperation between a transplant physician and a derma- is dose-dependent. Treatment consists of decreasing the
tologist in the management of solid-organ transplant recipi- glucocorticoid dose and a trial of topical therapies including
ents should be evident. Even in the pretransplant period, a benzoyl peroxide, erythromycin, and clindamycin.52 Oral
dermatologist may serve a role in assessing posttransplant therapies, including doxycycline and other oral antibiotics
risk for patients with a history of high-risk cutaneous malig- similarly used for acne, spironolactone in nonchildbearing
nancies.24 A recent study suggested that patient education, women, and isotretinoin, may be required for more resis-
understanding, and compliance with photoprotective mea- tant cases.53
sures were significantly lower in patients who had never
attended a specialty OTR dermatology clinic compared
AZATHIOPRINE
with patients being treated in a specialty OTR dermatology
clinic.50 The treatment of OTRs with skin cancer is complex Cutaneous side effects of azathioprine are rarely reported.
and requires longer appointment times, more frequent fol- Hair loss has been reported in 108 of 200 (54%) transplant
low-up, and a substantial amount of dermatologic surgical recipients examined, and it was concluded that diffuse alo-
procedures. Patients should be educated about this process. pecia was due to azathioprine in several of these cases.51 In
Understandably, patients often become overwhelmed by the addition, changes in hair color and texture were seen in 22%
numerous dermatologic surgical procedures. Among derma- of the study population (attributed to azathioprine-induced
tologists, this is often referred to as “Mohs fatigue,” in which diffuse alopecia).51 An erythema nodosum hypersensitivity
patients may often ask for less-invasive procedures such as reaction has also been reported, but in patients receiving
electrodessication and curettage, even with the understand- azathioprine for inflammatory bowel disease, which may
ing that these modalities are less effective, to avoid another have been a result of the underlying illness rather than a
invasive procedure. Therefore in OTRs with cutaneous direct effect of azathioprine.54
malignancy, a specialty OTR dermatology clinic can be espe-
cially beneficial to prevent, diagnose, and manage the bur-
CYCLOSPORINE
den of skin oncology in this patient population.50
Cyclosporine has largely been replaced by more effective
and safer immunosuppressant medications. Furthermore,
Immunosuppressive Drug the doses used today are lower than those historically used.
These trends have decreased the frequency of cutaneous
Cutaneous Side Effects adverse effects, which tend to be dose-dependent. The most
common cyclosporine-induced mucocutaneous effects
CORTICOSTEROIDS
are hypertrichosis (affecting up to 100% of those taking
Most transplant immunosuppression regimens include this drug) and gingival hyperplasia (affecting 2%–81%).55
corticosteroids, often early in the posttransplant course. Hypertrichosis does not appear to be an androgen-mediated
Cutaneous effects of corticosteroid use are well recognized side effect because cyclosporine-induced hypertrichosis is
and include cushingoid effects: redistribution of body fat, not confined to androgen-dependent areas of skin and is
purpura, striae, telangiectasia, and thinning of the skin.51 independent of sex hormone levels.55 Hypertrichosis is more
Corticosteroids stimulate the pilosebaceous unit, possi- common the longer that transplant recipients are exposed
bly through an androgen-mediated mechanism, and this to cyclosporine.55 Gingival hyperplasia is more common
586 Kidney Transplantation: Principles and Practice

with increasing time posttransplantation and younger


transplant recipients exposed to cyclosporine.56 In addi-
tion, pilosebaceous lesions are also a common development
as cyclosporine may be partially eliminated through seba-
ceous glands.57 Clinical lesions include sebaceous hyperpla-
sia, epidermal cysts, keratosis pilaris, and more nonspecific
follicular eruptions.58

MYCOPHENOLATE MOFETIL
Mycophenolate mofetil seems to have a low incidence of
skin side effects, with fewer side effects documented com-
pared with azathioprine.59 The most frequent side effect
related to mycophenolate mofetil is aphthous stomatitis,
which can present more severely in patients who are on
mycophenolate mofetil and mammalian target of rapamy- Fig. 34.4 Seborrheic keratoses on the trunk of a male patient, illus-
cin (mTOR) inhibitors (sirolimus, everolimus).60 There is trating the number and variation in shape, size, and color that may be
increased susceptibility to herpes simplex and zoster and observed in these lesions.
cytomegalovirus (CMV) infections.59
(see Fig. 34.2). AK results in mild dysplasia in the epidermis
on histopathology. Clinically, AKs present as erythematous
TACROLIMUS
scaling macules and papules. These may be very hyperkera-
Tacrolimus has few reported mucocutaneous side effects.61 totic scales, some with a cutaneous horn. The pathogenesis
In one study, where 15 transplant recipients were switched of actinic keratosis is identical to SCC.
from cyclosporine to tacrolimus, gingival hyperplasia In the general population, the likelihood of an invasive
resolved in all patients within 1 year and hypertrichosis SCC evolving from a given AK has been estimated to occur
resolved in all cases within 6 months.61 Alopecia has been at a rate of 0.075 to 0.096% per lesion per year.4 This may
reported in transplant recipients taking tacrolimus, and be higher in OTRs.
in one case series it occurred in 29% of kidney transplant In OTRs, the actinic burden may be very high and dis-
recipients when other potential causes of alopecia were tressing to the patient. From the clinical perspective, dif-
ruled out.62 ferentiating AK from SCC may be difficult, and a biopsy is
warranted for lesions not responding to traditional therapy
for actinic keratosis.
SIROLIMUS AND EVEROLIMUS
The mainstay of treatment for an isolated AK is cryo-
Cutaneous side effects in kidney transplant recipients receiv- therapy. However, OTRs often have numerous AKs, render-
ing sirolimus may be severe and are well characterized.63 ing cryotherapy impractical. In particular, male Caucasian
Aphthous ulceration is significantly associated with siroli- OTRs often develop the “transplant scalp” which involves the
mus therapy and was observed in 60% of patients.63 Aggres- development of numerous AKs on the scalp.4 This increases
sive use of a potent topical steroid may aid in treatment of the likelihood of developing SCC with numerous lesions.
these ulcers. Disorders of the pilosebaceous unit were fre- Also, this can make surgical margin control for SCC on the
quently observed, with acneiform eruptions being the most scalp difficult. For transplant scalp, field treatment with a
common and observed in 46% of patients.63 Chronic edema topical therapy is recommended. Treatment options are sim-
was seen in 55% and angioedema in 15%.63 Mucous mem- ilar to those mentioned for NMSC including 5-­fluorouracil
brane pathologies were also very common.63 Alopecia of cream, imiquimod, and photodynamic therapy.4 For par-
the scalp was observed in 11% and hypertrichosis was seen ticularly hyperkeratotic lesions, curettage may be necessary
in 16%.63 During the 3-month period after completion of before treatment. Sun protection should be advised.
the study, 12% of patients had to stop sirolimus secondary
to cutaneous effects, including hidradenitis suppurativa, SEBORRHEIC KERATOSIS
severe acne, severe limb edema, and nail disorders.63
Seborrheic keratoses (SKs) are common benign skin lesions
that are almost ubiquitous among older individuals. SKs
present as tan to brown, waxy, “stuck-on” appearing pap-
Premalignant and Benign ules (Fig. 34.4). These may be more common in OTRs; how-
Cutaneous Tumors in Renal ever these may appear similarly to viral warts, which occur
more commonly in OTRs. SKs are also more prevalent in
Transplant Patients lighter skinned, older patients who have been transplanted
the longest.64 SKs may become inflamed and be treated
ACTINIC KERATOSIS
with cryotherapy or curettage.
Actinic keratoses (AKs) are precursor lesions of SCC. AKs SKs are generally felt to be benign; however, if there is a
are among the most frequently encountered skin lesions in suspicious lesion in an OTR, this should be biopsied. A retro-
clinical dermatology practice. They present on sun-dam- spective study of SCC arising in SKs demonstrated a higher
aged skin of the head, neck, upper trunk, and extremities risk in OTRs.65 HPV testing was not done in this study.65
34 • Cutaneous Disease in Kidney Transplantation Patients 587

POROKERATOSIS
Porokeratoses are benign skin lesions that may rarely have
malignant potential to develop SCC. These present as tan-
to-brown macules or thin plaques with central atrophy and
with a peripheral collarette of scale. Disseminated superfi-
cial actinic porokeratosis (DSAP) located on the extremi-
ties is often seen in OTRs with chronic ultraviolet exposure
history. The incidence of porokeratoses in OTRs, from two
population-based studies, is between 8% to 10.68%.66
Immunosuppression has been suggested to play a role
in the development of porokeratosis, and the incidence of
porokeratoses has been directly linked to strength of immu-
nosuppressive therapy.67 The risk of malignant transfor-
mation of porokeratosis to SCC in the general population is
approximately 7%, which may be greater in OTRs second-
ary to immunosuppression, larger lesion sizes, and config- Fig. 34.5 Extensive common warts on the hands of a kidney transplant
uration.67 Treatment is challenging and mostly cosmetic; recipient.
cryotherapy often leads to unwanted scarring. If required,
treatment can be approached through topical chemothera-
pies including 5-fluorouracil or imiquimod, photodynamic cutaneous infection was viral warts, which was found in
therapy, and chemical peels.68 Sun protection should be 38% of patients and increased in frequency in concordance
advised. with duration from transplant75 (Fig. 34.5). These results
were consistent with several previous studies.75
The severity of cutaneous warts may be pronounced
Infections and Inflammatory in immunosuppressed patients (see Fig. 34.5). Acquired
epidermodysplasia verruciformis (EV), which is a specific
Cutaneous Findings in OTRs pathology finding and suggests an increased susceptibility
to HPV infection, has been reported in immunocompro-
CUTANEOUS INFECTIONS
mised patients.76 There may be an association with wors-
Overall, the most frequently encountered cutaneous infec- ening disease with azathioprine; however, this requires
tions in transplant recipients are superficial fungal infec- further study.76
tions and viral warts. HPV infections in OTRs can be challenging to treat and
are an additional risk factor for NMSC, especially in this
Viral Infections patient population.77 Treatment can either be attempted
Given the suppression of T cell immunity and the role of through physical destruction (cryotherapy, curettage,
T lymphocytes in combating viral pathogens, viral infec- laser) or topical therapies including salicylic acid, podophyl-
tions tend to predominate 1 to 6 months posttransplanta- lum, cantharidin, trichloroacetic acid, topical cidofovir,
tion.69 Antiviral prophylaxis has significantly decreased the topical vitamin D, retinoids, 5-floururacil, or imiquimod.72
incidence of herpes virus (HHV) infections that historically Even with clearance, viral warts are a chronic condition,
occurred between 1 and 6 months posttransplantation.70,71 and recurrence should be anticipated.
However, reactivation of HHV represents a significant risk in Less frequently, molluscum contagiosum (MC), a pox-
this patient population and is important to diagnose and treat. virus that infects squamous epithelia, has been shown to
The most common HHV infections in OTRs are herpes sim- occur in nearly 7% of pediatric OTRs and extensively in
plex viruses (HSV types 1 and 2); varicella zoster virus (VZV); immunocompromised patients including OTRs.78,79 MC
cytomegalovirus; Epstein-Barr virus; and HHV-6, HHV-7, can be treated by surgically removing the umbilicated core,
and HHV-8, which is the causative agent of KS.72 HSV infec- cryoablation, electrodessication, cantharidin, trichloroace-
tion is most commonly manifested through mucocutaneous tic acid, podophyllum, and topical tretinoin.72 Other rare
lesions in OTRs and must be distinguished between siroli- viral infections include viral-associated trichodysplasia spi-
mus-induced mucositis in certain patients.72 The risk of HSV nulosa and acquired epidermodysplasia verruciformis.80,81
infection decreases 6 months after transplantation as immu-
nosuppression is tapered. However, when lesions develop, Fungal Infections
they can be painful, persistent, and rarely disseminate to dis- Cutaneous fungal infections also occur frequently because
tant organs.73 The most common treatments are acyclovir of the suppression of cell-mediated immunity. In the early
and valacyclovir, which can be used acutely and as suppres- posttransplantation period, when immunosuppression is
sive therapy. If VZV infection disseminates in a transplant greatest, disseminated candidiasis and aspergillosis, both
patient, intravenous acyclovir is the preferred treatment. primary cutaneous and secondary cutaneous resulting
After the sixth postoperative month, OTRs continue to be from invasive disease, are frequently seen, but with insti-
at high risk for developing community-acquired infections, tutional variability in incidence. Immunosuppressive doses
and over time more indolent infections begin to develop, also tend to be high 1 to 6 months posttransplantation and
which are frequently caused by HPVs.69,74 In a prevalence patients continue to be at significant risk of developing
study of renal transplant recipients, the most common an opportunistic deep fungal infection. Common mycoses
588 Kidney Transplantation: Principles and Practice

Fig. 34.7 Fungal nail infection (onychomycosis) affecting toenails.


Fig. 34.6 Pityriasis versicolor: pigmented macular lesions with superfi-
cial scaling over the shoulder region.
dermatitis (SD), acrochordons, porokeratosis, and sebor-
include dimorphic fungi infections, zygomycoses, phaeohy- rheic warts.64,86
phomycosis, hyalohyphomycosis, cryptococcosis, and con- In particular, SD in transplant recipients may be severe
tinued risk from Candida and Aspergillus. Skin lesions range and require treatment. SD has been reported to occur in
from nonerythematous papules, erythematous macules, 9.5% of renal transplant recipients, which is much higher
papules and nodules (+/− necrosis), hemorrhagic bullae, than SD prevalence of 1% to 5% reported in the normal
abscesses, and cellulitis. Cutaneous fungal infections may adult population.86 Although the precise etiology of SD is
occur in isolation or be a clue to underlying systemic infec- unclear, the impaired T cell immunity in OTRs may lead to
tion. Early recognition of systemic infection is imperative an altered immune response to Malassezia species and more
given the potential mortality associated with disseminated prevalent SD.86 Treatment with combination topical anti-
disease.82 As clinical presentation is variable and relatively fungals, steroids, or calcineurin inhibitors remain effective
nonspecific, a high index of suspicion and early biopsies of therapies for SD in OTR.86
skin lesions for histologic examination and culture are criti-
cal for diagnosis. Therapy usually requires systemic anti-
fungal compounds including triazoles of voriconazole and Conclusion
posaconazole, amphotericin B, and echinocandins such as
caspofungin, micafungin, and anidulafungin, but surgi- OTRs suffer from a higher burden of malignant, infectious,
cal debridement/excision may be an adjunct in localized inflammatory, and benign skin disease than the general
disease.82 population. In particular, the risk of NMSC in OTRs war-
Overall, superficial fungal infections are more common rants multidisciplinary care between a patient’s trans-
infections in OTRs. Mucocutaneous candidiasis is frequent, plant physician and a dermatologist. Patient education of
especially early in posttransplantation.83 Nystatin or topi- increased risk of cutaneous malignancy and the impor-
cal azoles are commonly used and are effective treatments tance of prevention are of the utmost importance in the care
for superficial candida infections. Dermatophyte infections of OTRs.
often present atypically because of a lack of erythema that
is normally produced by local inflammation.84 Pityriasis References
versicolor has been identified as the most common fungal 1. Zwald FO, Brown M. Skin cancer in solid organ transplant recipi-
infection in OTRs in several studies and can be treated with ents: advances in therapy and management: part I. Epidemiology of
skin cancer in solid organ transplant recipients. J Am Acad Dermatol
topical or oral medications, most commonly topical azole 2011;65:253–61. Quiz 62.
antifungals85 (Fig. 34.6). Other frequent infections include 2. Garrett GL, Blanc PD, Boscardin J, et al. Incidence of and risk factors
chronic tinea pedis and onychomycosis, both of which are for skin cancer in organ transplant recipients in the United States.
more common with chronic immunosuppression85 (Fig. JAMA Dermatol 2017;153:296–303.
34.7). Oral terbinafine is the treatment of choice and has 3. Urwin HR, Jones PW, Harden PN, et al. Predicting risk of nonmela-
noma skin cancer and premalignant skin lesions in renal transplant
the benefit of little interaction with immunosuppressants, recipients. Transplantation 2009;87:1667–71.
but liver function should be monitored during therapy. 4. Otley CC, Staskp T. Skin disease in organ transplantation. New York:
Cambridge University Press; 2008.
5. Arron ST, Jennings L, Nindl I, et al. Viral oncogenesis and its role in
Common Inflammatory Disorders nonmelanoma skin cancer. Br J Dermatol 2011;164:1201–13.
6. Genders RE, Mazlom H, Michel A, et al. The presence of betapapillo-
mavirus antibodies around transplantation predicts the development
Although several of the inflammatory dermatoses encoun- of keratinocyte carcinoma in organ transplant recipients: a cohort
tered in OTRs are due to drug effects, several conditions study. J Invest Dermatol 2015;135:1275–82.
arise from an immunosuppressed state or hypersensitiv- 7. Chung CL, Nadhan KS, Shaver CM, et al. Comparison of posttrans-
plant dermatologic diseases by race. JAMA Dermatol 2017;153(6):
ity reaction, including folliculitis (which may be bacterial 552–8.
or fungal or viral), drug rash, keratosis pilaris, hair loss of 8. Vinzon SE, Braspenning-Wesch I, Muller M, et al. Protective vaccina-
telogen effluvium-type, sebaceous hyperplasia, seborrheic tion against papillomavirus-induced skin tumors under immunocom-
34 • Cutaneous Disease in Kidney Transplantation Patients 589

petent and immunosuppressive conditions: a preclinical study using a 31. Markovic SN, Erickson LA, Rao RD, et al. Malignant melanoma in the
natural outbred animal model. PLoS Pathog 2014;10:e1003924. 21st century, part 1: epidemiology, risk factors, screening, preven-
9. Winkelhorst JT, Brokelman WJ, Tiggeler RG, Wobbes T. Incidence and tion, and diagnosis. Mayo Clin Proc 2007;82:364–80.
clinical course of de-novo malignancies in renal allograft recipients. 32. Morton DL, Thompson JF, Cochran AJ, et al. Sentinel-node biopsy or
Eur J Surg Oncol 2001;27:409–13. nodal observation in melanoma. N Engl J Med 2006;355:1307–17.
10. Sheil AG, Disney AP, Mathew TH, Amiss N. De novo malignancy 33. Faries MB, Thompson JF, Cochran AJ, et al. Completion dissection or
emerges as a major cause of morbidity and late failure in renal trans- observation for sentinel-node metastasis in melanoma. N Engl J Med
plantation. Transplant Proc 1993;25:1383–4. 2017;376:2211–22.
11. Humphreys TR, Shah K, Wysong A, Lexa F, MacFarlane D. The role 34. Robert C, Karaszewska B, Schachter J, et al. Improved overall survival
of imaging in the management of patients with nonmelanoma skin in melanoma with combined dabrafenib and trametinib. N Engl J Med
cancer: when is imaging necessary? J Am Acad Dermatol 2017; 2015;372:30–9.
76:591–607. 35. Robert C, Schachter J, Long GV, et al. Pembrolizumab versus ipilim-
12. Bavinck JN, Tieben LM, Van der Woude FJ, et al. Prevention of skin umab in advanced melanoma. N Engl J Med 2015;372:2521–32.
cancer and reduction of keratotic skin lesions during acitretin ther- 36. Arron ST, Canavan T, Yu SS. Organ transplant recipients with Merkel
apy in renal transplant recipients: a double-blind, placebo-controlled cell carcinoma have reduced progression-free, overall, and disease-
study. J Clin Oncol 1995;13:1933–8. specific survival independent of stage at presentation. J Am Acad Der-
13. Kovach BT, Sams HH, Stasko T. Systemic strategies for chemopre- matol 2014;71:684–90.
vention of skin cancers in transplant recipients. Clin Transplant 37. Garrett GL, Zargham H, Schulman JM, Jafarian F, Yu SS, Arron ST.
2005;19:726–34. Merkel cell carcinoma in organ transplant recipients: case reports and
14. Zwald FO, Brown M. Skin cancer in solid organ transplant recipients: review of the literature. JAAD Case Rep 2015;1:S29–32.
advances in therapy and management: part II. Management of skin 38. Nghiem PT, Bhatia S, Lipson EJ, et al. PD-1 Blockade with pembrolizumab
cancer in solid organ transplant recipients. J Am Acad Dermatol in advanced Merkel-cell carcinoma. N Engl J Med 2016;374:2542–52.
2011;65:263–79. Quiz 80. 39. McCoppin HH, Christiansen D, Stasko T, et al. Clinical spectrum of
15. Jirakulaporn T, Endrizzi B, Lindgren B, Mathew J, Lee PK, Dudek AZ. atypical fibroxanthoma and undifferentiated pleomorphic sarcoma
Capecitabine for skin cancer prevention in solid organ transplant in solid organ transplant recipients: a collective experience. Dermatol
recipients. Clin Transplant 2011;25:541–8. Surg 2012;38:230–9.
16. Chen AC, Martin AJ, Choy B, et al. A phase 3 randomized trial 40. Chiereghin A, Barozzi P, Petrisli E, et al. Multicenter prospective study
of nicotinamide for skin-cancer chemoprevention. N Engl J Med for laboratory diagnosis of HHV8 infection in solid organ donors and
2015;373:1618–26. transplant recipients and evaluation of the clinical impact after trans-
17. Jenni D, Karpova MB, Muhleisen B, et al. A prospective clinical trial plantation. Transplantation 2017;101:1935–44.
to assess lapatinib effects on cutaneous squamous cell carcinoma and 41. Karia PS, Azzi JR, Heher EC, Hills VM, Schmults CD. Association of
actinic keratosis. ESMO Open 2016;1:e000003. sirolimus use with risk for skin cancer in a mixed-organ cohort of
18. Siano M, Molinari F, Martin V, et al. Multicenter phase II study of solid-organ transplant recipients with a history of cancer. JAMA Der-
panitumumab in platinum pretreated, advanced head and neck squa- matol 2016;152:533–40.
mous cell cancer. Oncologist 2017;22(7). 782-70. 42. Lim WH, Russ GR, Wong G, Pilmore H, Kanellis J, Chadban SJ. The
19. Degache E, Crochet J, Simon N, et al. Major response to pembroli- risk of cancer in kidney transplant recipients may be reduced in those
zumab in two patients with locally advanced cutaneous squamous maintained on everolimus and reduced cyclosporine. Kidney Int
cell carcinoma. J Eur Acad Dermatol Venereol 2018;32(7):e257–8. 2017;91:954–63.
20. Taylor P. Regeneron and Sanofi bag breakthrough status for PD-1 43. Asgari MM, Arron ST, Warton EM, Quesenberry CP, Weisshaar D.
latecomer. Fierce Biotech website 2017. Available online at: http:// Sirolimus use and risk of cutaneous squamous cell carcinoma (SCC)
www.fiercebiotech.com/biotech/regeneron-and-sanofi-bag-break- in solid organ transplant recipients (SOTRs). J Am Acad Dermatol
through-status-for-pd-1-latecomer. Accessed September 21, 2017. 2015;73:444–50.
21. Sekulic A, Migden MR, Basset-Seguin N, et al. Long-term safety and 44. Cowen EW, Nguyen JC, Miller DD, et al. Chronic phototoxicity and
efficacy of vismodegib in patients with advanced basal cell carci- aggressive squamous cell carcinoma of the skin in children and
noma: final update of the pivotal ERIVANCE BCC study. BMC Cancer adults during treatment with voriconazole. J Am Acad Dermatol
2017;17:332. 2010;62:31–7.
22. Mohan SV, Chang J, Li S, Henry AS, Wood DJ, Chang AL. Increased 45. Clancy CJ, Nguyen MH. Long-term voriconazole and skin cancer: is
risk of cutaneous squamous cell carcinoma after vismodegib therapy there cause for concern? Curr Infect Dis Rep 2011;13:536–43.
for basal cell carcinoma. JAMA Dermatol 2016;152:527–32. 46. Singer JP, Boker A, Metchnikoff C, et al. High cumulative dose expo-
23. Bhutani T, Abrouk M, Sima CS, et al. Risk of cutaneous squamous cell sure to voriconazole is associated with cutaneous squamous cell
carcinoma after treatment of basal cell carcinoma with vismodegib. carcinoma in lung transplant recipients. J Heart Lung Transplant
J Am Acad Dermatol 2017;77(4):713–8. 2012;31:694–9.
24. Zwald F, Leitenberger J, Zeitouni N, et al. Recommendations for solid 47. Mansh M, Binstock M, Williams K, et al. Voriconazole exposure and
organ transplantation for transplant candidates with a pretransplant risk of cutaneous squamous cell carcinoma, aspergillus colonization,
diagnosis of cutaneous squamous cell carcinoma, Merkel cell carcinoma invasive aspergillosis and death in lung transplant recipients. Am J
and melanoma: a consensus opinion from the International Transplant Transplant 2016;16:262–70.
Skin Cancer Collaborative (ITSCC). Am J Transplant 2016;16:407–13. 48. Zwald FO, Spratt M, Lemos BD, et al. Duration of voriconazole expo-
25. Arron ST, Raymond AK, Yanik EL, et al. Melanoma outcomes in sure: an independent risk factor for skin cancer after lung transplanta-
transplant recipients with pretransplant melanoma. Dermatol Surg tion. Dermatol Surg 2012;38:1369–74.
2016;42:157–66. 49. Ibrahim SF, Singer JP, Arron ST. Catastrophic squamous cell carci-
26. Zwald FO, Christenson LJ, Billingsley EM, et al. Melanoma in solid noma in lung transplant patients treated with voriconazole. Dermatol
organ transplant recipients. Am J Transplant 2010;10:1297–304. Surg 2010;36:1752–5.
27. Hollenbeak CS, Todd MM, Billingsley EM, Harper G, Dyer AM, Leng- 50. Ismail F, Mitchell L, Casabonne D, et al. Specialist dermatology clinics
erich EJ. Increased incidence of melanoma in renal transplantation for organ transplant recipients significantly improve compliance with
recipients. Cancer 2005;104:1962–7. photoprotection and levels of skin cancer awareness. Br J Dermatol
28. Ascha M, Ascha MS, Tanenbaum J, Bordeaux JS. Risk factors 2006;155:916–25.
for melanoma in renal transplant recipients. JAMA Dermatol 51. Koranda FC, Dehmel EM, Kahn G, Penn I. Cutaneous complica-
2017;153:1130–6. tions in immunosuppressed renal homograft recipients. JAMA
29. Brewer JD, Christenson LJ, Weaver AL, et al. Malignant melanoma in 1974;229:419–24.
solid transplant recipients: collection of database cases and compari- 52. Dessinioti C, Antoniou C, Katsambas A. Acneiform eruptions. Clin
son with surveillance, epidemiology, and end results data for outcome Dermatol 2014;32:24–34.
analysis. Arch Dermatol 2011;147:790–6. 53. Ponticelli C, Bencini PL. Nonneoplastic mucocutaneous lesions in
30. Milton CA, Barbara J, Cooper J, Rao M, Russell C, Russ G. The trans- organ transplant recipients. Transpl Int 2011;24:1041–50.
mission of donor-derived malignant melanoma to a renal allograft 54. de Fonclare AL, Khosrotehrani K, Aractingi S, Duriez P, Cosnes J,
recipient. Clin Transplant 2006;20:547–50. Beaugerie L. Erythema nodosum-like eruption as a manifestation of
590 Kidney Transplantation: Principles and Practice

azathioprine hypersensitivity in patients with inflammatory bowel 71. Meyers JD, Wade JC, Mitchell CD, et al. Multicenter collaborative
disease. Arch Dermatol 2007;143:744–8. trial of intravenous acyclovir for treatment of mucocutaneous herpes
55. Lindholm A, Pousette A, Carlstrom K, Klintmalm G. Ciclosporin-­ simplex virus infection in the immunocompromised host. Am J Med
associated hypertrichosis is not related to sex hormone levels follow- 1982;73:229–35.
ing renal transplantation. Nephron 1988;50:199–204. 72. Tan HH, Goh CL. Viral infections affecting the skin in organ transplant
56. Daley TD, Wysocki GP, Day C. Clinical and pharmacologic correla- recipients: epidemiology and current management strategies. Am J
tions in cyclosporine-induced gingival hyperplasia. Oral Surg Oral Clin Dermatol 2006;7:13–29.
Med Oral Pathol 1986;62:417–21. 73. Smith SR, Butterly DW, Alexander BD, Greenberg A. Viral infections
57. Heaphy Jr MR, Shamma HN, Hickmann M, White MJ. Cyclosporine- after renal transplantation. Am J Kidney Dis 2001;37:659–76.
induced folliculodystrophy. J Am Acad Dermatol 2004;50:310–5. 74. Berkhout RJ, Bouwes Bavinck JN, ter Schegget J. Persistence of
58. Bencini PL, Montagnino G, Sala F, De Vecchi A, Crosti C, Tarantino A. human papillomavirus DNA in benign and (pre)malignant skin
Cutaneous lesions in 67 cyclosporin-treated renal transplant recipi- lesions from renal transplant recipients. J Clin Microbiol 2000;
ents. Dermatologica 1986;172:24–30. 38:2087–96.
59. Wang K, Zhang H, Li Y, et al. Safety of mycophenolate mofetil versus 75. Rudlinger R, Smith IW, Bunney MH, Hunter JA. Human papillomavi-
azathioprine in renal transplantation: a systematic review. Trans- rus infections in a group of renal transplant recipients. Br J Dermatol
plant Proc 2004;36:2068–70. 1986;115:681–92.
60. Campistol JM, de Fijter JW, Flechner SM, Langone A, Morelon E, 76. Ovits CG, Amin BD, Halverstam C. Acquired epidermodysplasia
Stockfleth E. mTOR inhibitor-associated dermatologic and mucosal verruciformis and its relationship to immunosuppressive therapy:
problems. Clin Transplant 2010;24:149–56. report of a case and review of the literature. J Drugs Dermatol
61. Busque S, Demers P, Saint-Louis G, et al. Hypertrichosis and gingival 2017;16:701–4.
hypertrophy regression in renal transplants following the substitution 77. Benton EC, McLaren K, Barr BB, et al. Human papilloma virus infec-
of cyclosporin by tacrolimus. Ann Chir 1999;53:687–9. tion and its relationship to skin cancer in a group of renal allograft
62. Tricot L, Lebbe C, Pillebout E, Martinez F, Legendre C, Thervet E. recipients. Curr Probl Dermatol 1989;18:168–77.
Tacrolimus-induced alopecia in female kidney-pancreas transplant 78. Euvrard S, Kanitakis J, Cochat P, Cambazard F, Claudy A. Skin dis-
recipients. Transplantation 2005;80:1546–9. eases in children with organ transplants. J Am Acad Dermatol
63. Mahe E, Morelon E, Lechaton S, et al. Cutaneous adverse events in 2001;44:932–9.
renal transplant recipients receiving sirolimus-based therapy. Trans- 79. Mansur AT, Goktay F, Gunduz S, Serdar ZA. Multiple giant mollus-
plantation 2005;79:476–82. cum contagiosum in a renal transplant recipient. Transpl Infect Dis
64. Lally A, Casabonne D, Waterboer T, et al. Association of seborrhoeic 2004;6:120–3.
warts with skin cancer in renal transplant recipients. J Eur Acad Der- 80. Henley JK, Hossler EW. Acquired epidermodysplasia verruciformis
matol Venereol 2010;24:302–7. occurring in a renal transplant recipient. Cutis 2017;99:E9–12.
65. Conic RZ, Napekoski K, Schuetz H, Piliang M, Bergfeld W, Atanas- 81. Sperling LC, Tomaszewski MM, Thomas DA. Viral-associated tricho-
kova Mesinkovska N. The role of immunosuppression in squamous dysplasia in patients who are immunocompromised. J Am Acad Der-
cell carcinomas arising in seborrheic keratosis. J Am Acad Dermatol matol 2004;50:318–22.
2017;76:1146–50. 82. Virgili A, Zampino MR, Mantovani L. Fungal skin infections in organ
66. Herranz P, Pizarro A, De Lucas R, et al. High incidence of porokerato- transplant recipients. Am J Clin Dermatol 2002;3:19–35.
sis in renal transplant recipients. Br J Dermatol 1997;136:176–9. 83. Ribeiro PM, Bacal F, Koga-Ito CY, Junqueira JC, Jorge AO. Presence
67. Zenarola P, Melillo L, Lomuto M, Carotenuto M, Gomes VE, of Candida spp. in the oral cavity of heart transplantation patients.
Marzocchi W. Exacerbation of porokeratosis: a sign of immunodepres- J Appl Oral Sci 2011;19:6–10.
sion. J Am Acad Dermatol 1993;29:1035–6. 84. Miele PS, Levy CS, Smith MA, et al. Primary cutaneous fungal infec-
68. Stoff B, Salisbury C, Parker D, O’Reilly Zwald F. Dermatopathology tions in solid organ transplantation: a case series. Am J Transplant
of skin cancer in solid organ transplant recipients. Transplant Rev 2002;2:678–83.
(Orlando) 2010;24:172–89. 85. Shuttleworth D, Philpot CM, Salaman JR. Cutaneous fungal infection
69. Fishman JA. Infection in solid-organ transplant recipients. N Engl J following renal transplantation: a case control study. Br J Dermatol
Med 2007;357:2601–14. 1987;117:585–90.
70. Rubin RH, Kemmerly SA, Conti D, et al. Prevention of primary cyto- 86. Lally A, Casabonne D, Newton R, Wojnarowska F. Seborrheic derma-
megalovirus disease in organ transplant recipients with oral ganciclo- titis among Oxford renal transplant recipients. J Eur Acad Dermatol
vir or oral acyclovir prophylaxis. Transpl Infect Dis 2000;2:112–7. Venereol 2010;24:561–4.
35 Cancer in Dialysis and
Transplant Patients
ANGELA CLAIRE WEBSTER, BRENDA MARIA ROSALES, and JOHN F.
THOMPSON

CHAPTER OUTLINE Cancer in Dialysis Patients Chronic Antigenic Stimulation and Immune
Cancer Risk in Dialysis Patients Regulation
Etiology of the Increased Risk of Cancer in Environmental Factors
Dialysis Patients Immunosuppressive Treatments
Types of Cancer in Dialysis Patients Types of Cancer in Kidney Transplant
Renal Tract Malignancy Recipients
Thyroid Cancer Posttransplant Lymphoproliferative
Myeloma Disorder
Dialysis in Patients With a History of Cancer Kaposi’s Sarcoma
Cancer Screening and Prevention in Dialysis Transplantation in Recipients With a History
Patients of Cancer
Management of Cancer in Dialysis Patients Transmission of Cancer From the Donor
Survival in Dialysis Patients who Develop Screening for Cancer in Kidney Transplant
Cancer Recipients
Cancer in Kidney Transplant Recipients Time of Cancer Presentation
Cancer Risk in Kidney Transplant Recipients Management of Cancer in Kidney Transplant
Recipients
Etiology of the Increased Risk of Cancer in
Kidney Transplant Recipients Survival in Kidney Transplant Recipients
Who Develop Cancer
Impaired Immune Surveillance
Conclusions
Oncogenic Viruses

Dialysis patients and kidney transplant recipients have con- Generally, the types of cancer were similar to the cancers
siderably greater cancer risk than the general population. observed with increased frequency in transplant recipi-
This chapter discusses cancer in dialysis patients and kid- ents. Most common were cancers of the urinary tract, but
ney transplant recipients, with the exception of skin malig- cancers of the tongue, liver, lower genital tract in women,
nancies, which are considered separately in Chapter 34. external genitalia in men, and thyroid, lymphomas, and
multiple myeloma, had increased incidence.

Cancer in Dialysis Patients CANCER RISK IN DIALYSIS PATIENTS


Soon after the first reports of cancer arising de novo in kid- Some of the most comprehensive long-term data on the
ney transplant recipients,1 it was suggested that dialysis development of malignancy in dialysis patients and kid-
patients were also at heightened risk of cancer.2 Subse- ney transplant recipients are available from the Australia
quent reports confirmed that the incidence of malignancy and New Zealand Dialysis and Transplant (ANZDATA)
was higher while on dialysis than in the general population. Registry. This registry has collected information on all
Most early reports were about cancers affecting the renal patients in Australia and New Zealand who have been
tract, either directly or indirectly. It is now clear that there treated with dialysis or received a kidney transplant since
is an overall increase in incidence of malignancy in patients 1963. Although there are several much larger registries
with chronic kidney disease (CKD).3 The first study large in the world, the population base and completeness of the
enough to study the relationship between cancer, includ- information recorded sets the ANZDATA registry apart
ing less common types, estimate small increases in risk, from most others. The 2012 ANZDATA report examined
and seek associations with the causes of CKD and modali- the incidence of cancer in 50,635 patients (with 145,043
ties of dialysis (hemodialysis or peritoneal dialysis) was by person-years of follow-up) treated with dialysis and 17,150
Maisonneuve.4 This confirmed an overall increased risk of patients (159,413 person-years of follow-up) after a first
cancer in patients with end-stage kidney disease (ESKD). kidney transplant in Australia and New Zealand, compared
591
592 Kidney Transplantation: Principles and Practice

with the general population incidence, between 1982 and patients treated in other countries have also been reported
2009.5 Indirect standardization was used, standardizing (summarized in Table 35.1).6–8 These estimations dem-
for differences in age, sex, and calendar year, to calculate onstrate the remarkable similarities in incidence trends.
standardized incidence ratios (SIR) with their 95% confi- When considering cancer risk by site it is apparent that
dence (CIs). SIR can be interpreted as risk ratio, where an the pattern of increased risk is varied. For many cancers,
SIR value of 1 is risk equal to that of the general population for example cancers of the lung and colon, there is a slight
of similar age and sex, living in an equivalent time period increase in risk among dialysis patients, with a somewhat
in the same country, and an SIR of 2 is double the risk, greater increase after transplantation. For several other
etc. Comprehensive data on the standardized incidence of cancers, however, the risk increase after transplantation
cancer in both kidney transplant recipients and dialysis is more marked. Most of these are known or postulated to

TABLE 35.1 Risk of Cancer After Commencement of Dialysis and After a First Kidney Transplant Across Different Countries
Dialysis Before First Transplant After First Transplant
Standardized Incidence Ratio [95% CI] Standardized Incidence Ratio [95% CI]
Australia United States Denmark Australia United States Denmark
Cancer Site ICD-O (2012)b (2016)c (2017)d (2012)a (2016)b (2017)c
INFECTION RELATED
Kaposi’s sarcoma C46 8.88 [4.62–17.07] 6.4 [2.8–13] 22.29 [15.06–32.98] 55 [44–68]
All lymphoma C81–85, C96 1.13 [0.91–1.40] 9.64 [8.73–10.66]
Non-Hodgkin’s 1.7 [1.5–2] 1.6 [1.2–2.2] 5.9 [5.5–6.3] 6.1 [4.6–8.2]
lymphoma
Hodgkin’s lym- 0.87 [0.45–1.5] 3.4 [2.6–4.3]
phoma
Liver C22 2.41 [1.81–3.21] 1.8 [1.5–2.2] 2.80 [1.91–4.12] 1 [0.79–1.3]
Stomach C16 1.55 [1.22–1.97] 1.4 [1.1–1.7] 1.44 [0.99–2.09] 1.6 [1.3–1.9]
Oropharynx 1.2 [0.88–1.7] 1.3 [0.97–1.7]
Anus 2.6 [1.7–3.8] 4.8 [3.7–6.2]
Uterus C54–55 1.36 [0.96–1.89] 0.93 [0.72–1.2] 1.69 [1.17–2.45] 0.94 [0.75–1.2]
Cervix C53 2.81 [1.91–4.13] 0.89 [0.59–1.3] 4.81 [3.58–6.47] 1.1 [0.75–1.5]
Ovary C56 0.96 [0.60–1.55] 1.22 [0.73–2.07]
Vulva
Penis and male C60, 63 1.40 [0.45–4.35] 10.94 [6.06–19.75]
genital
Other genital sites 3 [2–4.2] 5.1 [4–6.5]
IMMUNE RELATED
Trachea bronchus C33–34 1.70 [1.53–1.88] 1.76 [1.51–2.06]
lung
Lunga 1.2 [1.1–1.3] 1.6 [1.5–1.7]
Melanoma C43 1.08 [0.88–1.20] 1.5 [1.2–1.8] 2.74 [2.44–3.09] 2.8 [2.5–3.2]
Nonepithelial skin 2.5 [1.5–3.9] 13 [11–15]
Lip C00 0.13 [0.03–0.05] 3.5 [1.7–6.2] 3.9 [2.0–7.5] 18 [15–22] 12.3
[6.1–24.5]
ESKD-RELATED
Kidney, ureter, C64–66, 88 5.99 [5.35–6.70] 8.58 [7.52–9.78]
urethra
Kidney C64 9 [8.4–9.6] 2.8 [2.2–3.7] 6.4 [5.9–6.8] 6.9 [5.0–9.6]
Other urinary tract 1.6 [1.3–1.9] 1.9 [1.6–2.2]
Thyroid C73 4.32 [3.33–5.62] 4 [3.5–4.6] 3 [1.7–5.5] 4.12 [3.13–5.44] 2.9 [2.5–3.4] 4.7 [2.3–9.9]
Multiple myeloma C90 7.09 [6.12–8.22] 1.8 [1.5–2.2] 2.23 [1.49–3.32] 1.8 [1.4–2.1]
Other
Colorectal C18–20 1.03 [0.92–1.16] 1.2 [1.1–1.3] 1.44 [0.99–2.09] 1.1 [0.96–1.2]
Pancreas C25 1.14 [0.86–1.51] 1.1 [0.86–1.4] 1.27 [0.84–1.93] 1.5 [1.3–1.8]
Leukemia C91–95 1.09 [0.03–1.43] 1.4 [1.1–1.8] 1.81 [1.34–2.46] 1.8 [1.5–2.1]
Prostatea C61 0.58 [0.5–0.66] 0.85 [0.78–0.92] 0.82 [0.68–0.98] 0.92 [0.85–0.98]
Breasta C50 1.28 [1.11–1.47] 1.2 [1–1.3] 1.22 [1.03–1.44] 0.95 [0.86–1.0]
Esophagusa C15 1.61 [1.18–2.18] 0.96 [0.68–1.3] 3.93 [2.91–5.29] 1.3 [1–1.7]
aEvidence of infection inconclusive (Vadjic et al., 2006).
Oropharynx cancer includes cancers of the tongue, tonsils, and other oropharynx sites.
Other genital cancers include cancers of the vagina, vulva, and penis.
Nonepithelial skin is defined as skin cancers excluding melanoma, Kaposi’s sarcoma, and squamous and basal cell carcinomas.
Data from:
bWebster AC, Peng A, Kelly PJ. Cancer. In: ANZDATA Registry Report 2012. 35th Annual Report. Adelaide, South Australia: Australia and New Zealand Dialysis and

Transplant Registry; 2012.


cYanik EL, Clarke CA, Snyder JJ, Pfeiffer RM, Engels EA. Variation in cancer incidence among patients with ESRD during kidney function and nonfunction intervals.

J Am Soc Nephrol 2016;27:1495–504.


dHortlund M, Arroyo Muhr LS, Storm H, Engholm G, Dillner J, Bzhalava D. Cancer risks after solid organ transplantation and after long-term dialysis. Int J Cancer

2017;140:1091–101.
35 • Cancer in Dialysis and Transplant Patients 593

have a viral etiology; for example, carcinoma of the cervix, cancers.17 In both dialysis patients and transplant recipi-
lymphoma, and Kaposi’s sarcoma. ents, the increased risk of lymphoma is thought likely to be
due to activation of dormant Epstein-Barr virus (EBV).18
ETIOLOGY OF THE INCREASED RISK OF CANCER IN
DIALYSIS PATIENTS TYPES OF CANCER IN DIALYSIS PATIENTS
The etiology of cancer risk in dialysis patients includes the Renal Tract Malignancy
presence of chronic infection (especially in the urinary Risk of renal tract malignancies is particularly high for
tract), a depressed immune system, previous treatment the dialysis population; risk of renal cancer SIR, 4.03;
with immunosuppressive or cytotoxic drugs, nutritional 95% CI 3.88 to 419 and bladder SIR, 1.57; 95% CI 1.51 to
deficiencies, and altered deoxyribonucleic acid (DNA) 1.64.11 Population-specific SIRs are listed in Table 35.1.
repair mechanisms.9 Importantly, cancer is not related The increase in renal tract malignancies during dialysis
to dialysis modality, but rather the uremic state.4 Ure- in the US population is largely driven by localized kidney
mia is associated with impaired T cell immunity and a cancers.8 In the dialysis population, the risk of developing
state of chronic inflammation, which lead to DNA muta- cancer of the kidney or bladder is relatively (but not abso-
tions in proliferating cells and deregulatory release of lutely) greater at younger ages, and in women rather than
cytokines implicated in cancer development and progres- men. The SIR for kidney cancer increases significantly
sion. CKD also leads to the accumulation of carcinogenic with time on dialysis, whereas the SIR for bladder cancer
compounds to which dialysis patients are exposed from is progressively decreased.19 Increased risk for cancers of
the environment and possibly in the dialysate. Increas- the urinary tract is associated with increasing duration of
ing the frequency of dialysis has been associated with maintenance dialysis before first kidney transplant where
reduced genomic damage and plasma urea concentra- the adjusted hazard ratio (HR) for urinary tract cancers in
tions in patients with ESKD.10 Excess cancer risk may also recipients that were on dialysis for more than 4.5 years was
be due to an interaction of immune dysfunction induced 2.57 (95% CI 1.33–4.95) compared with those that were
by uremia with established risk factors (ultraviolet [UV] on dialysis for less than 1.5 years. However when looking
radiation, tobacco, alcohol).11 Several reports have also at the differences in absolute terms, this was not significant
demonstrated high levels of cumulative radiation dose in between groups.20 There is no excess risk of kidney cancer
patients with ESKD on dialysis.12 Although there have in patients with ESKD due to autosomal dominant polycys-
been no follow-up studies that have measured cumula- tic kidney disease.
tive radiation doses and cancer outcomes in patients with
ESKD, high exposure (cumulative effective dose >50 mSv) Thyroid Cancer
has been reported to increase cancer mortality by 5% in Patients on dialysis are at much higher risk of develop-
other populations.12 ing thyroid cancer than the general population or trans-
In addition to the persistent metabolic changes associ- plant recipients (see Table 35.1). CKD has been known
ated with ESKD, the underlying causative disease, and the to affect thyroid hormone metabolism. The prevalence of
development of certain complications of ESKD may also subclinical hypothyroidism (21.8%) and nodular thyroid
predispose to cancer. The risk of renal cell cancer (RCC) disease (24.1%) are almost three times higher in patients
is increased in patients with acquired cystic disease and with ESKD than age-, sex-, and weight-matched controls
seems to be related to the total duration of CKD, rather (7.1%).21 The risk of thyroid cancer is 10.1-fold higher
than the duration of dialysis.13 Other conditions predis- (95% CI 1.12–91.0) in patients with ESKD and secondary
posing to cancer include Balkan nephropathy and anal- hyperparathyroidism.22 It is thought that a low glomerular
gesic nephropathy, both of which are associated with filtration rate changes the metabolism of thyroid hormones,
a high risk of developing tumors of the renal pelvis and although the exact mechanism remains unknown. It may
ureter.15 have to do with altered metabolism of iodine, decreased
The increased risk of some cancer types is rapidly reversed peripheral sensitivity of hormones, or autoimmune thyroid-
when immunosuppression is reduced or withdrawn after itis. Another possible explanation for the observed increase
kidney transplant failure. These cancer types include Kapo- in risk of thyroid tumors is the repeated imaging of the neck
si’s sarcoma, non-Hodgkin lymphoma, melanoma, and to investigate secondary hyperparathyroidism. Studies
squamous cell carcinomas of the lip. However, the risk of assessing cumulative doses of radiation have been too small
cancer at other sites remains significantly elevated after to prove any association; however, the observation that
iatrogenic immunosuppression is ceased. These cancer the frequency of thyroid tumors increases with duration on
types include leukemia, lung cancer, and cancers related to dialysis supports this hypothesis. Any contribution of over-
ESKD.14 detection through increases in incidental thyroid imaging
The frequency of viral infections in dialysis patients is in dialysis and transplant patients is difficult to quantify.
poorly documented, but there is no doubt that patients
with ESKD have a greater than normal exposure to hepa- Myeloma
titis B and hepatitis C viruses,15 and this probably accounts Renal disease is common in myeloma and can affect the
for the observed excess of liver cancer. Human papillo- glomeruli, tubules, and interstitium in isolation or in com-
mavirus (HPV) is associated with cancers of the tongue, bination. CKD is noted at presentation in up to half of newly
cervix, vagina, vulva, and penis.16 HIV is also associated diagnosed myeloma patients but is frequently responsive
with increased risk of Kaposi’s sarcoma, non-Hodgkin’s to the correction of factors contributing to acute injury.
lymphoma, and Hodgkin’s lymphoma, lip, and cervical The presence and severity of kidney disease correlates with
594 Kidney Transplantation: Principles and Practice

patient survival, and overall prognosis is related to response duration of dialysis, and when cancer is detected by screen-
of the renal disease to therapy.23 In the past decade there ing, rather than after causing symptoms.30 Early diagnosis
have been major advances in myeloma therapy and man- of RCC by regular imaging of patients with ESKD who are
agement, including autologous stem cell transplant, the use on dialysis would result in an improved outcome.31
of novel therapeutic agents such as protease inhibitors, and
immunomodulatory drugs (bortezomib, lenalidomide, and MANAGEMENT OF CANCER IN DIALYSIS
thalidomide), and the use of the free light chain assay pro- PATIENTS
viding greater diagnostic precision, which has significantly
improved survival. A recent study predicted 5-year survival For dialysis patients who have surgical treatment for malig-
of between 12% and 32% dependent on age and improved nancy, postoperative complications are much higher.32
survival in myeloma patients on peritoneal dialysis com- Radical nephrectomy may be superior to partial nephrec-
pared with hemodialysis (HR 0.7, 95% CI 0.6–0.9).24 tomy for treatment of localized RCCs, causing 79 fewer
deaths per 1000, with no difference in recurrence.33 Many
DIALYSIS IN PATIENTS WITH A HISTORY OF chemotherapy agents are excreted by the kidney, meaning
dosage and scheduling adaptation for those with ESKD.34
CANCER
Using available specific drug management recommenda-
Rarely, CKD may be a consequence of malignancies directly, tions for drug adjustment and avoiding premature elimina-
in the case of myeloma, or via glomerulopathy, as those tion of drug during dialysis is paramount. Treatment with
arising in lung or colon cancer,25 possibly as a result of radioactive iodine can be undertaken for thyroid cancer in
tumor-associated antibodies. Nephrotic syndrome is most dialysis patients, but dosage adjustment and consideration
often associated with Hodgkin’s disease. Malignant dis- of bystander exposure is necessary because iodine is cleared
ease of the kidney or ureter can impair kidney function by mainly by the kidneys or by the dialysis process.35
causing obstruction, and occasionally kidney dysfunction
results from a treatment-related nephropathy secondary to SURVIVAL IN DIALYSIS PATIENTS WHO
toxicity from radiation or drugs. DEVELOP CANCER
CANCER SCREENING AND PREVENTION IN Cancer-specific mortality data in dialysis patients is rela-
tively sparse. In Japan, a nation with a large population of
DIALYSIS PATIENTS
people on long-term dialysis, an analysis of deaths caused
There are currently no standard recommendations for by cancer (including renal tract tumors) revealed that the
cancer screening in the dialysis population. Some authors relative risk (RR) of cancer mortality was greatly increased
have suggested that routine cancer screening of patients on compared with the general population (male RR 2.48;
long-term dialysis is not cost effective as, unlike the general female RR 3.99).36 A higher cumulative incidence of can-
population, early detection may not lead to improved sur- cer-specific mortality has been found in patients who initi-
vival in those with life-limiting ESKD, and treatment effects ated dialysis after 2003, compared with previous years.11
may not be as certain.26 Others have argued that selective This may be due to improved overall survival in dialysis
screening in younger patients and for known cancer types patients, allowing time for cancer development.
are warranted.
Cost-effectiveness analyses can help put expectations
from screening programs into context. Breast cancer
Cancer in Kidney Transplant
screening in women on dialysis has resulted in an absolute Recipients
reduction in breast cancer mortality of 0.1%, with a net
gain in life expectancy of only 1.3 days. The total incre- The magnitude of the risk of some cancers increases further
mental cost to screen and save one cancer death approxi- after transplantation (see Table 35.1). Cancer risk increases
mated $403,000 per life-years saved from breast cancer.27 steadily over time, with absolute increase strongly depen-
Similarly, the incremental cost of colorectal cancer screen- dent on age at the time of transplantation.
ing has been calculated at $122,977 per life-year saved for
dialysis patients not listed on the transplant waiting list, CANCER RISK IN KIDNEY TRANSPLANT
and $85,095 for patients on the waiting list, which greatly RECIPIENTS
exceeded the typical thresholds for acceptable cost effec-
tiveness.28 These costs were largely dependent on the test Early reports of de novo cancers arising in immunosup-
characteristics of the screening test. In a study where the pressed transplant recipients based on single-center and
colorectal cancer screening test had poor sensitivity but rea- registry reports indicated that malignancies, excluding
sonable specificity, 69% of cases of advanced cancer would nonmelanoma skin cancers (NMSC), arose in 2% to 8% of
have been missed by immunochemical fecal occult blood patients.
test screening alone. Thus surveillance colonoscopy may be When incidence is based on time since transplantation, a
a more appropriate approach in patients with ESKD.29 more extreme picture emerges, with 34% to 50% of immu-
For cancers without a screening program in the general nosuppressed transplant recipients developing cancer after
population, but which occur more commonly in those on 20 years or more.37 Long-term data from the ANZDATA
dialysis, there are some situations where targeted screen- registry reveal that at 30 years the incidence of nonskin
ing may be warranted. Screening may be valuable for cancer is 33%, with some form of cancer (either skin or
RCC, where survival is best in young patients with a short nonskin cancer) developing in 80% of recipients overall.38
35 • Cancer in Dialysis and Transplant Patients 595

Transplant registry data from other countries also show The age trend for lymphoma, colorectal cancer, and breast
a substantial risk of cancer development in kidney trans- cancer risk was similar: Melanoma showed less variability
plant recipients, which steadily increases with time since across age cohorts, whereas prostate cancer showed no
transplantation.39–41 risk increase. Within the transplanted population, risk was
Fig. 35.1 and Table 35.2 demonstrate cancer epidemiol- affected by age differently for each sex (P = 0.007). Risk
ogy after transplantation. Long-term data from ANZDATA was increased by prior malignancy (HR 1.40, CI 1.3–1.89)
indicate that 1642 (10.8%) of 15,183 kidney transplant and white race (HR 1.36, CI 1.12–1.89), but reduced by
recipients developed cancer.42 Cancer rates were similar diabetic ESKD (HR 0.67, CI 0.50–0.89). As well as demon-
to those in the general population 20 to 30 years older. strating how absolute risk differs across patient groups, Fig.
Risk was inversely related to age (SIR 15–30 for children, 35.1 allows the risk of cancer developing after transplanta-
2 if >65 years). Females aged 25 to 29 had rates equiva- tion to be estimated, based on clinical details known at the
lent to women aged 55 to 59 in the general population. time of transplantation. For example, men aged 45 to 54

.5
<35 years
Transplanted female Transplanted male
Proportion with at least 1 non-skin cancer

General population female General population male


.4

.3

.2

.1

0
0 5 10 15 20
Time in years
A

.5
35–44 years
Transplanted female Transplanted male
Proportion with at least 1 non-skin cancer

General population female General population male


.4

.3

.2

.1

0
0 5 10 15 20
Time in years
B

Fig. 35.1 Cumulative risk of cancer (excluding nonmelanocytic skin and lip cancer) in kidney transplant recipients by age at transplantation, with
expected cumulative risk for a comparable general population of the same age and sex. (A) Less than 35 years; (B) 35 to 44 years; (C) 45 to 54 years; (D)
55 years and over. (Adapted from Webster AC, Craig JC, Simpson JM, Jones MP, Chapman JR. Identifying high risk groups and quantifying absolute risk of
cancer after kidney transplantation: a cohort study of 15,183 recipients. Am J Transplant 2007;7(9):2140–51.)
596 Kidney Transplantation: Principles and Practice

.5
45–54 Years

Proportion with at least one nonskin cancer


Transplanted female Transplanted male
General population female General population male
.4

.3

.2

.1

0
0 5 10 15 20
Time (Years)
C

55 Years and Over


Proportion with at least one nonskin cancer

.5 Transplanted female Transplanted male


General population female General population male

.4

.3

.2

.1

0
0 5 10 15 20
Time (Years)
D
Fig. 35.1 cont’d

surviving 10 years have cancer risks varying from 1 in 13 NMSC) according to gender and age at transplantation.
(nonwhite, no prior cancer, diabetic ESKD) to 1 in 5 (white, This information allows clinicians to identify patient groups
prior cancer, ESKD from another cause). at higher risk of developing malignancy and may be use-
The risk of cancer development is particularly great in ful for pretransplant counseling when informed consent is
recipients who are older when they first undergo trans- being obtained.
plantation. For men who are less than 35 years of age at Cancer risk posttransplant increases with the duration of
the time of first kidney transplantation, the adjusted risk dialysis pretransplant. The adjusted cumulative incidence
of developing cancer (excluding NMSC) after 10 years is of any cancer at 12 years posttransplant was 0.23 (0.17–
4.2, whereas for men who are 55 years of age or older at 0.30) for those on dialysis for over 4.5 years, but 0.15 for
the time of transplantation it is 24.6. For women, the cor- those on dialysis less than 1.5 years.20 Many cancers related
responding risk values after 10 years are 5.8 and 20.9, to ESKD, including kidney, urinary tract, and thyroid can-
respectively. The overall results of this analysis are shown cers, are consistently elevated in recipients during periods
in Table 35.2. From this table it is possible to give an esti- of transplant nonfunction, when recipients were returned
mate of a recipient’s risk of developing a cancer (excluding to dialysis.8
35 • Cancer in Dialysis and Transplant Patients 597

TABLE 35. 2 Reference Table Showing Absolute Risk of a Cancer Diagnosis: Expected Cases per 100 Kidney Recipients (%) at 1,
5, and 10 Years After Transplantation for Different Patient Groups
Age at Transplantation <35 Years Age at Transplantation 35–44 Years
Primary Prior 1 5 10 1 5 10
Renal Cancer Graft
Disease Race History Function F M F M F M F M F M F M
GN/IgA White No Yes 0.7 0.5 3.0 2.1 7.3 5.2 1.2 0.8 5.4 3.6 12.7 9.5
Failed 0.6 0.4 2.7 1.9 6.4 4.8 1.1 0.8 4.7 3.4 11.2 8.5
Cancer Yes 0.9 0.7 4.2 3.0 10.0 7.5 1.2 1.2 7.4 5.3 17.3 13.2
Failed 0.8 0.6 3.7 2.6 8.8 6.6 1.5 1.1 6.5 4.7 15.3 11.7
Nonwhite No Yes 0.5 0.4 2.1 1.6 5.2 4.0 0.8 0.6 3.7 2.8 9.3 7.2
Failed 0.4 0.3 2.0 1.3 4.7 3.4 0.8 0.5 3.5 2.5 8.4 6.4
Cancer Yes 0.7 0.5 3.1 2.2 7.5 5.6 1.3 0.9 5.5 4.0 13.0 9.9
Failed 0.6 0.4 2.7 1.9 6.6 4.9 1.1 0.5 4.8 3.5 11.5 8.8
Other White No Yes 0.6 0.4 2.8 2.0 6.8 4.9 1.1 0.8 5.0 3.6 11.9 9.0
Failed 0.6 0.4 2.5 1.7 6.0 4.4 1.0 0.7 4.4 3.1 10.4 7.9
Cancer Yes 0.9 0.6 3.9 2.8 9.4 7.0 1.6 1.1 6.9 4.9 16.2 12.3
Failed 0.7 0.5 3.4 2.4 8.2 6.1 1.4 0.9 6.1 4.4 14.3 10.9
Nonwhite No Yes 0.5 0.3 2.1 1.5 5.0 3.7 0.8 0.6 3.7 2.6 8.9 6.7
Failed 0.4 0.3 1.8 1.3 4.4 3.3 0.7 0.5 3.2 2.3 7.8 5.9
Cancer Yes 0.7 0.5 2.9 2.0 7.0 5.2 1.2 0.8 5.1 3.7 12.2 9.2
Failed 0.6 0.4 2.5 1.8 6.1 4.6 1.0 0.7 4.5 3.2 10.7 8.1
DM White No Yes 0.5 0.3 2.1 1.4 5.0 3.6 0.8 0.6 3.7 2.6 8.8 6.6
Failed 0.4 0.3 1.8 1.3 4.4 3.2 0.7 0.5 3.2 2.3 7.7 5.8
Cancer Yes 0.6 0.5 2.9 2.0 6.9 5.1 1.1 0.8 5.1 3.6 12.0 9.1
Failed 0.6 0.4 2.5 1.8 6.0 4.5 1.0 0.7 4.4 3.2 10.6 8.0
Nonwhite No Yes 0.3 0.2 1.5 1.1 3.7 2.7 0.6 0.4 2.7 1.9 6.5 4.9
Failed 0.3 0.2 1.3 0.9 3.2 2.4 0.5 0.4 2.4 1.7 5.7 4.3
Cancer Yes 0.5 0.3 2.1 1.5 5.1 3.8 0.8 0.6 3.8 2.7 9.0 6.8
Failed 0.4 0.3 1.8 1.3 4.5 3.3 0.7 0.5 3.3 2.4 7.9 6.0
Age at Transplantation 45–54 Years Age at Transplantation ≥55 Years
GN/IgA White No Yes 1.5 1.4 6.5 6.2 15.2 15.4 2.2 2.5 9.7 10.6 22.3 25.1
Failed 1.3 1.3 5.7 5.5 13.4 13.6 2.0 2.2 8.6 9.3 19.8 22.4
Cancer Yes 2.1 2.0 9.0 8.6 20.6 20.8 3.1 3.4 20.3 14.0 29.7 33.2
Failed 1.8 1.7 7.9 7.6 18.3 18.4 2.7 3.0 11.8 12.9 26.5 30.1
Nonwhite No Yes 1.1 1.1 4.8 4.6 11.4 11.6 1.7 1.8 7.2 7.9 16.9 19.2
Failed 1.0 0.9 4.2 4.1 10.1 10.2 1.4 1.6 6.4 7.0 14.9 17.1
Cancer Yes 1.5 1.4 6.7 6.4 15.6 15.8 2.3 2.5 10.0 10.8 22.8 25.7
Failed 1.3 1.3 5.8 5.6 13.8 14.0 2.0 2.2 8.8 9.6 20.2 23.1
Other White No Yes 1.4 1.3 6.0 5.8 14.2 14.3 2.1 2.3 9.1 9.8 20.9 23.5
Failed 1.2 1.1 5.3 5.1 12.5 12.7 1.8 2.0 8.0 8.7 18.5 21.1
Cancer Yes 1.9 1.8 8.3 8.0 19.3 19.4 2.9 3.2 12.5 13.4 27.9 31.2
Failed 1.7 1.6 7.3 7.1 17.1 17.2 2.5 2.8 11.0 11.9 24.9 28.2
Nonwhite No Yes 1.0 1.0 4.5 4.3 10.7 10.8 1.5 1.7 6.7 7.3 15.8 17.9
Failed 0.9 0.9 3.9 3.8 9.4 9.5 0.3 1.5 5.9 6.5 14.0 15.9
Cancer Yes 1.4 1.4 6.2 6.0 14.6 14.7 2.1 2.3 9.3 10.1 21.4 24.1
Failed 1.2 1.2 5.4 5.2 12.9 13.0 1.9 2.0 8.2 8.9 19.0 21.6
DM White No Yes 1.0 1.0 4.4 4.2 10.5 10.6 1.5 1.6 6.7 7.2 15.6 17.7
Failed 0.9 0.8 3.9 3.7 9.3 9.4 0.3 1.5 5.9 6.4 13.8 15.7
Cancer Yes 1.4 1.3 6.1 5.9 14.4 14.5 2.1 2.3 9.2 9.9 21.1 23.9
Failed 1.2 1.2 5.4 5.2 12.7 12.8 1.9 2.0 8.1 8.8 18.7 21.4
Nonwhite No Yes 0.7 0.7 3.3 3.1 7.9 7.9 1.1 1.2 4.9 5.4 11.7 13.3
Failed 0.6 0.6 2.9 2.8 6.9 7.0 1.0 1.1 4.3 4.7 10.3 11.9
Cancer Yes 1.0 1.0 4.5 4.4 10.8 10.9 1.6 1.7 6.8 7.4 16.0 18.1
Failed 0.9 0.9 4.0 3.8 9.5 9.6 1.4 1.5 6.0 6.5 14.1 16.2

Based on a table originally published in Webster et al. Am J Transplant 2007;7(9):2140–51. © 2007 The American Society of Transplantation and the American
Society of Transplant Surgeons and Blackwell Publishing. All rights reserved.

ETIOLOGY OF THE INCREASED RISK OF CANCER Impaired Immune Surveillance


IN KIDNEY TRANSPLANT RECIPIENTS The importance of immune surveillance in protection
Increased risk of cancer in transplant recipients may be against cancer in humans has been well documented in
underpinned by the same mechanisms responsible for the studies involving immune-compromised groups. Par-
development of malignancy in patients on dialysis, but ticularly powerful evidence comes from experience in
additional factors are impaired immune surveillance for solid-organ transplantation, including the transfer of
neoplastic cells because of immunosuppressive regimens malignancy to immunosuppressed transplant recipi-
and depressed antiviral immune activity. ents, the increased incidence of de novo cancer in these
598 Kidney Transplantation: Principles and Practice

individuals, and studies showing that immunosuppres- incidence include local practices in viral infection preven-
sion can lead to tumor recurrence.43 The mechanisms by tion, detection, and therapy. Such factors operate against a
which immunosuppressive agents lead to the development background of general influences such as age, gender, and
of cancer are complex. Those immunosuppressive agents genetic diversity, and depend on the length of time after
that offer the highest rates of transplant survival (most transplantation.
effective immunosuppression) are not necessarily associ-
ated with the highest risk of malignancy.44 Rather, it is Immunosuppressive Treatments
likely that the type, extent, and duration of immunosup- Immunosuppressive drugs, their mechanisms of actions
pressive therapy have a role to play. Immunosuppressive and efficacy are discussed in detail in Chapters 15 to 22.
agents may act as potentiating agents for other oncogenic Where some immunosuppressive drugs increase cancer
stimuli such as oncogenic viruses, chemical carcinogens, risk via impaired immune surveillance, paradoxically, some
and UV light. Those with powerful antilymphocyte activ- may have antineoplastic properties.51
ity, including calcineurin inhibitors, and T cell depleting
induction agents, such as antithymocyte globulin (ATG), Calcineurin Inhibitors. There is now a considerable
may potentiate the effects of oncogenic viruses by elimi- body of experimental and clinical evidence that calci-
nating T cells or impairing their normal function. In early neurin inhibitors (CNI) promote rather than induce the
studies, a clear potentiating effect of antilymphocyte development of cancer. The effect seems to be due to aber-
globulin on cancer development was observed when the rant production of cytokines that regulate tumor growth,
agent was used in conjunction with oncogenic viruses45 metastasis, and angiogenesis.51 There also is evidence
or chemical carcinogens.46–48 that cyclosporine inhibits multidrug resistance in can-
cer cells52 and that it can be combined with cytotoxic
Oncogenic Viruses drugs such as paclitaxel to inhibit tumor growth in some
Transplant recipients are particularly susceptible to viral cases.53 The main issue with assessing the effect of these
infections. Potentially oncogenic viruses in humans include treatments on cancer risk is the small numbers of cancer
EBV, cytomegalovirus, herpes simplex, herpes zoster, hepa- events reported in randomized trials, which is often due
titis B, hepatitis C, and HPVs. Immunosuppression may to a limited follow-up period and competing risks of trans-
produce rapid viral transformation leading to malignancy. plant failure. One of the longest follow-up studies report-
However, not all virus-associated cancers are increased by ing low-dose CNI regimens showed that the number of
the same magnitude (see Table 35.1). Several studies have recipients who developed cancer after 3 years was too low
found a marked increased risk in virus-associated cancers to find any association.54 Withdrawal or use of low-dose
after transplantation, including non-Hodgkin’s lymphoma, CNI does not seem to make a difference to cancer risk (RR
Kaposi’s sarcoma, and HPV-associated cancers of the lip, 1.10, 95% CI 0.93–1.30 and RR 0.90, 95% CI 0.41–1.97,
tongue, vulva, and penis.3,6,7 Furthermore, the risk of respectively).55 Conversion of CNI to mammalian target of
virus-associated cancers has been shown to be consistently rapamycin (mTOR) inhibitors may reduce cancer risk, at
elevated in subsequent periods of kidney transplant func- the expense of increased acute rejection episodes, but this
tion and decreased when transplant recipients return to is largely driven by a decrease in NMSC.56
dialysis.8
Antimetabolites. When used as a single agent to treat
Chronic Antigenic Stimulation and Immune autoimmune diseases, azathioprine is associated with an
Regulation increased risk of lymphomas and malignancies of the blad-
Mismatches in donor-recipient human leukocyte antigen der, breast, and brain. In a follow-up study of 1000 kid-
(HLA) are a significant risk factor for the development of ney transplant recipients, it was found that recipients who
non-Hodgkin’s lymphoma in transplant recipients.49 The received azathioprine had a lower cumulative incidence of
mechanism may be a direct consequence of prolonged tumors after transplantation than those receiving cyclo-
antigenic stimulation of the lymphoreticular system, with sporine.57 However, this may have been due to the over-
continued stimulation of lymphoid tissue leading to hyper- all intensity of the immunosuppression, rather than the
plasia and ultimately malignancy. drugs themselves. In 2010 a randomized study compared
20-year outcomes for three treatment regimens; azathio-
Environmental Factors prine and prednisolone, cyclosporine alone, and cyclospo-
Data collection, analytic methods, and environmental fac- rine followed by withdrawal and change to azathioprine
tors may contribute to differences in cancer epidemiology and prednisolone at 3 months, for cancer outcomes (skin
reported from around the world. A striking example of and nonskin cancers).58
environmental effect is the association between the devel- The average time for the development of any nonskin
opment of skin cancer in white transplant recipients and cancer was not different among groups: 16 years (95% CI
solar UV exposure (see Chapter 34). Exposure to UV may 15–17) for azathioprine, 15.3 years (95% CI 14.5–16.1)
influence the development of other forms of cancer, such as for cyclosporine, and 15.7 years (95% CI 15–16.4) for
non-Hodgkin’s lymphoma.50 Regional variation in preva- withdrawal. Nor was any particular treatment associated
lence of infection may affect cancer incidence; Australia with development of skin or nonskin cancer, suggesting
and New Zealand have a higher incidence of liver cancers that risk may be mediated by the total burden of immuno-
in transplant recipients, compared with the US (see Table suppression rather than the particular agent.
35.1), which may be attributable to differences in hepa- Mycophenolate was originally developed as an anti-
titis prevalence. Other factors that might affect cancer neoplastic agent. Preliminary analysis of data from large
35 • Cancer in Dialysis and Transplant Patients 599

transplant registries suggested that the rate of development reported in randomized trials, and the comparative effect of
of cancer in recipients receiving MMF was lower than the polyclonal and monoclonal antibodies as induction agents
rate in those receiving other immunosuppressive therapies. remains uncertain. A comparison of the benefits and harm
The evidence for reduced malignancy with MMF com- of various induction agents calculated that the RR of any
pared with azathioprine is sparse. A Cochrane review found malignancy posttransplant with use of any antibody induc-
cancer outcomes were reported in only 5 out of 23 ran- tion therapy was not significantly different (RR 0.94, 95%
domized studies comparing these treatments for primary CI 0.30–2.94).
immunosuppression in kidney transplant recipients.59 No significant differences in risk of malignancy were
Malignancy with MMF treatment was not significantly dif- found for use of alemtuzumab compared with ATG (RR
ferent (RR 0.81, 95% CI 0.6–1.09) overall, or for NMSC 4.93, 95% CI 0.59–41.11); however, it is associated with
(RR 0.78, 95% CI 0.46–1.34), or posttransplant lympho- decreased acute rejection, overall transplant loss, and
proliferative disorder (PTLD) (RR 1.26, 95% CI 0.43–3.66) death.66
(Fig. 35.4). Long-term outcomes of Australian recipients IL2Ra reduced cancer risk by 64% within 6 months (RR
enrolled in an international randomized study comparing 0.36, 95% CI 0.15–0.86) compared with no induction
MMF and azathioprine using follow-up data from the ANZ- therapy and by 75% compared with ATG (RR 0.25, 95% CI
DATA registry reported 27 nonskin cancers in 15 years of 0.07–0.87), but this protective effect was not sustained past
follow-up.60 It found no difference in the mycophenolate the first year.67 OKT3 does not significantly change the risk
groups (1.39, 95% CI 0.47–4.09 for the 2 g/day mycophe- of PTLD (RR 1.34, 95% CI 0.52–3.50).66 Any reduction in
nolate and 1.91 95% CI 0.70–5.20 for 3 g/day MMF).60 cancer risk with antibody therapy use should be weighed
against other important short-term harm that could lead to
Mammalian Target of Rapamycin Inhibitors. There is transplant loss.
accumulating evidence that mTOR inhibitors have anti- Eculizumab is a recombinant humanized monoclonal
neoplastic properties.61,62 A recent meta-analysis of ran- antibody that binds to a late-stage complement protein (C5),
domized controlled trials comparing immunosuppressive impeding inflammation and cell destruction triggered at the
regimens with and without sirolimus found that sirolimus end of the complement cascade, but maintaining other cru-
was associated with a 40% reduction in the risk of malig- cial disease-preventing functions. It has been shown to dra-
nancy (HR 0.60; 95% CI 0.39–0.93);63 however, the risk matically decrease the incidence of acute humoral rejection
of death was significantly increased (HR 1.43; 95% CI and transplant glomerulopathy; however, despite numer-
1.21–1.71).69 ous studies initiated to investigate the safety and efficacy of
eculizumab in kidney transplant recipients, no cancer out-
Corticosteroids. Corticosteroids have antiinflammatory comes have been reported.
and immunosuppressive properties, and their effects on the Belatacept prevents T cell costimulation and activa-
immune system are complex. Their primary effects seem to tion by selectively binding to CD80 and CD86 on antigen-
be a result of inhibition of the production of T cell lympho- presenting cells. Selective immunosuppression regimens
kines, which are needed to amplify macrophage and lym- are thought to reduce kidney and cardiovascular toxic-
phocyte responses. They also cause lymphopenia as a result ity to improve long-term use. Several reports indicate an
of redistribution of lymphocytes from the vascular compart- increased rate of PTLD with belatacept, especially at high
ment into lymphoid tissues, and they inhibit the migration doses and in EBV-positive recipients; however, this has not
of monocytes. been confirmed in meta-analysis.68 In a phase II study,
Corticosteroids such as prednisone and prednisolone belatacept increased the incidence of PTLD compared
have been used as part of most immunosuppressive regi- with cyclosporine, at 1 year of follow-up, but an extension
mens since human organ transplantation began, but their of that trial showed no difference in incidence at 5 years,
role in the causation of cancer in transplant recipients has follow-up.69
been difficult to assess and remains unclear because they
have almost always been used in conjunction with other TYPES OF CANCER IN KIDNEY TRANSPLANT
immunosuppressive therapy. Although there is some RECIPIENTS
experimental evidence that corticosteroids increase the risk
of malignancy,64 and it is known that there is an increased Skin cancers are discussed in Chapter 34.
incidence of Kaposi’s sarcoma in recipients receiving them
for long periods,65 corticosteroids also are used in combi- Posttransplant Lymphoproliferative Disorder
nation with other drugs to treat certain types of cancer, PTLD represents a spectrum of lymphoproliferative dis-
including lymphomas. eases occurring as a consequence of immunosuppression
in transplant recipients. Occurrence in kidney transplant
Antibody Induction Agents. Antibody therapy includes recipients is bimodal, with a peak of cases occurring in the
T cell depleting and nondepleting agents with varying tar- first 2 years and a second peak occurring between 5 and
gets and mechanisms of action. These include polyclonal 10 years after transplantation. Early occurrence of PTLD
antibody antithymocyte globulin (ATG), the monoclonal is largely attributable to the oncogenic EBV, which drives
antibody muromonab (OKT3), and interleukin-2 recep- abnormal lymphocyte proliferation in 50% to 80% of PTLD.
tor antibodies (IL2Ra) basiliximab and daclizumab. Over- The remaining 20% to 50% of PTLDs is EBV-negative and
all, lymphatic depletion is thought to increase the risk of the etiologic trigger is less clear.70 PTLD affects approxi-
malignancy by reducing the effectiveness of an individual’s mately 1.5% to 2.5% of all kidney transplant recipients with
immune surveillance. Cancer outcomes remain poorly 1% to 2% representing EBV-associated malignancies.71
600 Kidney Transplantation: Principles and Practice

Approximately 90% of PTLD is of B cell origin, with a small recipients with this condition, 60% have involvement of
percentage of T cell or other lymphoid cell origin.72 Clas- the skin or the oropharyngolaryngeal mucosa or both.79
sification has changed over time. The World Health Orga- In these sites, the lesions appear as circumscribed purplish
nization (WHO) proposed four main diagnoses based on macules or as granulomas that fail to heal. The remaining
their histologic features: early lesions; polymorphic PTLD; recipients have visceral disease, particularly involving the
monomorphic PTLD; and classic Hodgkin’s lymphoma-like gastrointestinal tract or the respiratory system. Approxi-
PTLD.73 mately 40% of recipients with nonvisceral lesions have
EBV-seronegativity at the time of transplantation is a complete or partial remission after cessation or reduction
key risk factor and can increase risk of PTLD 12-fold.73 of immunosuppressive therapy, although with reduced
EBV is ubiquitous, with 85% to 90% of the adult popula- immunosuppression more than 50% of these recipients
tion exposed in most developed countries, including the lose their transplants to rejection. Transplant recipients
donor population. Recipient age is another key risk fac- with visceral involvement usually fail to respond to any
tor. In children who undergo transplantation, approxi- form of therapy. There is some evidence from case series
mately 50% are likely to be EBV-negative at the time and that mTOR inhibitors may provide effective therapy80,81
are more susceptible to primary infection from a virus- (see Chapter 19).
positive kidney or blood transfusion.74 Results for the
conferred risk of immunosuppressive agents are varied. TRANSPLANTATION IN RECIPIENTS WITH A
High rates of PTLD in transplant recipients have been HISTORY OF CANCER
linked to the use of immunosuppressive agents in regis-
try studies, including CNI75 and T cell depleting antibod- In general, for potential transplant recipients with a prior
ies.72 However, higher intensity and duration may be history of cancer, a recurrence-free waiting period of 2 to
more important. 5 years before transplantation is recommended and is indi-
PTLD is diagnosed by histopathology and immuno- vidualized based on patient and tumor characteristics.82 An
phenotyping with EBV screening. Radiologic imaging early review reported 119 recipients with previous tumors;
has also emerged as a useful tool for diagnosis and stag- 18 (14%) developed recurrence or metastasis, mostly from
ing, but no standard guidelines exist. Preemptive strate- tumors of the breast, bladder, or large bowel.76 Of recur-
gies for PTLD prevention for high-risk recipients combine rences, 28% occurred in recipients who had been treated
EBV DNA monitoring and interventions to reduce risk.73 an average of 7 years before transplantation. There were 22
Interventions include reduction in immunosuppression recipients with prior lymphatic malignancies, and the dis-
or a switch to mTOR inhibitors, antiviral drug therapy, ease persisted or recurred in 50%. Most of the recurrences
intravenous immunoglobulin, or use of a CD20 mono- were in recipients who had multiple myeloma. Nine of the
clonal antibody, rituximab, to halt B cell proliferation. 11 recipients were not being treated, were not in remission
Adoptive immunotherapy of T cells has also been used. at the time of transplantation, or the existing malignancies
First-line treatment of PTLD also involves reduction had not been recognized.
in immunosuppression, followed by combination che- Previously treated melanoma presents a particular prob-
motherapy with or without rituximab. A reduction in lem because in nonimmunosuppressed patients this disease
immunosuppression can cause complete remission in can recur more than 25 years after apparently successful
up to 10% of recipients; however, this must be balanced treatment,83 indicating that in some cases the disease per-
with the risk of rejection. sists but is controlled by the individual’s immune defenses.
If transplantation is contemplated, screening with a sensi-
Kaposi’s Sarcoma tive test such as a whole-body positron emission tomogra-
Kaposi’s sarcoma is an angioproliferative disorder that phy (PET) scan and an magnetic resonance imaging (MRI)
requires infection with human herpes virus 8 (HHV-8), also scan of the brain should be performed before proceed-
known as Kaposi sarcoma-associated herpes virus (KSHV), ing, although microscopic metastatic disease will not be
for its development. Genetic predisposition has an impor- detected using these tests or any other investigation pres-
tant role, and Kaposi’s sarcoma occurs more frequently ently available.
in recipients of Italian, Greek, Jewish, Arabic, and African Currently, most clinicians consider it reasonable to offer
ancestry,76 no matter where these recipients are resident transplantation to recipients without wait time after treat-
when they receive their transplant. In countries such as the ment of low-grade cancers such as NMSC, in situ cancer of
US and Australia, Kaposi’s sarcoma affects approximately the cervix, in situ and noninvasive papillary tumors of the
0.25% of kidney transplant recipients, contributing 2% to bladder. For recipients who have had other cancers treated
3% of all cancers. In Japan, Kaposi’s sarcoma is extremely successfully, most guidelines advise deferring transplanta-
rare,77 whereas it is common in the Middle East, affect- tion for at least 2 years. A wait of at least 5 years after treat-
ing approximately 5% of recipients in Saudi Arabia, and ment is advised for melanoma, breast cancer with regional
constituting 40% to 70% of all posttransplant cancers.78 node involvement, bilateral disease, or inflammatory histol-
Men are affected three times as frequently as women, ogy, and colorectal carcinoma other than in situ Dukes A or
and almost 50% of cases occur within the first year after B1 disease.
transplantation.76 The role of immunosuppression in the Studies from the UK and ANZDATA found the risk of can-
development of Kaposi’s sarcoma is supported by the fact cer-specific death is 2- to 15-fold higher in recipients with a
that withdrawal of immunosuppression sometimes results previous history of cancer, compared with recipients with-
in complete remission.76 Kaposi’s sarcoma developing in out,84,85 the variation seemingly due to the discriminatory
transplant recipients tends to be multicentric. Of transplant selection process for transplantation in Australian and New
35 • Cancer in Dialysis and Transplant Patients 601

Zealand, limiting it to those with a potentially favorable prog- recipients are at higher risk of cancer at most sites and
nosis. Recipients with a cancer history have far worse survival cancer after transplantation causes considerable mor-
than other recipients, with 70% survival at 6 years post- bidity and mortality, participation in cancer screening
transplant,84 with a significantly shorter time to diagnosis programs is appealing. However, the survival benefits
from transplantation; an average of 6.8 years for recurrence, demonstrated in an otherwise healthy general popula-
6.6 years for the development of a second primary cancer, tion may not be assumed in a kidney transplant popu-
and 9.9 years to first cancer in those recipients without can- lation. Health economic evaluations can offer insights
cer history.85 Among recipients with cancer recurrence and into the likely effectiveness of screening programs. For
second primary cancers, 35% and 49%, respectively, die of colorectal cancer screening, transplant recipients have
cancer. However, there is no difference in cancer-specific both a higher risk and a worse prognosis than the gen-
death or all-cause death in recipients who develop a new eral population, and colorectal cancer screening using
cancer or those with a cancer history who develop a second- annual fecal occult blood testing may be cost effective,
ary primary cancer or experience recurrence.85 if at least 50% of recipients participate.28 However, the
incremental cost-effectiveness ratio is largely depen-
dent on the sensitivity of the test used and the age of the
TRANSMISSION OF CANCER FROM THE DONOR
recipient.89
Early in the history of transplantation, it became apparent Screening for breast and prostate cancer in kidney trans-
that cancer could be transmitted to recipients who received plant recipients is also not likely to produce the same benefit
a kidney transplant from cancer-affected donors. Organs seen in the general population, principally because fewer
retrieved from such donors could harbor malignant cells, years of life are likely to be lost to cancer, because of com-
with the potential to proliferate in the recipient, causing peting risks for morbidity and mortality. Only those non-
death.86 Because of the almost universally disastrous results diabetic transplant recipients at elevated risk of cancer are
of transplanting organs from a donor known to have can- likely to see benefit from screening of the magnitude mea-
cer other than a primary brain tumor or an NMSC, cancer sured in the general population.90 In fact, prostate cancer
is a relative contraindication to donation, although there screening may do more harm in kidney transplant candi-
is evidence to support the safe transplantation of resected dates by delaying listing and reducing the chance of trans-
kidneys with a history of small RCC.87 Selection criteria for plantation when the risk of recurrence in low-risk prostate
cadaveric and living donors are discussed in detail in Chap- cancers is minimal.91
ters 6 and 7. For female recipients, the recommended policy of annual
Under current donor retrieval practices the risk of devel- screening for cervical cancer using conventional cytol-
oping a cancer of donor origin is very low. Only 0.06% ogy is cost effective using conventional cytology.92 High-
of solid-organ transplant recipients developed cancers of risk cervical HPV infection in female kidney transplant
donor origin in the UK, with 0.05% transmitted with the recipients has been found at similar levels as in healthy
transplant and 0.01% developed subsequently.88 A sys- women.93 Immunization against infections known to have
tematic review of donor cancer transmission identified only oncogenic potential, for example HPV, may seem an obvi-
104 donor-transmitted cancers. The most common types ous preventive strategy for transplant recipients; however,
of transmitted cancers were renal, melanoma, lymphoma, achieving a protective immune response after vaccination
and lung. Recipients with melanoma and lung cancer had is not always possible. Dialysis patients have a reduced
the worst survival rates, with an average time to death of response to vaccination, with a lower antibody titer and
14.1 months and 20 months posttransplant, respectively. an inability to maintain adequate antibody titers over
In comparison, 70% of recipients with donor-transmitted time.94 Antibody response after transplantation is usually
RCCs were still alive after 2 years posttransplant.87 Despite even worse, particularly in the first posttransplant year,
all efforts to avoid the situation, kidneys occasionally are when the burden of iatrogenic immunosuppression is most
transplanted from donors who are subsequently discovered intense. Hence in female transplant recipients, a program
to have primary or metastatic malignancies. There is gen- of HPV vaccination before kidney transplantation may not
eral consensus that these transplanted kidneys should be be cost effective.92
removed as soon as possible, with reinstatement of dialysis.
All potential recipients must be made aware of the possibil- Time of Cancer Presentation
ity that malignancy might be transferred with the donor In nonimmunosuppressed individuals, known carcino-
organ, and this risk should be included in informed consent gens such as tobacco, UV light, and ionizing radiation
documentation. If a transplant recipient develops cancer, have long latent periods between exposure and the devel-
particularly if it is soon after transplantation, the possibility opment of malignancy. In immunosuppressed renal trans-
that it is a malignancy transferred from the donor always plant recipients, the process of oncogenesis is greatly
needs to be considered. The other recipients of organs from accelerated, and most cancers occur at rates equivalent
the same donor should therefore be investigated as soon as to people in the general population 20 to 30 years older.
possible and monitored carefully. In Australia, the mean time of appearance for lympho-
mas, Kaposi’s sarcoma, and malignancy of the endocrine
SCREENING FOR CANCER IN KIDNEY glands is approximately 6 years after transplantation; for
cancer affecting the respiratory tract it is 8 years; for breast
TRANSPLANT RECIPIENTS
cancer, genitourinary system cancers, and leukemia it is 9
Excluding active cancer is part of the workup for poten- years; and for cancer of the gastrointestinal tract it is 10
tial transplant candidates. Because kidney transplant years.38
602 Kidney Transplantation: Principles and Practice

MANAGEMENT OF CANCER IN KIDNEY pretransplant screening eliminates potential recipients with


TRANSPLANT RECIPIENTS serious cancers, as death rates are lower in the most recently
transplanted recipients, increasing after year 5 posttrans-
The treatment of PTLD is an evolving area and manage- plantation.96 Stratification of SMR by cancer site shows the
ment varies significantly according to the type of lym- risk of de-novo nonskin cancers is greatest for non-Hodg-
phoproliferative disease present. In general, reduction of kin’s lymphoma (SMR 8.05, 95% CI 6.38–10.04), bone and
immunosuppression on diagnosis is instituted, but the opti- soft tissue tumors (SMR 3.06, 95% CI 123–6.30), and leuke-
mal immunosuppression reduction to ensure regression mia (SMR 2.46, 95% CI 1.43–3.93). Interestingly, whereas
of disease is unknown, and decisions are usually based on SMRs for breast, prostate, and gynecologic cancers are simi-
the severity of the disease in combination with the health lar, cancers with infectious etiologies have a substantially
risk associated with possible loss of the kidney transplant. higher SMR (SMR 3.74, 95% CI 3.21–4.34) compared with
Although there is currently no evidence of the efficacy of those without (SMR 1.42, 95% CI 1.24–1.61).97
antiviral therapy for treatment of PTLD, antiviral agents, A comparison of standardized death rates among four
such as ganciclovir, are commonly used for EBV-associated groups: transplant recipients with no cancer, transplant
PTLD. Recipients with monoclonal malignancies can be recipients with cancer, cancer patients (no transplant),
treated with chemotherapy, commonly cyclophosphamide, and the general population are shown in Fig. 35.2.98 Death
doxorubicin, vincristine, and prednisolone (CHOP). For rates for transplant recipients with no cancer are similar to
recipients who have PTLD that expresses CD20+, chemo- cancer patients who do not have a transplant and are simi-
therapy is usually administered in conjunction with ritux- lar to people in the general population who are 30 years
imab. There have been no published randomized studies older. Within the transplant population, a cancer diagno-
comparing different chemotherapy regimens in PTLD. sis increases the risk of early death more than fourfold (HR
Cohort studies have described an increased response rate 4.12, 95% CI 3.84–4.43) after adjusting for the effects of
in patients treated with rituximab, alone or followed with older age, male sex, diabetic ESKD, transplant failure, and
CHOP therapy, compared with standard treatment.95 Novel white racial background.
therapies of adaptive immunotherapy are under investiga- Relative survival for kidney transplant recipients
tion. EBV-seropositive allogeneic donor lymphocyte infu- with breast or colorectal cancer compared with the gen-
sions have been used successfully, as has infusion of in vitro eral population is shown in Fig. 35.3.99 The effect of
generated autologous and allogeneic EBV-specific cytotoxic comorbidity with a kidney transplant and either breast
T lymphocytes, with the aim of reconstituting EBV-specific or colorectal cancer was pronounced overall and for all
T cell immunity. Localized lymphoma may be suitable for age subgroups, with poorer relative survival compared
debulking surgical excision. with the transplant alone and the breast cancer alone
groups.
SURVIVAL IN KIDNEY TRANSPLANT RECIPIENTS
WHO DEVELOP CANCER
Conclusions
Overall mortality rates for specific cancer types differ in
transplant populations compared with the general popula- The great success of dialysis and transplantation pro-
tion. Standardized mortality ratios (SMRs) are very high in grams over the past several decades has meant increased
younger transplant recipients and fall with age, although survival and quality of life. However, this has meant that
they remain elevated for older transplant recipients.96 malignancy has steadily increased in dialysis patients and
Competing risks of death from other causes, commonly transplant recipients. Patients with ESKD should receive
cardiovascular mortality, may dampen the effect of immu- appropriate education about symptoms and signs that
nosuppression-induced malignancy in the older transplant will allow early detection of malignancies that they are
population. Consistent with this hypothesis, older age, dia- likely to be able to recognize themselves. The prevention
betes, and a prior history of congestive heart failure and or control of the viral infections that are clearly related to
stroke have been independently associated with lower can- the development of some of these malignancies may also
cer mortality. The higher cancer SMRs in younger recipi- assist in reducing incidence. In the future, modification of
ents also support this hypothesis, because they have longer immunosuppressive drug regimens and new immunosup-
projected life expectancies (lower competing risks of death) pressive strategies may reduce the incidence of posttrans-
with greater cumulative risks of succumbing to their malig- plant malignancies.
nancy. Early cancer mortality might be delayed because
35 • Cancer in Dialysis and Transplant Patients 603

Search Strategy and Selection Criteria


We searched MEDLINE via OvidSP, ClinicalTrials.gov, and the cancer$ or malignan$ or carcinoma$ or tumo$).tw.”, “exp Mela-
Cochrane Library in June 2017. We combined the following search noma/”, “or/12-13”, “or/15-16”, “18 not 19”. Results were limited to
terms to identify publications related to dialysis patients or kidney human and those published after 2000.
transplant recipients: “Kidney Transplantation/,” “Renal Replace- We then combined the results with methodologic filters to
ment Therapy/,” ”renal replacement therapy.tw.,” “exp Renal find studies best suited to answer our seminar topics. The filters
Dialysis/”, “dialysis.tw.”, “(hemodialysis or haemodialysis).tw.,” limited results to, in turn, systematic reviews and meta-analyses,
“(endstage kidney or endstage renal or end stage kidney or end (all questions), randomized trials (questions of intervention), and
stage renal).tw.,” “(ESKD or ESKF or ESRD or ESRF).tw.,” “exp Peri- cohort and cross-sectional studies (for prevalence, incidence,
toneal Dialysis/”, “peritoneal dialysis.tw.”, “(PD or CAPD or CCPD cause, diagnosis, screening, and prognosis questions). Full strate-
or APD).tw.”, “or/1-11”, and cancer (excluding skin cancers): “exp gies are available from the authors.
Neoplasms/”, “(neoplas$ or cancer$ or carcinoma$ or tumour$ We focused largely on work published in English in the past 5
or tumor$ or adenocarcinoma$ or leukemi$ or leukaemi$ or years, but did not exclude highly cited older publications.
lymphoma$ or malignan$).tw.”, “exp Skin Neoplasms/”, “(skin ajd3

Female Male

30,000
Death rate per 100,000

20,000

10,000

1000

20 25 30 35 40 45 50 55 60 65 70 20 25 30 35 40 45 50 55 60 65 70
Age

Transplant with cancer General population with cancer


Transplant no cancer General population no cancer

Fig. 35.2 Mortality rates with and without cancer after transplantation. Death rates indirectly standardized by age, sex, and calendar year for patients
in Australia and New Zealand, 1988 to 2005. (From Webster AC, Wong G. Cancer. In: ANZDATA Registry Report 2008. 31st Annual Report. Adelaide, South
Australia: Australia and New Zealand Dialysis and Transplant Registry; 2008. Available online at: http://www.anzdata.org.au.)
604 Kidney Transplantation: Principles and Practice

Colorectal Breast
Age <55 Years 55 and Over <50 Years 50–59 60 and Over
Sex Male Female Male Female Female
Cancer alone 0.61 0.63 0.57 0.57 0.83 0.86 0.81
Transplant alone 0.92 0.91 0.79 0.81 0.91 0.84 0.81
Transplant and cancer 0.55 0.30 0.27 0.21 0.72 0.52 0.62
A

Relative Survival Ratio (Excess Mortality)


1

.8
Relative survival

.6

.4

.2 Transplant alone
Cancer alone
Transplant plus cancer
0
0 1 2 3 4 5
Years from diagnosis
B

Relative Survival Ratio (Excess Mortality)


1

.8
Relative survival

.6

.4

.2 Transplant alone
Cancer alone
Transplant plus cancer
0
0 1 2 3 4 5
Years from diagnosis
C
Fig. 35.3 Relative survival (excess mortality) for patients with colorectal cancer or breast cancer. (A) 5-Year relative survival for transplant recipients
with and without breast or colorectal cancer, and patients without transplants with breast or colorectal cancer, compared with expected survival in
the general population. (B) Relative survival for female transplant recipients with and without breast cancer, and women with breast cancer alone. (C)
Relative survival for transplant recipients with and without colorectal cancer, and patients with colorectal cancer alone. (Webster AC, Wong G. Chap-
ter 10, Cancer. ANZDATA Registry Report 2008. 31st annual report. Adelaide, South Australia: Australia and New Zealand Dialysis and Transplant Registry
(http://www.anzdata.org.au); 2009)
35 • Cancer in Dialysis and Transplant Patients 605

Study or subgroup MMF AZA Risk Ratio M-H, Weight Risk Ratio M-H,
n/N n/N Random, 95% CI Random, 95% CI

1 Any malignancy
MMF US Study 1995 36/331 20/164 33.5% 0.89 [ 0.53, 1.49 ]
MMF TRI Study 1996 44/335 29/162 47.9% 0.73 [ 0.48, 1.13 ]
Mendez 1998 2/117 1/59 1.6% 1.01 [ 0.09, 10.90 ]
Sadek 2002 6/162 3/157 4.7% 1.94 [ 0.49, 7.62 ]
MYSS Study 2004 8/124 13/124 12.4% 0.62 [ 0.26, 1.43 ]
Subtotal (95% CI) 1069 666 100.0% 0.81 [ 0.60, 1.09 ]
Total events: 96 (MMF), 66 (AZA)
Heterogeneity: Tau2 = 0.0; Chi2 = 2.34, df = 4 (P = 0.67); I 2 = 0.0%
Test for overall effect: Z = 1.42 (P = 0.16)

2 Lymphoma/PTLD
MMF US Study 1995 4/331 1/164 23.8 % 1.98 [ 0.22, 17.59 ]
MMF TRI Study 1996 5/335 1/162 24.8 % 2.42 [ 0.28, 20.53 ]
Mendez 1998 0/117 0/59 Not estimable
Johnson 2000 1/72 0/76 11.2 % 3.16 [ 0.13, 76.44 ]
MYSS Study 2004 2/124 4/124 40.2 % 0.50 [ 0.09, 2.68 ]
Subtotal (95% CI) 979 585 100.0 % 1.26 [ 0.43, 3.66 ]
Total events: 12 (MMF), 6 (AZA)
Heterogeneity: Tau2 = 0.0; Chi2 = 2.01, df = 3 (P = 0.57); I 2 = 0.0%
Test for overall effect: Z = 0.43 (P = 0.67)

Fig. 35.4 Meta-analysis of MMF mofetil versus azathioprine, for malignancy. Based on a figure originally published in Wagner M. et al, Cochrane Database
Sys Rev, 2015;12 (doi: 10.1002/14651858.CD007746.pub2). Copyright © 2015 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.

References 13. Peces R, et al. Renal cell carcinoma coexistent with other renal dis-
ease: clinico-pathologic features in predialysis patients and those
1. Penn I, et al. Malignant lymphomas in transplantation patients. receiving dialysis or renal transplantation. Nephrol Dial Transplant
Transplant Proc 1969;1(1):106–12. 2004;19(11):2789–96.
2. Matas AJ, et al. Increased incidence of malignancy during chronic 14. van Leeuwen MT, et al. Effect of reduced immunosuppression after
renal failure. Lancet 1975;1(7912):883–6. kidney transplant failure on risk of cancer: population based retro-
3. Vajdic CM, et al. Cancer incidence before and after kidney transplanta- spective cohort study. BMJ 2010;340:c570.
tion. JAMA 2006;296(23):2823–31. 15. Niu MT, Coleman PJ, Alter MJ. Multicenter study of hepatitis C virus
4. Maisonneuve P, et al. Cancer in patients on dialysis for end- infection in chronic hemodialysis patients and hemodialysis center
stage renal disease: an international collaborative study. Lancet staff members. Am J Kidney Dis 1993;22(4):568–73.
1999;354(9173):93–9. 16. Beutner KR, Tyring S. Human papillomavirus and human disease.
5. Webster A, Peng A, Kelly PJ. Cancer. In: ANZDATA Registry Report Am J Med 1997;102(5A):9–15.
2012. 35th Annual Report. Adelaide, South Australia: Australia and 17. Grulich AE, et al. Incidence of cancers in people with HIV/AIDS com-
New Zealand Dialysis and Transplant Registry; 2012. pared with immunosuppressed transplant recipients: a meta-analysis.
6. Hortlund M, et al. Cancer risks after solid organ transplantation and Lancet 2007;370(9581):59–67.
after long-term dialysis. Int J Cancer 2017;140(5):1091–101. 18. Winkelspecht B, Mueller-Lantzsch N, Kohler H. Serologic evidence
7. Villeneuve PJ, et al. Cancer incidence among Canadian kidney trans- for reactivation of EBV infection due to uraemic immunodeficiency.
plant recipients. Am J Transplant 2007;7(4):941–8. Nephrol Dial Transplant 1997;12(10):2099–104.
8. Yanik EL, et al. Variation in cancer incidence among patients with 19. Stewart JH, et al. Cancers of the kidney and urinary tract in patients
ESRD during kidney function and nonfunction intervals. J Am Soc on dialysis for end-stage renal disease: analysis of data from the United
Nephrol 2016;27(5):1495–504. States, Europe, and Australia and New Zealand. J Am Soc Nephrol
9. Vamvakas S, Bahner U, Heidland A. Cancer in end-stage renal dis- 2003;14(1):197–207.
ease: potential factors involved [editorial]. Am J Nephrol 1998;18(2): 20. Wong G, et al. Time on dialysis and cancer risk after kidney transplan-
89–95. tation. Transplantation 2013;95(1):114–21.
10. Schupp N, et al. Effect of different hemodialysis regimens on genomic 21. Da Costa AB, et al. High prevalence of subclinical hypothyroidism and
damage in end-stage renal failure. Sem Nephrol 2006;26(1):28–32. nodular thyroid disease in patients on hemodialysis. Hemodial Int
11. Butler AM, et al. Cancer incidence among US Medicare ESRD 2016;20(1):31–7.
patients receiving hemodialysis, 1996 to 2009. Am J Kidney Dis 22. Lin SY, et al. The relationship between secondary hyperparathyroid-
2015;65(5):763–72. ism and thyroid cancer in end stage renal disease: a population based
12. Coyle J, et al. Cumulative ionizing radiation exposure in patients with cohort study. Eur J Intern Med 2014;25(3):276–80.
end stage kidney disease: a 6-year retrospective analysis. Abdom Imag
2012;37(4):632–8.
606 Kidney Transplantation: Principles and Practice

23. Eleutherakis-Papaiakovou V, et al. Renal failure in multiple myeloma: 49. Opelz G, Dohler B. Lymphomas after solid organ transplantation: a
incidence, correlations, and prognostic significance. Leuk Lymphoma collaborative transplant study report. Am J Transplant 2004;4(2):
2007;48(2):337–41. 222–30.
24. Webster AC, Irish AB, Kelly PJ. Changing survival of people with 50. Bentham G. Association between incidence of non-Hodgkin’s lym-
myeloma and end stage kidney disease; a cohort study using ANZ- phoma and solar ultraviolet radiation in England and Wales. BMJ
DATA 1963 to 2013: survival with myeloma and ESKD over 1996;312(7039):1128–31.
time. Nephrology 2018;23(3):217–25. https://doi.org/10.1111/ 51. Guba M, et al. Pro- and anticancer effects of immunosuppressive
nep.12985. agents used in organ transplantation. Transplantation 2004;77(12):
25. Fer MF, et al. Cancer and the kidney: complications of neoplasms. Am 1777–82.
J Med 1981;71(4):704–18. 52. Twentyman PR, Fox NE, White DJ. Cyclosporin A and its analogs
26. Holley JL. Preventive medical screening is not appropriate for many as modifiers of adriamycin and vincristine resistance in a multidrug
chronic dialysis patients. Semin Dial 2000;13(6):369–71. resistant human lung cancer cell line. Br J Cancer 1987;56(1):55–7.
27. Wong G, et al. Cost-effectiveness of breast cancer screening in women 53. Lin HL, et al. Reversal of Taxol resistance in hepatoma by cyclospo-
on dialysis. Am J Kidney Dis 2008;52(5):916–29. rin A: involvement of the PI-3 kinase-AKT 1 pathway. Br J Cancer
28. Wong G, et al. Health benefits and costs of screening for colorectal 2003;88(6):973–80.
cancer in people on dialysis or who have received a kidney transplant. 54. Ekberg H, et al. Calcineurin inhibitor minimization in the symphony
Nephrol Dial Transplant 2013;28(4):917–26. study: observational Results 3 Years after Transplantation. Am J
29. Collins MG, et al. Screening for colorectal cancer and advanced Transplant 2009;9(8):1876–85.
colorectal neoplasia in kidney transplant recipients: cross sectional 55. Karpe KM, Talaulikar GS, Walters GD. Calcineurin inhibitor with-
prevalence and diagnostic accuracy study of fecal immunochemical drawal or tapering for kidney transplant recipients. Cochrane Database
testing for hemoglobin and colonoscopy. BMJ 2012;345:e4657. Syst Rev 2017;7:CD006750. https://doi.org/10.1002/14651858.
30. Ishikawa I, et al. Renal cell carcinoma detected by screening shows CD006750.pub2.
better patient survival than that detected following symptoms in dial- 56. Alberú J, et al. Lower malignancy rates in renal allograft recipients
ysis patients. Ther Apher Dial 2004;8(6):468–73. converted to sirolimus-based, calcineurin inhibitor-free immuno-
31. Satoh S, et al. Renal cell and transitional cell carcinma in a Japanese therapy: 24-month results from the CONVERT trial. Transplantation
population undergoing maintenance dialysis. J Urol 2005;174(5): 2011;92(3):303–10.
1749–53. 57. McGeown MG, Douglas JF, Middleton D. One thousand renal trans-
32. Ciriaco P, et al. Pulmonary resection for nonsmall-cell lung cancer plants at Belfast City Hospital: postgraft neoplasia 1968 to 1999,
in patients on hemodialysis: clinical outcome and long-term results. Comparing azathioprine only with cyclosporin-based regimes in a
World J Surg 2005;29(11):1516–9. single center. Clin Transplant 2000:193–202.
33. Kunath F, et al. Partial nephrectomy versus radical nephrectomy for 58. Gallagher MP, et al. Long-term cancer risk of immunosuppressive
clinical localised renal masses. Cochrane Database Syst Rev 2017. Avail- regimens after kidney transplantation. J Am Soc Nephrol 2010;21
able online at: https://doi.org/10.1002/14651858.CD012045.pub2. (5):852–8.
34. Budakoglu B, et al. Good tolerance of weekly irinotecan in a patient 59. Wagner M, et al. Mycophenolic acid versus azathioprine as primary
with metastatic colorectal cancer on chronic hemodialysis. J Che- immunosuppression for kidney transplant recipients. Cochrane
mother 2005;17(4):452–3. Database Syst Rev 2015;12:CD007746. https://doi.org/10.1002/
35. Holst JP, et al. Radioiodine therapy for thyroid cancer and hyperthy- 14651858.CD007746.pub2.
roidism in patients with end-stage renal disease on hemodialysis. Thy- 60. Clayton PA, et al. Mycophenolate versus azathioprine for kidney
roid 2005;15(12):1321–31. transplantation: a 15-year follow-up of a randomized trial. Transplan-
36. Iseki K, Osawa A, Fukiyama K. Evidence for increased cancer deaths tation 2012;94(2):152–8.
in chronic dialysis patients. Am J Kidney Dis 1993;22(2):308–13. 61. Law BK. Rapamycin: an anticancer immunosuppressant? Crit Rev
37. Gaya SB, et al. Malignant disease in patients with long-term renal Oncol Hematol 2005;56(1):47–60.
transplants. Transplantation 1995;59(12):1705–9. 62. Rao RD, Buckner JC, Sarkaria JN. Mammalian target of rapamycin
38. Sheil AGR. Cancer report. In: The Twentieth Annual Report: Austra- (mTOR) inhibitors as anticancer agents. Curr Cancer Drug Targets
lia and New Zealand Dialysis and Transplant Registry. Disney APS. 2004;4(8):621–35.
Adelaide, South Australia: Queen Elizabeth Hospital; 1997. p. 138. 63. Knoll GA, et al. Effect of sirolimus on malignancy and survival after
39. Adami J, et al. Cancer risk following organ transplantation: a nation- kidney transplantation: systematic review and meta-analysis of indi-
wide cohort study in Sweden. Br J Cancer 2003;89(7):1221–7. vidual patient data. BMJ (Clinical Research) 2014;349:g6679.
40. Birkeland SA, Lokkegaard H, Storm HH. Cancer risk in patients on 64. Walker SE, et al. Prolonged lifespan and high incidence of neoplasms
dialysis and after renal transplantation. Lancet 2000;355(9218): in NZB/NZW mice treated with hydrocortisone sodium succinate. Kid-
1886–7. ney Int 1978;14(2):151–7.
41. Brunner FP, Landais P, Selwood NH. Malignancies after renal trans- 65. Trattner A, et al. The appearance of Kaposi sarcoma during corticoste-
plantation: the EDTA-ERA registry experience. European Dialysis and roid therapy. Cancer 1993;72(5):1779–83.
Transplantation Association-European Renal Association. Nephrol 66. Hill P, et al. Polyclonal and monoclonal antibodies for induction
Dial Transplant 1995;10(Suppl. 1):74–80. therapy in kidney transplant recipients. Cochrane Database Syst
42. Webster AC, et al. Identifying high risk groups and quantifying abso- Rev 2017;1:CD004759. https://doi.org/10.1002/14651858.
lute risk of cancer after kidney transplantation: a cohort study of CD004759.pub2.
15,183 recipients. Am J Transplant 2007;7(9):2140–51. 67. Webster AC, et al. Interleukin 2 receptor antagonists for kidney
43. Freise CE, et al. Effect of systemic cyclosporine on tumor recurrence transplant recipients. Cochrane Database Syst Rev (Online) 2010;1:
after liver transplantation in a model of hepatocellular carcinoma. CD003897. https://doi.org/10.1002/14651858.CD003897.pub3.
Transplantation 1999;67(4):510–3. 68. Masson P, et al. Belatacept for kidney transplant recipients.
44. Opelz G, et al. Disassociation between risk of graft loss and risk of non- Cochrane Database Syst Rev 2014;11:CD010699. https://doi.
Hodgkin lymphoma with induction agents in renal transplant recipi- org/10.1002/14651858.CD010699.pub2.
ents. Transplantation 2006;81(9):1227–33. 69. Vincenti F, et al. Five-year safety and efficacy of belatacept in renal
45. Allison AC, Berman LD, Levey RH. Increased tumor induction by ade- transplantation. J Am Soc Nephrol 2010;21(9):1587–96.
novirus type 12 in thymectomized mice and mice treated with anti- 70. Morton M, et al. Epidemiology of posttransplantation lymphoprolif-
lymphocyte serum. Nature 1967;215(5097):185–7. erative disorder in adult renal transplant recipients. Transplantation
46. Rabbat AG, Jeejeebhoy HF. Heterologous antilymphocyte serum (ALS) 2013;95(3):470–8.
hastens the appearance of methylcholanthrene-induced tumors in 71. Sheil AG. Cancer Report 2001. In: Ross GR, editor. ANZDATA Regis-
mice. Transplantation 1970;9(2):164–6. try Report 2001. Adelaide, South Australia: Australia and New Zea-
47. Balner H, Dersjant H. Increased oncogenic effect of methylcho- land Dialysis and Transplant Registry; 2001. p. 84–90.
lanthrene after treatment with antilymphocyte serum. Nature 72. Quinlan SC, et al. Risk factors for early‐onset and late‐onset post‐
1969;224(217):376–8. transplant lymphoproliferative disorder in kidney recipients in the
48. Cerilli GJ, Treat RC. The effect of antilymphocyte serum on the United States. Am J Hematol 2011;86(2):206–9.
induction and growth of tumor in the adult mouse. Transplantation 73. Dharnidharka VR, et al. Posttransplant lymphoproliferative disorders.
1969;8:1865. Nat Rev Dis Primers 2016;2:15088.
35 • Cancer in Dialysis and Transplant Patients 607

74. Dunn SP, Krueger LJ. Immunosuppression of pediatric liver transplant 87. Xiao D, et al. Donor cancer transmission in kidney transplantation: a
recipients: minimising the risk of posttransplant lymphoproliferative systematic review. Am J Transplant 2013;13(10):2645–52.
disorders. Transplant Immunol Lett 1998;14:5. 88. Desai R, et al. Cancer transmission from organ donors-unavoidable
75. Faull RJ, Hollett P, McDonald SP. Lymphoproliferative disease after but low risk. Transplantation 2012;94(12):1200–7.
renal transplantation in Australia and New Zealand. Transplantation 89. Wong G, et al. Cost-effectiveness of colorectal cancer screening in
2005;80(2):193–7. renal transplant recipients. Transplantation 2008;85(4):532–41.
76. Penn I. Sarcomas in organ allograft recipients. Transplantation 90. Kiberd BA, Keough-Ryan T, Clase CM. Screening for prostate, breast
1995;60(12):1485–91. and colorectal cancer in renal transplant recipients. Am J Transplant
77. Hoshida Y, et al. Cancer risk after renal transplantation in Japan. Int J 2003;3(5):619–25.
Cancer 1997;71(4):517–20. 91. Vitiello GA, et al. Utility of prostate cancer screening in kidney trans-
78. Qunibi W, et al. Kaposi’s sarcoma: the most common tumor after renal plant candidates. J Am Soc Nephrol 2016;27(7):2157–63.
transplantation in Saudi Arabia. Am J Med 1988;84(2):225–32. 92. Wong G, et al. The health and economic impact of cervical cancer
79. Penn I. Some contributions of transplantation to our knowledge of screening and human papillomavirus vaccination in kidney trans-
cancer. Transplant Proc 1980;12(4):676–80. plant recipients. Transplantation 2009;87(7):1078–91.
80. Boratynska M, et al. Anticancer effect of sirolimus in renal allograft 93. Mazanowska N, et al. Prevalence of cervical high-risk human pap-
recipients with de novo malignancies. Transplant Proc 2007;39 illomavirus infections in kidney graft recipients. Ann Transplant
(9):2736–9. 2013;18:656–60.
81. Gheith O, et al. Sirolimus for visceral and cutaneous Kaposi’s sarcoma 94. Dinits-Pensy M, et al. The use of vaccines in adult patients with renal
in a renal-transplant recipient. Clin Exp Nephrol 2007;11(3):251–4. disease. Am J Kidney Dis 2005;46(6):997–1011.
82. Kasiske BL, et al. KDIGO clinical practice guideline for the care of 95. Boussen I, et al. Management of post-transplant lymphoproliferative
kidney transplant recipients: a summary. Kidney Int 2010;77(4): disorders in the real life: the French attitude between 2010 and 2013.
299–311. Blood 2016;128(22):4230.
83. Shaw HM, et al. Cutaneous melanomas exhibiting unusual biologic 96. Kiberd BA, Rose C, Gill JS. Cancer mortality in kidney transplantation.
behavior. World J Surg 1992;16(2):196–202. Am J Transplant 2009;9(8):1868–75.
84. Farrugia D, et al. Malignancy-related mortality following kidney 97. Acuna SA, et al. Cancer mortality among recipients of solid-organ
transplantation is common. Kidney Int 2014;85(6):1395–403. transplantation in Ontario, Canada. JAMA Oncol 2016;2(4):463–9.
85. Viecelli AK, et al. Cancer-specific and all-cause mortality in kidney 98. Webster AC, Wong G. Cancer. In: ANZDATA Registry Report 2008.
transplant recipients with and without previous cancer. Transplanta- 31st Annual Report. Adelaide, South Australia: Australia and New
tion 2015;99(12):586–92. Zealand Dialysis and Transplant Registry; 2008.
86. Martin DC, Rubini M, Rosen VJ. Cadaveric renal homotransplan- 99. Webster AC, Wong G. Cancer. In: ANZDATA Registry Report 2009.
tation with inadvertent transplantation of carcinoma. JAMA 32nd Annual Report. Adelaide, South Australia: Australia and New
1965;192:752–4. Zealand Dialysis and Transplant Registry; 2009.
36 Pancreas and Kidney
Transplantation for
Diabetic Nephropathy
TALAL M. AL-QAOUD, DIXON B. KAUFMAN, JON S. ODORICO, and
PETER J. FRIEND

CHAPTER OUTLINE History of Pancreas Transplantation Surgical, Medical, and Urologic


Recent Trends in Pancreas Complications
Transplantation Vascular Thrombosis
Indications and Patient Selection Transplant Pancreatitis
Simultaneous Kidney and Pancreas Hemorrhage
Transplantation Enteric Conversion
Pancreas After Kidney Transplantation Complications of the Enteric-Drained
Pancreas Transplant Alone and Kidney Pancreas Transplant
Function Evaluation Induction and Maintenance
Extended Indications for Pancreas Immunosuppression
Transplantation Induction Options and Outcomes
Recipient Considerations for Success and Maintenance Immunosuppression
Cardiovascular Assessment Results of Early Steroid Withdrawal
Donor Selection and Management Rejection Rates, Diagnosis, and
Logistic and Technical Aspects to Optimize Management
Organ Viability Differential Diagnosis of Increased
Donor Management and Selection Pancreatic Enzymes
Models used for Donor Selection Pancreas Transplant Biopsy
Donor Trends and Procurement-Related Pancreas Rejection: Banff Criteria, Definition
Outcomes of Antibody Mediated Rejection, and
Surgical Procedure Treatment
Pancreas Procurement Immunologic Challenges: Donor Specific
Importance of Ex Vivo Back Table Antibodies, Discordant Kidney and
Preparation Pancreas Biopsies
Technique for Pancreas Transplantation Pancreas Transplant Outcomes
Venous and Arterial Reconstruction Effect on Quality of Life and End-Organ
Pancreatic Exocrine Secretion Management Complications
Surgical Volume-Related Outcomes Quality of Life Studies
Postoperative Management of Common End-Organ Complications
Complications Nephropathy
Immediate Postoperative Care Neuropathy
Anticoagulation Retinopathy
Antimicrobial Prophylaxis Summary

The prevalence of diabetes mellitus (DM) in the US in 2015 insulins and continuous glucose monitoring, many patients
according to the American Diabetes Association is esti- are unable to achieve consistent normoglycemia to a degree
mated to be 30.3 million Americans, 9.4% of the popula- necessary to prevent glucose lability and end-organ dam-
tion. Diabetes remains the seventh leading cause of death in age.1 Invariably, patients are plagued by the development
the US in 2015, with 79,535 death certificates listing it as and progression of secondary complications of nephropa-
the underlying cause of death and a total of 252,806 death thy, neuropathy, and retinopathy, as well as macrovascular
certificates listing diabetes as an underlying or contributing and microvascular complications, which can appear years
cause of death.1 Despite advances in genetically modified to decades after disease onset. Evidence from the Diabetes
608
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 609

Fig. 36.1 A historical technique used for pancreatico-duodenal trans-


plantation, now seldom used. Exocrine drainage of grafts using a Fig. 36.2 Another historical technique employing enteric drainage via
cutaneous duodenostomy. (Reproduced from Lillehei, R.C., et al., Pancre- a Roux en Y limb of recipient bowel. Note the long donor duodenum
atico-duodenal allotransplantation: experimental and clinical experience. and end to end anastomosis to native jejunum (Reproduced from Lille-
Ann Surg 1970;172(3): p. 405–36.) hei, R.C., et al., Pancreatico-duodenal allotransplantation: experimental
and clinical experience. Ann Surg 1970;172(3): p. 405–36.)

Control and Complication Trial (DCCT) demonstrating that


intensive insulin therapy can mitigate secondary complica- enthusiasm and several centers embarked on their own
tions of diabetes gave added impetus to apply and hone pan- cases in the 1970s and 1980s. As experience expanded,
creas transplantation for patients with insulin-dependent surgical complications began to accumulate, resulting in
diabetes mellitus (IDDM).2 The majority of pancreas trans- graft and patient survivals ranging from a few months to
plants are performed simultaneously in conjunction with a 1 year.
kidney transplant for patients with IDDM, primarily type 1 In the mid 80s, Sollinger, Corry, and Ngheim devised
diabetes (T1D) and DM nephropathy. Sequential pancreas the bladder drainage technique, initially with a duodenal
after kidney transplants also achieve the goals of correcting button (Fig. 36.3) and then modified to a duodenal seg-
diabetes and eliminating the need for dialysis. ment.18 For the next decade, the bladder drainage tech-
nique with duodenocystostomy was the predominant
technique for pancreas transplants. The bladder drainage
History of Pancreas technique was associated with a low acute complication
Transplantation rate and was useful for measuring urinary amylase level,
which helped in monitoring for rejection. At around the
Oskar Minkowski and Joseph von Mering in 1889 demon- same time, use of University of Wisconsin (UW) organ
strated that pancreatic extirpation in dogs produced severe preservation solution and Cyclosporin A (CsA)-based
diabetes, partially reversed by implanting a small portion immunosuppression became routine and undoubtedly
of pancreas subcutaneously.3 Dr. P. Watson at the Bristol had a beneficial effect on improving technical success
Royal Infirmary, England, implanted three extracts from rates and graft survival. However, chronic urologic and
a freshly slaughtered sheep into a 15-year old boy with metabolic complications frequently plagued patients lead-
diabetes; unfortunately, due to severe diabetic ketoacido- ing to posttransplant morbidity, and gradually, in the mid
sis the patient expired 3 days later.4 The identification and 1990s, centers shifted back to enteric drainage, using a
isolation of the glycemia-lowering substance contained in graft duodenojejunostomy with or without a Roux-en-Y
pancreatic extracts by Banting, Best, Collip and Macleod,5,6 anastomosis. Today, a duodenojejunostomy without a
and experimental pancreas allotransplantation in dogs,7– Roux-en-Y anastomosis continues to be the most com-
14 prepared initial attempts to transplant the pancreas in monly used technique for managing exocrine secretions.
humans. Drs. William D. Kelly and Richard C. Lillihei per- Portal venous drainage was introduced by Sir Roy Calne
formed the first successful human pancreas allograft on in 1984 for segmental pancreas grafts as a potentially
December 16th, 1966, at the University of Minnesota.15 more physiologic technique.19
The graft was a pancreatic duct-ligated segmental graft With the introduction of CsA, tacrolimus, and myco-
transplanted simultaneously with a kidney in a uremic phenolate mofetil (MMF), rates of acute rejection dropped
patient resulting in immediate insulin independence. The dramatically. A number of other strategies such as T
patient expired 2 months later due to sepsis with a func- cell depleting antibodies,20 single antigen bead Luminex
tioning graft. After this initial transient success, additional human leukocyte antigens (HLA) alloantibody determi-
cases were reported, primarily using intestine-based exo- nation and virtual crossmatching,21 pancreatic allograft
crine drainage techniques (Figs. 36.1 and 36.2).16 Of the biopsies,22 and adoption of a standardized rejection grad-
initial series performed at the University of Minnesota, 11 ing schema23 have contributed to better outcomes in
were performed in uremic patients and 3 in nonuremic the modern era. Additional history has been detailed
patients.17 Some initial technical successes bolstered elsewhere.4,24
610 Kidney Transplantation: Principles and Practice

PTA

1500
SPK
PAK

500 1000
Transplants
All

0
2004 2006 2008 2010 2012 2014 2016
Year
Fig. 36.4 Annual number of US transplants. PAK, pancreas after kidney;
PTA, pancreas transplant alone; SPK, simultaneous pancreas and kid-
ney transplant. (Reproduced from Kandaswamy R, et al. OPTN/SRTR 2015
Annual Data Report: Pancreas. Am J Transplant 2017;17(Suppl 1):117–73.)

new patients on the waiting list dropped from a high of


2067 in 2004 to 1476 in 2015, the lowest level in the
past decade. As a result, the majority (∼70%) of pancreas
transplant centers in the US perform 10 or fewer pan-
creas transplants annually. Data from the Global Obser-
vatory on Donation and Transplantation show that 780
pancreas transplants were carried out in 2016 across the
28 countries of the European Union (population 506 mil-
lion). National transplant rates varied from less than one
procedure per million population per year to more than
three transplants per million population, as in Norway,
the UK, and the Czech Republic.27 A larger proportion of
European transplants are carried out as SPK transplanta-
tion than is the case in the US, where the PAK procedure
is more common. Patients with life-threatening hypogly-
caemia unawareness may be offered either solid-organ
PTA or islet transplantation. Most recent data from the
Fig. 36.3 A historical technique using duodenal bulb bladder drain- UK (2016–2017) show that 76% of all pancreas trans-
age technique. (Reproduced from Sollinger, H.W., et al., One thousand plantation (including islets) was in the category of SPK,
simultaneous pancreas-kidney transplants at a single center with 22-year 8% as PAK and PTA combined, and 16% as islet trans-
follow-up. Ann Surg 2009;250(4): p. 618–30.)
plantation. As a proportion of solid-organ pancreas
transplants, 91% were SPK.28
In the US, the vast majority of pancreas transplants
Recent Trends in Pancreas (∼90%) are performed in patients with insulin-dependent
Transplantation T1D as the underlying disease. However, increasingly
insulin-dependent type 2 diabetes (T2D) patients with
Approximately 900 to 1000 pancreas transplants are residual endogenous insulin production (positive fasting
performed annually in the US, approximately 90% serum C-peptide) are receiving pancreas (SPK and PAK)
involving a simultaneous pancreas and kidney (SPK) and kidney transplants, as T2D becomes the predominant
transplant for patients with IDDM and renal failure. cause of kidney disease in the US. T2D recipients now
The other 10% of pancreas transplants are equally split account for >10% of SPK transplants, and >6% of PAK
between sequential pancreas after kidney transplants and PTA transplants each. Waitlisted T2D candidates
(PAK) and pancreas transplants alone (PTA) in patients increased from a low of 7.3% in 2007 to over 10% cur-
with preserved renal function and difficult to control rently. Almost all transplants are from deceased donors,
diabetes associated with frequent severe hypoglycemia. with only five living donor cases performed over the past
Analysis of the annual data reports from the Organ 10 years in the US, and these were performed at only two
Procurement and Transplantation Network/Scientific centers. Living donor pancreas transplantation is more
Registry of Transplant Recipients (OPTN/SRTR),25 and common in Asia.
International Pancreas Transplant Registry (IPTR) com- The reasons for the decline in pancreas transplant vol-
paring the periods 2005 to 2009 with 2010 to 201426 umes in the US are not fully understood, especially in the
have demonstrated the number of US pancreas trans- context of steadily improving patient and graft survival.
plants declined by over 20%, whereas the overall num- Potential reasons include improvements in insulin delivery
ber of pancreas transplants performed outside the US has technologies, resulting in delayed progression to advanced
increased (Fig. 36.4). The greatest decline is observed diabetic nephropathy, and shift toward more obesity affect-
in the PAK population, followed by the SPK popula- ing patients with T1D.29 A recent decrease in donor organ
tion. The PTA population has remained steady at about quality is also likely a contributing factor. The US donor pop-
200 to 300 transplants per year. The number of active ulation is becoming increasingly aged, obese, and diabetic,
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 611

all factors that adversely affect pancreatic graft functional the same donor because the immunosuppressive medica-
outcomes. Despite these adverse trends, overall short-term tions needed are similar to those for a kidney transplant
technical success rates of pancreas transplantation have alone. As long as the cardiac and surgical risks of the dual
improved. The trend in pancreas transplantation follows operation are considered acceptable, the benefits of adding
the rule of 90s: 90% are SPKs, 90% are from conventional a pancreas transplant to ameliorate diabetes can help pro-
donors (body mass index [BMI] <30), 90% enteric drainage, long the survival of the transplanted kidney by preventing
90% systemic venous drainage, 90% receive cell-depleting recurrent diabetic nephropathy31 and is associated with
induction agent, 90% receive tacrolimus and MMF, 90% prolonged patient survival compared with a deceased or liv-
have T1D, 90% have a PRA <20%, 90% of recipients have ing donor kidney (LDK) alone in the long term.31 Moreover,
a BMI less than 30, and the 1-year and 5-year graft survival one operation achieves both elimination of dialysis and
is above 90%.34 And by the rule of 30s: 30% are at least restoration of normoglycemia. The pancreas donors also
50 yrs old, 30% experience acute rejection, 30% undergo generally represent the best 15% to 25% of kidney donors,
relaparotomy for postoperative complications, 30% of dual- which implies a high quality cadaver kidney is available for
organ biopsies are discordant, 30% develop donor-specific these patients. Depending on the allocation schema, the
antibodies, and 30% remain steroid-free long term.30 waiting time for an SPK transplant is usually less than that
The allocation policies for solid-organ pancreata to for a cadaver kidney.
patients with diabetes differ across the world and are con- Conventionally, pancreas transplantation was consid-
tinually undergoing refinement. In the US, recent changes ered suitable only for insulin-dependent T1D, lean, ketosis-
in the United Network for Organ Sharing (UNOS) Pancreas prone patients, with absent insulin secretion and C-peptide
Allocation System have led to an increase in pancreas levels that were extremely low or undetectable. In contrast,
transplant volumes (especially SPK transplants) and give patients with T2D are classically obese, insulin resistant,
priority to utilization of the pancreas as a solid organ over with inappropriately “normal” to elevated C-peptide levels.
that of islets. The UK pancreas allocation policy is based on Data from multiple centers support the notion that pan-
four objectives: (1) equality of access irrespective of geo- creas transplantation in insulin-dependent patients with
graphic location or the transplant center at which patients T2D with detectable C peptide, few comorbidities, and rea-
are registered; (2) maximum benefit with respect to utiliza- sonable insulin requirements is successful and comparable
tion of the organ and outcome of the transplant; (3) priority to T1D recipients of SPK.32–35 As the number of uremic
for sensitized and hard-to-match patients; and (4) alloca- T2D patients is increasing, UNOS has issued regulations
tion of suitable donor organs in appropriate numbers to islet that define eligibility criteria for patients with measurable
transplantation. For every donor organ, all patients on the C-peptide as a surrogate for T2D. These regulations impose
waiting list are awarded points, based on seven criteria: (1) limits by restricting eligibility in those patients having a
HLA mismatch; (2) waiting time; (3) degree of sensitization; C-peptide of greater than 2 ng/mL by imposing a maximum
(4) geography (travel time); (5) donor BMI; (6) dialysis sta- allowable BMI (initially 28 kg/m2). UNOS policy mandates
tus; and (7) donor to recipient age matching. The algorithm a 6-month review process with allowance for reduction (or
by which points are allocated (including the weighting of increase) in the BMI eligibility threshold by plus or minus
each criterion) is based on a series of simulations, designed 2 kg/m2 depending on the makeup and/or size of the wait-
to achieve the optimum outcome, balancing fairness with list, to a maximum of 30 kg/m2. The majority of T2D are
utility. The reduction in pancreas transplant activity that SPK and PAK recipients, but because T2D patients less
has occurred in the US in recent years has not been experi- commonly experience the problematic hypoglycemia and
enced in Europe, possibly because transplant rates had not related glycemic lability more often seen in long-standing
previously achieved the same levels. However, pancreas T1D, non-T1D account for very few PTA transplants per-
transplant rates in the larger European countries (e.g., the formed in the US.
UK) are relatively static. The evidence would suggest that
future developments will come as a result of improving PANCREAS AFTER KIDNEY TRANSPLANTATION
outcomes rather than donor organ availability. Early mor-
bidity, mostly associated with reperfusion pancreatitis and The second category for pancreas transplantation is patients
later graft failure, probably related to the problems of graft with IDDM who have received a previous kidney trans-
monitoring and undiagnosed rejection, are now the major plant from either a living or deceased donor. This group,
limiting factors. along with PTA, accounts for approximately 10% to 15%
of patients receiving pancreas transplants. Given the high
mortality of waitlisted patients for SPK (46% at 4 years) and
Indications and Patient Selection the proven benefit of kidney transplant for patients with
T1D,36 pursuing a kidney transplant before pancreas trans-
SIMULTANEOUS KIDNEY AND PANCREAS plant is a viable option for patients in regions with long
wait times for SPK or those who have a well-matched living
TRANSPLANTATION
donor kidney. Because PAK patients are already on calci-
Juvenile onset, autoimmune T1D is the principal indication neurin inhibitors (CNIs), the threshold for satisfactory kid-
for pancreas transplantation. Patients that develop chronic ney function is much lower than that for PTA patients and
kidney disease (CKD) stages 3 to 5 are potential candidates many patients can be successfully transplanted with a PAK
for SPK transplantation. According to current UNOS crite- with an eGFR of approximately 45 to 50 mL/min/1.73 m2
ria, candidates with an estimated glomerular filtration rate with an expectation of little change in eGFR after pancreas
(eGFR) <20 mL/min/1.73 m2 are eligible for listing. Such transplantation. Current literature supports equivalent
persons are excellent candidates for an SPK transplant from patient survival and improved GFR in patients who receive
612 Kidney Transplantation: Principles and Practice

a pancreas after a living donor renal transplant (LDRT) neoplasms), trauma, and various pediatric genetic or con-
versus patients who receive a LDRT alone up to 10 years genital abnormalities, and separately, in patients with
posttransplant.37 Factors that may lead to increased odds cystic fibrosis who develop both pancreatic exocrine and
of kidney loss include pre-PAK GFR <45 mL/min/1.73 m2, endocrine insufficiency. In these postpancreatectomy and
pre-PAK acute kidney rejection episodes, pre-PAK protein- cystic fibrosis patients, there is the added benefit of restoring
uria, pancreas transplant interval of >1 year after kidney exocrine function with an enteric-drained pancreas graft.44
transplant, and post-PAK pancreas allograft failure.38 In Pancreas transplantation is not indicated for the major-
other words, the ideal patient likely to benefit from PAK is ity of patients with IDDM, such as those who have normal
one deemed fit for surgery with minimal immunologic risk, renal function and do not exhibit a labile course, hypogly-
GFR of ≥50 mL/min/1.73 m2, without proteinuria or pre- cemic unawareness, or any evidence of nephropathy. For
ceding kidney rejection and a recent kidney transplant. patients that do have an indication for pancreas transplan-
tation, it is important to rule out significant medical contra-
PANCREAS TRANSPLANT ALONE AND indications in a similar manner as applies to other areas of
transplantation. These general contraindications include:
KIDNEY FUNCTION EVALUATION
recent malignancy, active or chronic untreated infection,
The third category for pancreas transplantation is in non- advanced forms of major extrarenal complications (i.e.,
uremic, nonkidney transplant patient with insulin-depen- uncorrected coronary artery disease [CAD] or severe aor-
dent diabetes who typically has hypoglycemia unawareness toiliac disease), life expectancy of less than 3 years, morbid
and/or severe metabolic instability and/or who has failed obesity with numerous metabolic complications, poor com-
attempts to manage their diabetes with insulin delivery and pliance, active substance abuse, and uncontrolled psychi-
glucose-sensing technology. These patients with longstand- atric disorder.
ing diabetes have extremely labile disease such that there is
difficulty with day-to-day living, associated with frequent RECIPIENT CONSIDERATIONS FOR SUCCESS AND
emergency room and inpatient hospitalizations for hypo- CARDIOVASCULAR ASSESSMENT
glycemia or diabetic ketoacidosis.39–41 Other patients may
have significant difficulty with hypoglycemic unawareness Advanced CAD is the most important comorbidity to con-
that results in unconsciousness without warning. This can sider in patients with IDDM and DM nephropathy. It has
be a devastating condition for these patients that ultimately been estimated that uremic, diabetic patients carry a near
affects their employment, their ability to keep a license to 50-fold greater risk of cardiovascular events then the gen-
drive, and concern about suffering lethal hypoglycemia eral population. Because of the neuropathy associated with
while asleep.39–41 The indications for a PTA are currently diabetes, patients are often asymptomatic because isch-
identical to patients being considered for an islet transplant: emia-induced angina is not perceived. The prevalence of
recurrent hypoglycemic/hyperglycemic attacks, ketoacido- significant (>50% stenosis) CAD in patients with diabetes
sis, inability to achieve adequate control with exogenous starting treatment for end-stage renal disease is estimated to
insulin therapy and newer automated technologies, or its be 40% to 60%. As such, screening studies to detect signifi-
failure to mitigate complications. cant and treatable CAD are important. Risk factors such as
The perfect candidate for a PTA includes patients with age, duration of dialysis, duration of diabetes, and smoking
preserved renal function; GFR >80 mL/min/1.73 m2 and/ and family history should be assessed but are not sufficiently
or serum creatinine <1.5 mg/dL and no proteinuria. In predictive, and standard cardiac risk assessment tools have
circumstances where a PTA candidate has marginal renal not been validated widely in this population. Hence, we
function, a trial of tacrolimus therapy can be used to pre- have adopted an aggressive policy of coronary angiography
dict the effect of CNI therapy on postoperative native kid- in nearly all dialysis patients and the majority of nonuremic
ney function. If native kidney functional reserve is deemed patients. Techniques for coronary angiography have
insufficient, then the patient is best served by waiting for improved significantly and by avoiding ventriculograms
kidney function to further deteriorate to an eGFR of 20 mL/ the contrast dye load and consequently the nephrotoxic risk
min/1.73 m2 or less. The risk of CKD is estimated to be 10% can be reduced considerably. Patients with coronary lesions
to 15% at 5 years post PTA,42,43 with a mean GFR decrease amenable to angioplasty and/or stenting or bypass grafting
of 33 mL/min/1.73 m2. Risk factors associated with renal should be treated, reevaluated, and then reconsidered for
failure include BMI >30 and age younger than 30 (attrib- transplantation. The goal of revascularization is to dimin-
uted to enhanced recipient immune status and more fre- ish the perioperative risk of significant myocardial ischemia
quent rejection episodes). and cardiac events, and to prolong the duration of life after
transplantation. Patients should have their cardiac status
EXTENDED INDICATIONS FOR PANCREAS reassessed at regular intervals (every 1 to 2 years).
Diabetic retinopathy is a nearly ubiquitous finding in
TRANSPLANTATION
patients with diabetes and end-stage renal disease. Signifi-
Indications for pancreas transplantation extend beyond cant vision loss may have occurred including blindness.
classic insulin-dependent T1D and T2D patients. Pan- Blindness is not an absolute contraindication to transplan-
creas transplantation is sometimes recommended for some tation because many blind patients lead very independent
patients with IDDM due to pancreatectomy or chronic pan- lives. Although rarely a problem, it should be confirmed that
creatitis, including failed total pancreatectomy and islet a patient with significant vision loss has an adequate support
autotransplantation.45,46 This cohort includes diabetes system to help with travel and medication administration if
seen after pancreatectomy either for chronic pancreatitis, needed. Acute normalization of glycemia has been associ-
pancreatic neoplasms (i.e., intraductal papillary mucinous ated with transient worsening of retinopathy, and stability
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 613

of retinopathy and its treatment should be ensured before pancreas allocation system currently dictates that T2D SPK
pancreas transplantation; annual ophthalmologic exami- candidates have to meet the following criteria for accrual
nations are recommended before and after transplantation. of waitlist time for pancreas transplantation: insulin
Autonomic neuropathy is prevalent and may manifest dependence, C-peptide ≤2 ng/mL with no BMI restriction
as gastropathy, cystopathy, and orthostatic hypotension. (assumed to be T1D) or on insulin and C-peptide >2 ng/mL
Impaired gastric emptying (gastroparesis) is an important with a BMI ≤30 kg/m2 (assumed to be T2D). However, the
consideration with significant implications in the post- BMI qualifier may be eliminated in the future in the US. In
transplant period. Patients with severe gastroparesis may the UK, BMI is factored into the allocation algorithm.
have difficulty tolerating the oral immunosuppressive med- Many centers are hesitant to perform a pancreas trans-
ications, predisposing to subtherapeutic levels and rejection plant in patients older than age 50 years. The latest
episodes. Patients typically require motility agents such as SRTR report25 shows that pancreas transplants declined
metoclopramide or erythromycin and, as such, gastropare- sharply in recipients aged older than 50 years, from 244
sis is not considered a contraindication to pancreas trans- in 2014 to 185 in 2015. Transplants in younger recipi-
plantation at many centers. Neurogenic bladder, manifested ents (age <35 years) increased from 252 in 2014 to 279
by inability to sense bladder fullness and empty the bladder, in 2015. In the context of improved outcomes and the
predisposes to urine reflux and high post-void residuals. This DM population now living longer, older patients should
may adversely affect renal allograft function, increase the be considered for pancreas transplantation. Many larger
incidence of bladder infections and pyelonephritis, and pre- centers in the US and UK are now considering selected
dispose to graft pancreatitis. The combination of orthostatic older patients to be eligible as long as the patient has a
hypotension and recumbent hypertension results from dys- favorable cardiovascular risk profile and does not have
regulation of vascular tone. This has implications for blood additional comorbidities.
pressure control posttransplant, especially in patients with From historical retrospective single-center data, obesity
bladder-drained pancreas transplants that are predisposed has been considered a risk factor for reduced kidney and
to volume depletion. Therefore posttransplant antihyper- pancreas graft survival in SPK transplantation for many
tensive medication must be reassessed. Sensory and motor years.29 In a recent analysis of the UNOS/SRTR data,47 the
neuropathies are common in patients with longstanding effect of recipient BMI on pancreas transplant outcomes
diabetes. This may have implications for the rehabilitation in all three pancreas transplant categories was examined.
posttransplant. It also reflects potential risk for injury to the The authors demonstrated that: (1) overweight and obesity
feet and subsequent diabetic foot ulcers. are associated with a moderate increase in early mortal-
Uremic, diabetic patients experience an increased rate ity, (2) overweight and obesity are associated with a mod-
of vascular complications, including cerebral vascular erate increase in early pancreas graft loss, (3) obesity, but
accidents, transient ischemic attacks, and peripheral vas- not overweight, is associated with poorer long-term graft
cular disease. Deaths related to cerebral vascular disease survival, and (4) underweight is associated with poorer
are approximately twice as common in patients with dia- long-term patient survival. Although the mechanisms
betes versus no diabetes once end-stage renal disease has underlying these findings are not understood clearly at
occurred. Patients with diabetes suffer strokes more fre- this time, multiple single-center studies have demonstrated
quently and at a younger age than do age and gender match noninferior patient and graft survival outcomes in obese
nondiabetic stroke patients. Hypertension is the major risk compared with nonobese patients in both DM catego-
factor for stroke followed by diabetes, heart disease, and ries, but at the expense of a higher surgical complication
smoking. Given the high rate of peripheral vascular dis- rate.48–51 Based on these data, our recommendation is that
ease present in the pancreas transplant population, assess- each center should determine what recipient BMI they are
ing the adequacy of the iliac vessels before transplantation comfortable with transplanting, it should be individualized
is paramount. A plain computed tomography (CT) scan of to the recipient, and that preoperative weight loss should
the abdomen and pelvis is the best option for assessing tar- be strongly encouraged. Given the frequent comorbidities
get vessels, despite examination findings of palpable femo- of morbid obesity, few centers will tackle patients with BMI
ral pulses. Iliac artery calcifications can be easily detected, >35. Morbidly obese (World Health Organization [WHO]
as well as aid in operative planning. Additionally, diabetic Class 2) patients could be considered for possible bariatric
patients are at risk for amputation of the lower extremity. surgery before transplantation.48,52
These problems typically begin with a foot ulcer associated Pretransplant assessment of autoimmune markers (e.g.,
with advanced neuropathy, Charcot foot fractures and antibodies against glutamic acid decarboxylase, insulin-
pressure ulcers and/or tibioperoneal vascular disease. Sig- oma-associated antigen-2, zinc transporter-8, and insulin)
nificant distal vascular disease or amputation of the lower should be considered to establish a baseline before possible
extremities is not, however, an absolute contraindication to pancreas transplantation. After transplantation, a new or
transplantation. increasing titer of a T1D-specific autoantibody may indicate
Mental or emotional illnesses including neuroses and recurrent autoimmunity as a cause for pancreas graft dys-
depression are quite common. These illnesses may influence function and aid in the evaluation of any new-onset hyper-
the decision of steroid-free/sparing regimens to avoid the glycemia posttransplant.53
deleterious effects of high-dose steroids on mood disorders. Assessment of the degree of hypoglycemic unawareness
High BMI T1D and T2D patients are often not offered pan- is crucial. The most common tool used to assess hypoglyce-
creas transplantation. Currently in the US there is a restric- mia awareness is the Clarke Hypoglycemia Symptom Ques-
tion regarding which patients with T2D can be offered SPK tionnaire. The Clarke method54 comprises eight questions
transplantation, whereas there is no similar restriction characterizing the patient’s exposure to episodes of moder-
for PAK or PTA transplantation. Nonetheless, the UNOS ate and severe hypoglycemia. It also examines the glycemic
614 Kidney Transplantation: Principles and Practice

threshold for, and symptomatic responses to, hypoglyce- TABLE 36.1 Contraindications to Deceased Pancreas
mia. A score of 4 or greater implies impaired awareness of Procurement for Transplantation
hypoglycemia. Assessment of the frequency and severity of
severe hypoglycemia should be part of the evaluation of all Contraindications
patients considering pancreas transplantation. History of DM (type 1 and 2)
BMI >35 kg/m2
Previous pancreatic surgery
Moderate/severe pancreatic trauma
Donor Selection and Management Acute or chronic pancreatitis
Intraabdominal sepsis
Major active infection
LOGISTIC AND TECHNICAL ASPECTS TO Chronic alcohol use
OPTIMIZE ORGAN VIABILITY Prolonged hypotension or hypoxemia with significant end-organ
damage (kidney, liver, etc.)
Determining donor HLA typing, viral serologies, and cross- Prolonged warm ischemia in DCD donor (>30 min)
match results with patients on the pancreas transplant Procurement-related injuries
waiting list will permit the ideal allocation of the cadaveric Severe fatty infiltration of pancreatic parenchyma
pancreas (plus kidney with SPK transplant) before procure- Severe pancreatic edema
Relative contraindication: presence of replaced right HA off superior
ment of the organs. This sequence of events has several mesenteric artery
advantages. It will allow the transplant center perform-
ing the pancreas transplant the choice to also procure the BMI, body mass index; DCD, donors after cardiac death; DM, diabetes mellitus;
pancreas, it will allow patients to be admitted to the hospi- HA, hepatic artery.
tal and the reevaluation process to begin simultaneously,
rather than sequentially, to the procurement of the organs, Hyperglycemia and hyperamylasemia are frequently
and it will minimize the cold ischemia time of the pancreas observed in cadaveric organ donors. Hyperglycemia is not
before implantation. It is ideal to revascularize the pancreas a contraindication to pancreas procurement for patients
within 16 hours from the time of cross clamping at procure- who are known not to have T1D or T2D. Hyperglycemia
ment. Finally, it will also allow identification of 0-antigen is generally benign and caused by a combination of factors
mismatched donor-recipient pairs to be identified before including administration of pharmacologic doses of steroids
procurement that will minimize cold ischemia time if the to reduce brain swelling, high rate infusions of glucose-con-
organs need to be transported across the country. taining solutions (especially in patients with diabetes insipi-
dus), and increased sympathetic activity associated with
brain injury. A recent study also showed minimal associa-
DONOR MANAGEMENT AND SELECTION
tion of donor HbA1c levels with pancreas graft outcomes.58
Identification of suitable deceased organ donors for pan- Hyperamylasemia may be concerning but reports have
creas transplantation is one of the most important deter- suggested that it has little effect on pancreas graft function
minants of outcome. The contraindications to use a posttransplant. It is likely that this finding was due to vari-
deceased donor pancreas for transplantation are outlined able laboratory ranges and/or other nonpancreatic causes
in Table 36.1. Optimal management of the donor avoids for elevated enzymes. If the cause of hyperamylasemia is
organ edema and pancreatitis. due to severe hemorrhagic pancreatitis or due to pancre-
The criteria that determine an appropriate donor for pan- atic injury in the case of a donor with trauma, organ recov-
creas transplantation are more stringent than for kidney or ery will be ruled out at the time of procurement. However,
liver donors. Deceased pancreas organ donors are typically isolated hyperamylasemia, without hyperlipasemia can
between the ages of 10 and 55 years. The lower age limit also occur in the setting of head trauma and salivary gland
typically reflects the anticipated small size of the splenic injury. The hemodynamic stability and need for inotropic
artery that may rarely preclude successful construction support is an important consideration, as hypotension can
of the arterial Y-graft needed for pancreas allograft revas- cause pancreatitis in addition to acute kidney injury and
cularization. We have performed many transplants from hepatocellular injury. Donor hypotension may have more
younger and lower weight donors (weight 15–25 kg) and influence on the anticipated function of the kidney allograft
have observed noninferior outcomes in graft and patient than it does on initial endocrine function of the pancreas
survival, with no significant difference in graft loss second- allograft.
ary to technical reasons.55 The use of older donors has been Perhaps the most important determinant of the suitabil-
associated with increased technical failure due to pancreas ity of the pancreas for transplantation is direct examination
graft thrombosis, a higher incidence of posttransplant pan- of the organ during and after the surgical procurement. The
creatitis, and decreased pancreas graft survival rates.56 This experience of the procurement team is important for cor-
may be consequent to reduced tolerability of cold ischemia rect assessment of the suitability of the pancreas graft for
time, but this has not been rigorously studied. The weight transplantation. It is during procurement that judgment
of the cadaveric organ donor is an important consideration. regarding the color, degree of fibrosis, adipose tissue around
Obese donors over 100 kg are often not suitable. Obese and in between the acinar lobules, firmness or nodularity
patients may have a history of T2D, or the pancreas may be of the graft, atrophy, hemorrhagic pancreatitis, saponifica-
unsuitable because of a high degree of adipose infiltration. tion, donor trauma, and specific vascular anomalies can be
Many transplant surgeons find the pancreas donor risk accurately assessed. Pancreata with heavy infiltration of
index (PDRI)57 a useful tool to gauge whether the donor adipose tissue are believed to be relatively intolerant of cold
will provide a suitable pancreas for transplantation. preservation, and carry the potential of saponification and
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 615

on data from SPK, PAK, and PTA transplants.57 The model


included 10 donor factors: donor sex, age, black race, Asian
race, BMI, cause of death, creatinine, height, and DCD sta-
tus. The transplant factors included pancreas preservation
time and an interaction between PAK and the donor cause
of death. The model was stratified by transplant type to allow
differing failure rates for each type of pancreas transplant.57
In summary, the PDRI can be a useful tool in decision mak-
ing at time of organ offer. In bridging the gap between organ
availability and utilization, cautious utilization of higher
PDRI donors could be shifted toward recipients with median
characteristics awaiting an SPK rather than solitary trans-
plants, whereas aggressive utilization of these high-PDRI
donor organs for all transplant categories should be practiced
in high-volume centers as their outcomes appear to be non-
inferior compared with the “perfect” donor. Interestingly,
the PDRI has been tested in a European (UK) population,64
Fig. 36.5 Example of a high quality pancreas at time of organ recovery, demonstrating that this algorithm is predictive for graft sur-
note the color and absence of fatty infiltration of the organ. vival of SPK recipients; the range of scores in the UK popula-
tion (1021 transplants) was greater than that reported in the
reperfusion pancreatitis after revascularization. Fig. 36.5 is US cohort, consistent with greater utilization of higher-risk
an example of a perfect organ in situ before cross clamp. pancreas grafts. Indeed, the UK donor cohort was older (34.9
The important vascular anomaly that must be evalu- vs. 26.3 years, P < 0.0001) and comprised a greater propor-
ated during procurement is the occurrence of a replaced tion of DCD transplants (10.8% vs. 1.4%, P < 0.0001).
or accessory right hepatic artery (HA) originating from the Another useful tool is the composite risk model devised
superior mesenteric artery (SMA). This should also be reas- by the group from the University of Minnesota to predict
sessed ex vivo. technical failure (TF).56 Of the factors analyzed in their mul-
The use of donors after cardiac death (DCD) for pancreas tivariable model, donor BMI >30, donor Cr >2.5, donor age
transplantation is well reported.59,60 There is a higher rate >50 yrs, and preservation time >20 hours had the stron-
of ruling out the pancreas for transplantation at the time of gest association with technical failure. Bladder drainage of
DCD procurement than in stable conventional donor after exocrine secretions was protective. In their composite risk
brain death (DBD) organ donors. If the pancreas is deemed model, the presence of one risk factor did not significantly
suitable, there is the added consideration of the effect of increase risk of TF (HR 1.35, P = 0.35). Two risk factors in
delayed kidney graft function in a uremic SPK candidate. combination, however, increased the risk of TF greater than
The use of living related and unrelated pancreas donors has threefold whereas three risk factors increased risk greater
also been described. A distal pancreatectomy is performed than sevenfold (HR 7.66, P ≤ 0.01) (Fig. 36.8).
for a segmental pancreas transplant. Anecdotal cases of
combined live donor partial pancreatectomy and nephrec- DONOR TRENDS AND PROCUREMENT-RELATED
tomy have also been reported. These procedures are not OUTCOMES
widely performed and are confined to one or two pancreas
transplant programs, primarily in Asia.26 Pancreas donor characteristics, as analyzed by the IPTR and
The organ preservation solution may affect pancreas graft SRTR, have changed over the past decade. Deceased pan-
survival. In an adjusted analysis of UNOS data comparing creas donor demographics show a steady increase in propor-
the UW solution versus histidine-tryptophan-ketoglutarate tions of younger donors (18–34 years), from 49% in 2004 to
(HTK), HTK preservation was independently associated with 64% in 2015. Similarly, the proportion of older donors (age
an increased risk of pancreas graft loss (hazard ratio [HR] >50 years) decreased from 6% in 2004 to 2% in 2015.25
1.30, P = 0.014), especially in pancreas allografts with cold Proportions of white donors decreased from 72% in 2004 to
ischemic time ≥12 h (HR 1.42, P = 0.017). This reduced sur- 61% in 2015, with a corresponding increase in proportions
vival was seen in both SPK and PTA transplants (Fig. 36.6). of black donors from 13% to 22%. Discard rates of pancreata
Furthermore, HTK preservation was also associated with recovered for transplant were highest for donors aged older
1.54-fold higher odds of early (<30 days) pancreas graft loss than 50 years, and for high-BMI donors (>30). The higher
compared with UW (odds ratio [OR] 1.54, P = 0.008).61 discard rates from older and higher-BMI donors reflect the
shared experience and published risks associated with these
donors. The ratio of male to female donors, and DCD to DBD
MODELS USED FOR DONOR SELECTION
donors has remained constant. The rate of trauma as cause
A donor risk index (DRI) derived retrospectively from reg- of donor death increased significantly over time in SPK
istry analyses that controls for donor, recipient, and trans- and PAK transplants, and remained stable in PTA. Propor-
plant-related factors, has been developed for kidney (KDRI) tions of anoxia as a cause of death increased from 10% in
and liver (LDRI) donors.62,63 These DRIs include factors that 2004 to 27% in 2015. Proportions of cerebrovascular acci-
can be identified at the time of organ allocation and can help dent decreased from 21% to 11% and of head trauma from
predict the risk of early graft failure at 1 year. Such a DRI was 66% to 61%. The preservation time decreased significantly
constructed for pancreas allografts (PDRI; Fig. 36.7) based in SPK and PAK. In 2010 to 2014, 62% of all transplants
616 Kidney Transplantation: Principles and Practice

Graft Survival, SPK Graft Survival, Pancreas Alone


100 100

90 90

Percent Survival
Percent Survival

80 80

70 70

60 60

UW UW
HTK HTK
50 50

0 6 12 18 24 30 36 0 6 12 18 24 30 36
Months Months
A B
Fig. 36.6 Survival curves for SPK and pancreas alone transplants based on preservative solution. SPK graft survival by HTK versus UW (A), Solitary pan-
creas graft survival by HTK versus UW (B). HTK, histidine-tryptophan-ketoglutarate; SPK, simultaneous pancreas and kidney UW, University of Wisconsin.
(Reproduced from Stewart ZA, et al. Histidine-tryptophan-ketoglutarate (HTK) is associated with reduced graft survival in pancreas transplantation. Am J
Transplant 2009;9(1):217–21.)

1-Year SPK Pancreas Graft Survival by 1-Year PAK Pancreas Graft Survival by 1-Year PTA Pancreas Graft Survival by
DRI Among Median Recipient DRI Among Median Recipient DRI Among Median Recipient

100% 100%
100%
95% 95%
% Pancreas graft survival
% Pancreas graft survival

% Pancreas graft survival


95%

90% 90% 90%

85% 85% 85%

80% 80% 80%


75% 75%
75%
70%
70%
70%
65%
65%
0 60 120 180 240 300 360 65%
0 60 120 180 240 300 360
Time from transplant (in days) 0 60 120 180 240 300 360
Time from transplant (in days) Time from transplant (in days)
DRI: 0.64-0.85 (N=1387) DRI: 0.86-1.15 (N=1906)

DRI: 1.16-1.56 (N=1135) DRI: 1.57-2.11 (N=625) DRI: 0.64-0.85 (N=519) DRI: 0.86-1.15 (N=764)
DRI: 0.64-0.85 (N=134) DRI: 0.86-1.15 (N=250)

A DRI: 2.12-2.86 (N=218) B DRI: 1.16-1.56 (N=394) DRI: 1.57-2.11 (N=167)


C DRI: 1.16-1.56 (N=128) DRI: 1.57-2.11 (N=79)

Fig. 36.7 (A) Adjusted 1-year graft survival after simultaneous kidney-pancreas (SPK) transplant as a function of the pancreas donor risk index (PDRI).
(B) Adjusted 1-year graft survival after pancreas after kidney (PAK) transplant as a function of the PDRI. (C) Adjusted 1-year graft survival after pancreas
transplant alone (PTA) as a function of the pancreas donor risk index (PDRI). Curves shown reflect expected survival for an average recipient (41 years
old, BMI = 25, male, white, albumin 3.7 g/dL, PRA = 0, private insurance, no prior PTx, and enteric-drained) transplanted with pancreata from each PDRI
strata. BMI, body mass index. (Reproduced from Axelrod DA, et al. Systematic evaluation of pancreas allograft quality, outcomes and geographic variation in
utilization. Am J Transplant 2010;10(4):837–45.)

had a preservation time of less than 12 hours, and only 1% program. Analysis of the UK experience showed that DCD
reported a preservation time of over 24 hours. A compari- donors (but not recipients) were selected to minimize other
son of preservation times in all three categories showed a risk factors (e.g., age) and that the graft and patient survival
slightly longer preservation time for PTA compared with data were similar to contemporary data from DBD donor
SPK because of the higher rate of organ imports.26 transplants.60,65 Similarly, the maximum acceptable age
Criteria for organ donation tend to be more liberal in for donation has increased in the UK, with 23% of trans-
European countries than the US, particularly in the UK. plants from donors older than 50 years. The complications
Of all deceased donors in the UK, 42% are DCD; pancreas of transplantation are believed to be greater in organs from
transplantation from this source was first carried out donors with higher BMI, with very few solid-organ trans-
in 2005 and currently represents 25% of the national plants carried out from donors with BMIs greater than 30
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 617

Number Number
Cox model predicted risk of technical failure 0.6 of Risk 100.0% of Risk
Factors Factors
0 0
0.5

TF-Free Pancreas Graft Survival


1 80.0% 1
2 2
3 3
0.4
60.0%
0.3

40.0%
0.2

20.0%
0.1

0.0 0.0%

0 30 60 90 0 30 60 90
Days posttransplant Days posttransplant
A B
Fig. 36.8 The University of Minnesota Composite Risk Model prediction of technical failure risk and observed pancreas graft survival. (A) The final Cox
analysis was used to generate predictive model of technical failure risk according to the number of risk factors present. The risk model is adjusted for
bladder/enteric drainage and history of pancreatitis in the donor. (B) The observed technical failure-free pancreas graft survival in this cohort is depicted
according to number of risk factors by univariable Kaplan–Meier survival. (Reproduced from Finger EB, et al. A composite risk model for predicting technical
failure in pancreas transplantation. Am J Transplant 2013;13(7):1840–9.)

kg/m2; the UK organ allocation scheme directs such organs cold-stored. The donor iliac vessels are obtained for revas-
preferentially to islet isolation. cularization of the arterial supply and sometimes the portal
Pancreatic trauma occurring at the time of organ pro- vein. There are two general methods of organ procurement.
curement contributes to the growing rate of pancreas dis- Many programs prefer to perform an en-bloc removal of the
card, with more than 20% of pancreata discarded after liver and pancreas together and separate the two organs
recovery in the US due to procurement trauma.25 The UK at the back table. Other programs prefer to perform a more
transplant registry was analyzed for the frequency of pan- deliberate dissection of the pancreas and liver by mobiliz-
creatic injuries, factors associated with damage, and the ing the relevant vasculature before in situ flush preserva-
effects of these injuries on graft survival. More than 50% tion, after which the liver and pancreas are separated in
of recovered pancreata had at least one injury, most com- situ, ensuring the organs are kept cold and there is rapid
monly a short portal vein (21.5%) and capsular damage extirpation.
(13.6%),66 and more than 50% of organs retrieved for The relevant components of the in situ procurement pro-
whole organ or islet transplant purposes were discarded. cess are briefly described as follows:
Liver donation, procurement team origin, HA arising from   

the SMA, and increasing donor BMI were associated with ◼ L ong midline incision (± cruciate incision);
increased rates of pancreas damage on univariate analyses; ◼ Mobilization of ascending colon, control of the infrarenal
on multivariate analysis only the presence of a HA from the aorta, and identification of superior mesenteric artery;
SMA remained significant.66 Overall, there was no differ- ◼ C ontrol of the supraceliac aorta;
ence in graft survival between damaged and undamaged ◼ Identification of HA (ligation of gastroduodenal artery),
organs; however, graft loss was significantly more frequent splenic artery, portal vein, and division of common bile
in pancreata with arterial damage (P = 0.04) and in those duct;
with parenchymal damage (Fig. 36.9). Damage to the pan- ◼ Identification of replaced and/or accessory left and right
creas during organ recovery is more common than liver hepatic arteries;
and kidney recoveries, and hence scrupulous procurement ◼ Exposure of the anterior aspect of the pancreas for visual
technique and awareness of anatomic variants are essen- and manual inspection;
tial to reduce rates of procurement-related injuries leading ◼ Mobilization of the spleen by division of short gastric ves-
to discard.66 sels and dissection of its ligamentous attachments;
◼ Mobilization of head, tail, and body of the pancreas with
minimal manipulation of the organ;
◼ Nasogastric (NG) tube positioning into the proximal duo-
Surgical Procedure denum and irrigation with antibiotic solution;
◼ Removal of NG tube and division of the proximal duode-
PANCREAS PROCUREMENT
num proximal to the pylorus (this can be further resected
The pancreas must be procured with an intact vascular sup- on the back table);
ply that does not compromise the vascularity of the liver. ◼ Heparinization of the donor and infusion of intraaortic (±
The pancreas is procured with the spleen and duodenum intraportal, avoid inferior mesenteric vein (IMV) flush to
intact. The organ is perfused with preservation solution and prevent edema of the pancreas) preservation solution;
618 Kidney Transplantation: Principles and Practice

1.0 1.0

0.8 0.8
Cumulative survival

Cumulative survival
0.6 0.6

0.4 0.4

0.2 0.2

0.0 0.0

0.0 10.0 20.0 30.0 40.0 50.0 60.0 0.0 10.0 20.0 30.0 40.0 50.0 60.0
A Months posttransplantation B Months posttransplantation

1.0

0.8
Cumulative survival

0.6

0.4

0.2

0.0

0.0 10.0 20.0 30.0 40.0 50.0 60.0


C Months posttransplantation
Fig. 36.9 Graphical demonstration of pancreas graft survival, stratified by presence and type of pancreas damage. (A) Pancreas graft survival by pres-
ence of any damage (damage – gray [n = 233], no damage – black [n = 309]; P = 0.28). (B) Pancreas graft survival by presence of arterial damage (arterial
damage – gray [n = 10], no arterial damage – black [n = 528]; P = 0.04). (C) Pancreas graft survival by presence of parenchymal damage (damage – gray
[n = 9], no damage – black [n = 529]; P = 0.05). (Reproduced from Ausania F, et al. A registry analysis of damage to the deceased donor pancreas during
procurement. Am J Transplant 2015;15(11):2955–62.)

◼ Division of proximal jejunum, middle colic vessels, supe- IMPORTANCE OF EX VIVO BACK TABLE
rior mesenteric vessels distal to the pancreatic uncinate
PREPARATION
process;
◼ Division of celiac, SMA, splenic arteries; and portal vein; The back table preparation of the pancreaticoduodenal
◼ Procurement of liver, pancreaticoduodenosplenic allograft, allograft for transplantation requires careful and meticu-
and kidneys, and reflushing with fresh UW solution ex vivo lous surgical technique to ensure a properly revascularized
before packaging. After in situ flushing and during extirpa- pancreas with adequate duodenum and minimal extrane-
tion the organs must be kept cold; ous fibrotic and adipose tissue (Fig. 36.10).67 The quality of
◼ Procurement of donor iliac vessels; the back table surgical procedure has the greatest effect on
◼ Closure of incision. the outcome of the transplant surgery.
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 619

A B
Fig. 36.10 Back table preparation of the pancreaticoduodenal allograft. Prepared graft with a duodenal segment (left). Y-graft for anastomosis to the
donor splenic artery and donor superior mesenteric artery (right). (Reproduced from White SA, Shaw JA, Sutherland DE. Pancreas transplantation. Lancet
2009;373(9677):1808–17.)

The pancreaticoduodenosplenic allograft has previously recipient is undergoing a retransplant. Rescue of a short (2
been procured from the deceased donor by the procuring mm) portal vein can be accomplished with a portal venous
surgeons and placed in preservation solution and packed extension graft of donor external iliac vein.
according to local guidelines in a cold storage container An important anomaly that must be skillfully managed
for transportation to the transplantation center. The during procurement and reevaluated on the back table is
container with the allograft is brought into the operat- the occurrence of a replaced or accessory right HA originat-
ing room and the identification number and donor blood ing from the SMA. The presence of a replaced right HA is
group cross-referenced with the pancreas transplant recip- no longer an absolute contraindication for the use of the
ient. A basin filled with ice is filled with fresh, cold preser- pancreas for transplantation, and in our experience, it is
vation solution in an effort to dilute out possible bacteria extremely rare to discard a pancreas for this reason. Experi-
present in the preservation solution in which the pancreas enced procurement teams will be able to successfully sepa-
is stored which has been shown to occur nearly 30% of rate the liver and the pancreas either in situ, or on the back
the time. The organ is then removed from the transport table, without sacrificing quality of either organ for trans-
container and placed in the chilled preservation solution. plantation, in the majority of cases. However, there are a
Two surgeons typically perform the back table operative few important caveats that determine whether this is pos-
preparation. sible. The replaced right HA needs to be dissected down to
Visual inspection of the allograft is critical before mak- the SMA. If the replaced right HA traverses deep into the
ing the final decision to proceed with transplantation. The parenchyma of the head of the pancreas requiring exten-
organ should be evaluated for the color, degree of fibrosis, sive dissection, this may preclude using the pancreas for
adipose tissue around and in between the acinar lobules, transplantation as there would be a high likelihood of pan-
thickness of the mesentery, firmness or nodularity of the creatic enzyme leak. The SMA is often divided distal to the
graft, and specific vascular anomalies/injuries. Pancreata take-off of the replaced right HA, preserving it intact on a
with heavy infiltration of adipose tissue are relatively intol- short length of SMA with a carrel patch for the liver graft.
erant of prolonged (>18 hours) cold preservation and the Occasionally, there is a large inferior pancreaticoduodenal
potential of a high degree of saponification due to reperfu- arterial branch vascularizing the head of the pancreas that
sion pancreatitis after revascularization. These organs may originates proximal to, or at the junction of, the take-off of
be more suitable for islet isolation. the replaced right HA. The inferior pancreaticoduodenal
Inspection for parenchymal, vascular, and duodenal vessels are critical to vascularization of the head of the pan-
injuries, as well as vascular anomalies should be conducted creas because the gastroduodenal artery is routinely ligated
in a disciplined fashion. The divided small bowel mesentery during the process of HA immobilization for the liver trans-
should be inspected to ensure that it has not been divided plant. Therefore in the case of a very proximal take-off of the
excessively short, encroaching on the pancreatic paren- inferior pancreaticoduodenal artery (IPDA), the replaced
chyma and compromising the intraparenchymal vascu- right HA would have to be transected distal to the IPDA and
lar supply. The pancreatic portal vein must be carefully reconstructed to the hepatic arterial tree; otherwise divid-
inspected to ensure that it has not been cut excessively ing the SMA at the appropriate location for proper liver
short, damaging the joining splenic vein; a distance greater procurement would significantly impair vascularization of
than 1.5 cm from the splenic vein inlet is usually adequate. the head of the pancreas and preclude its use for transplan-
This can occur when the portal vein is put on stretch dur- tation. Evaluation of the arterial vascularity of the pancre-
ing liver procurement and over aggressively divided to aticoduodenal allograft can be tested on the back table by
generate excessive length for the liver transplant team, typ- several methods: (1) injection of Renografin into the supe-
ically when the procuring surgeons are told that the liver rior mesenteric artery or Y-graft and obtaining an x-ray;
620 Kidney Transplantation: Principles and Practice

(2) intraarterial injection of fluorescein visualization with a with complete celiac and SMA and intact gastroduodenal
Wood’s lamp; (3) performing a methylene blue angiogram; artery. In these cases a Y-graft reconstruction is not nec-
and (4) flushing the SMA with preservation solution and essary so long as the celiac and SMA arterial inflow are
observing if it exits the splenic artery and vice versa. Any of preserved. Exceptional duodenal graft perfusion will be
the these methods would ensure suitable vascular perfusion observed after recipient graft arterial revascularization.
of the pancreatic head and duodenum. Some groups routinely modify the outflow portal venous
The duodenum should be carefully inspected to rule out a vascular system, as well, but in the majority of cases it is
serosal injury. Many groups will open the duodenum at the not necessary as the portal vein can be dissected back to
distal corner staple line, to irrigate and drain the contents the bifurcation of the SMV and splenic vein. The portal vein
into a separate container, then close the opening. Some pro- is carefully mobilized to allow for appropriate length and
grams routinely culture the fluid and a small piece of duo- determination whether a short portal venous extension
denal tissue. graft utilizing donor external iliac vein would be useful. A
The main principles in allograft preparation are as fol- portal vein extension is accomplished using donor external
lows. The spleen is removed from the pancreas tail with iliac vein, aligning in the direction of blood flow using run-
secure ligatures on the large splenic vessels. The use of the ning 5-0 or 6-0 monofilament suture.
vascular stapler is also acceptable and may be more efficient. After vascular reconstruction, some groups infuse the
In some cases it may be useful to cut the tie off the com- pancreas Y-graft with 100 to 200 mL of fresh, cold, preser-
mon bile duct (CBD) and place a cannula (5 French feeding vation solution to examine for parenchymal vascular leaks
tube) temporarily to identify the location of the ampulla by that can be repaired before recipient revascularization. It is
manual palpation. A marking pen can be used to designate important not to use the solution in the basin for flushing
its location to accurately ensure its center position as the because small particulate matter will be introduced into
proximal and distal duodenum are shortened to an appro- the organ and increase risk of vascular compromise and
priate length (the tube is removed and the CBD retied). The thrombosis. Finally, the preservation solution should be
first portion of the duodenum is dissected a couple of cen- exchanged for fresh solution and plenty of ice resupplied
timeters distal to the pylorus and divided proximal to the in the basin underneath it. The total operative time for the
ampulla with the stapler. The third portion of the duode- back table work can exceed 3 hours in a complicated case,
num is dissected off the mesentery until adjacent to the pan- but ideally should take 1.5 to 2 hours. Using a Ligasure
creas and stapled. Effort should be made to bevel the staple device or harmonic scalpel device instead of ties for small
line such that the antimesenteric side of the bowel is shorter vessels may speed up the back table work.
than the mesenteric side. This may maximize vascularity to
the entire bowel wall. Next the staple lines are inverted with TECHNIQUE FOR PANCREAS
3-0 nylon or silk suture as interrupted stitches. It is impor- TRANSPLANTATION
tant to carefully and liberally invert the corners of the duo-
denum to avoid protrusion and eventual breakdown and The surgical techniques for pancreas transplantation are
leakage. The mesenteric vessels protruding through the diverse (Figs. 36.11–36.13). The principles are consistent,
pancreas uncinate may have been stapled or individually however, and include providing adequate arterial blood
ligated by the procurement team. The staple line on the root flow to the pancreas and duodenal segment, adequate
of the mesentery is oversewn with a running 3-0 monofila- venous outflow from the pancreas, and secure management
ment suture for reinforcement. The middle colic vessels are of the pancreatic exocrine secretions. Adequate exposure
secured. is also necessary for intraabdominal implantation of the
Modifying the inflow arterial system is performed rou- deceased donor kidney when a SPK transplant procedure
tinely. The donor splenic artery and the SMA are identified is performed. Proper exposure is accomplished by a midline
and prepared for reconstruction by removing surrounding incision because both organs can be implanted through the
ganglionic tissue. The reconstruction requires use of the single incision. After the midline incision, a modified Cattell-
donor iliac arterial complex: common, internal, and exter- Braasch maneuver is often used to properly expose the recip-
nal arteries. The iliac arterial complex is a critical inclusion. ient vessels for the pancreas transplant. This will expose the
The arterial complex should be carefully inspected to deter- right iliac vessels, aorta, and inferior vena cava. The pan-
mine whether injury at the internal/external junction has creas is ideally placed on the right iliac vascular supply. To
occurred (usually by excessive retraction during procure- properly expose the iliac vessels for the intraabdominal kid-
ment). The Y-graft is modified by dividing the internal and ney transplant, the descending colon and sigmoid are mobi-
external iliac arteries to an appropriate length tailored for lized medially by dividing along white line of Toldt lateral to
the end-to-end anastomoses on the splenic and superior the descending and sigmoid colon. The surgical techniques
mesenteric arteries of the pancreas allograft, respectively. for pancreas transplantation are defined according to the
Typically 7-0 or 6-0 monofilament suture is used as a anatomy of venous insulin delivery, pancreatic exocrine
running stich, but interrupted stiches would be appropri- drainage technique, and whether the pancreas graft is ori-
ate, especially if a pediatric donor pancreas is being trans- ented cephalad or caudad. Systemic venous insulin delivery
planted. Conversely, if a sufficient length of splenic artery via the iliac vein combined with enteric exocrine drainage
can be mobilized, it is possible to perform a direct end-to-side is the most commonly applied method, usually with the
anastomosis to the superior mesenteric artery. Occasionally pancreatic head cephalad. Another option is portal venous
the pancreas allograft is obtained without a simultaneous insulin delivery combined with enteric exocrine drainage
liver procurement. In this circumstance the procurement (this accounts for approximately 10% of cases). The other
team will return the pancreas allograft on an aortic patch common method is systemic venous insulin delivery via the
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 621

A B
Fig. 36.11 SPK transplantation with systemic endocrine and bladder exocrine drainage. (A) Pancreas transplantation with enteric exocrine drainage (B). SPK,
simultaneous pancreas and kidney. (Reproduced from Sollinger HW, et al. One thousand simultaneous pancreas-kidney transplants at a single center with 22-year
follow-up. Ann Surg 2009;250(4): 618–30.)

Fig. 36.12 Enteric conversion of previously bladder-drained pancreas Fig. 36.13 SPK transplantation with portal venous drainage. The pan-
transplant. (Reproduced from Sollinger HW, et al. One thousand simulta- creas is typically placed in the mid abdomen with anastomosis of the
neous pancreas-kidney transplants at a single center with 22-year follow- donor portal vein to the major branch of the recipient superior mes-
up. Ann Surg 2009;250(4):618–30.) enteric vein for venous drainage. (Reproduced from Redfield RR, et al.
Pancreas transplantation in the modern era. Gastroenterol Clin North Am
2016;45(1):145–66.)
iliac vein combined with bladder exocrine drainage with
the pancreatic head caudad. Enteric drainage is performed
VENOUS AND ARTERIAL RECONSTRUCTION
almost uniformly in SPK transplant cases. Bladder drainage
is used occasionally both in the US and Europe with solitary There are two choices for venous revascularization: sys-
pancreas transplants because of the opportunity to utilize temic and portal. Systemic venous reconstruction is used
urinary amylase monitoring for rejection in this category of in over 80% of cases. Systemic venous revascularization
cases because of the relatively higher rates of immunologic typically involves the distal vena cava/right common iliac
graft losses, although there is little published evidence yet to vein, or right external iliac vein (although it can also be
suggest that this benefits outcome.68 done on the left side). If portal venous drainage is used, it is
622 Kidney Transplantation: Principles and Practice

necessary to dissect the superior mesenteric vein at the root USA DD Primary Pancreas Transplants 1/1/1984 – 12/31/2016
of the mesentery. The pancreas portal vein is anastomosed % ED drained
end-to-side to the superior mesenteric vein. This places the 100
pancreas allograft anterior to the small bowel mesentery PAK
and influences the methodology of arterial revasculariza- 80 PTA
tion requiring the use of a long Y-graft placed through a SPK
60
window in the mesentery to reach the right common iliac
artery. The choice of using systemic versus portal venous 40
drainage tends to be made at the pancreas transplant center
level rather than according to case-by-case considerations 20
within a pancreas transplant program. Portal venous
drainage of the pancreas is more physiologic with respect 0

to immediate delivery of insulin to the recipient liver. How-

19
19 /85
19 /87
19 /89

19 /91
19 /93
19 /95
19 /97
20 /99
20 /01
20 /03
20 /05
20 /07
20 /09
20 /11
20 2/1
20 4/1
84
86
88
90

92
94
96
98
00
02
04
06
08
10
1
1 3
16 5
ever, portal drainage results in diminished circulating

/
insulin levels relative to those in systemic venous-drained Transplant Year
pancreas grafts. The route of venous drainage has no docu- Fig. 36.14 Percentage of enteric-drained pancreatic grafts in the US
mented clinically relevant differences in glycemic control or according to transplant category over time. PAK, pancreas after kid-
ney; PTA, pancreas transplant alone; SPK, simultaneous pancreas and
outcomes. kidney. (Reproduced from Professor Angelika Gruessner with permission.)
The pancreas graft arterial revascularization involves the
recipient right common or external iliac artery, with the
arterial Y-graft anastomosed in an end-to-side fashion. Posi- for rejection and the duodenal segment or head of the pan-
tioning of the head of the pancreas graft cephalad or caudad creas is amenable to transcystoscopic biopsy. However, the
is not relevant with respect to successful arterial revascular- switch away from bladder drainage eliminated the plethora
ization. The arterial anastomosis is typically performed after of complications that plagued these patients for many years
the venous anastomosis, except when the SMV is used for posttransplant, including cystitis, urethritis, urethral stric-
a portal-drained pancreas. In that case, it may be useful to ture and disruption, balanitis, hematuria, metabolic acido-
implant the straight portion of the arterial Y-graft graft-first sis, and frequent urinary tract infections and bladder leaks.
into the recipient iliac artery, then perform the venous anas- These were so prevalent that at some centers, up to 40% of
tomosis followed by rejoining of the donor iliac Y-graft to the all bladder-drained pancreas transplants met the indications
previously implanted portion. A well-perfused pancreas will for enteric conversion.69 Nowadays, pancreatic exocrine
have uniform pink color, including the duodenum. The duo- drainage is most commonly accomplished via the duodenal
denum will begin filling with secretions and it may be bene- segment of the pancreas graft with anastomosis to a loop
ficial to not let the duodenum become too distended, risking of the small intestine (typically proximal to midjejunum).
pancreatitis. The pancreas is carefully positioned according Enteric drainage of the pancreas allograft is physiologic with
to the orientation of the pancreatic head. If it is in the cepha- respect to the delivery of pancreatic enzymes and bicarbon-
lad position, the tail is positioned inferiorly into the pouch of ate into the intestines for reabsorption. The enteric-drained
Douglas or lateral to the cecum in the right paracolic gut- pancreas can be constructed with or without a Roux-en-Y
ter, depending on how the intestines were previously mobi- intestinal limb. The enteric anastomosis can be made side-
lized. Palpation of the vessels will confirm a strong splenic to-side or end-to-side with the duodenal segment of the pan-
arterial pulse and soft portal vein. Adequate mobilization of creas. The anastomosis may be hand sewn or accomplished
the recipient bowels may be beneficial to avoid significant with a stapler. The former is more common. Currently, pan-
downward (caudad) pressure on the pancreas and tension creas transplantation with direct enteric exocrine drainage
on the portal vein. The intestines are repositioned around is performed in 90% of cases in the US with the Roux limb
the allograft with the head of the pancreas oriented cepha- diversion technique being performed in approximately 20%
lad and superior adjacent to the midjejunum. of cases. Data from the IPTR have not convincingly shown
a benefit of primary Roux-en-Y diversion. A frequently cited
PANCREATIC EXOCRINE SECRETION disadvantage of jejunal drainage is the lack of access for safe
MANAGEMENT graft biopsy (compared with bladder drainage). The modi-
fication of duodeno-duodenostomy was first described by
The exocrine secretions of the transplanted pancreas must be the group in Liege, Belgium70 and has been adopted by the
controlled so that spillage and leakage does not occur post- Oslo group71 and others. Although clearly providing access
transplant. The two methods include enteric and bladder for diagnostic purposes, it does not resolve the challenge of
drainage. Enteric drainage refers to small bowel diversion surveillance, and there is no published evidence of a graft
in the recipient of the pancreas exocrine secretions. This is survival advantage in patients treated in this way.
generally done with a side-to-side enteric anastomosis with
or without a Roux limb; whereas bladder drainage refers
SURGICAL VOLUME-RELATED OUTCOMES
to draining exocrine secretions directly into the recipient
bladder via a duodenovesical anastomosis. For the recipient In a Eurotransplant analysis, unadjusted 1-year patient
of an SPK transplant, enteric drainage is the technique of survival was associated with mean center volume for each
choice and has been the predominant method over the past group; patient survival was best in high-volume centers,
20 years as illustrated in Fig. 36.14. The advantage of blad- compared with medium and low volume: 96.5%, 94%, and
der drainage is that it provides means of urinary monitoring 92.3%, respectively (P = 0.017). PDRI was highest in high
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 623

1.0 1.0

Pancreas graft survival (death-censored)


0.9
Patient survival

0.9

0.8

0.8

0.7
0 1 2 3 0 1 2 3
Years since transplantation Years since transplantation
0 days 180 days 1 year 3 years 0 days 180 days 1 year 3 years
Low volume Low volume
N at risk 382 353 332 216 N at risk 382 300 280 170
Patient survival 100% 94.2% 92.3% 88.3% Graft survival 100% 83.3% 81.6% 75.2%
Medium volume Medium volume
N at risk 399 353 329 170 N at risk 399 310 284 139
Patient survival 100% 95.1% 94.0% 92.4% Graft survival 100% 84.3% 83.2% 78.2%
High volume High volume
N at risk 382 360 346 236 N at risk 382 324 307 203
Patient survival 100% 97.4% 96.5% 93.6% Graft survival 100% 88.2% 86.0% 82.1%
A B
Fig. 36.15 Effect of center volume on outcomes in Eurotransplant Region. (A) Patient survival estimates stratified by low (blue line), medium (green line),
and high (yellow line) volume centers (P = 0.017). (B) Pancreas graft survival estimates stratified by low, medium, and high volume centers (P = 0.114).
(Reproduced from Kopp W, et al. Center volume is associated with outcome after pancreas transplantation within the Eurotransplant Region. Transplantation
2017;101(6):1247–53.)

volume centers: 1.38 versus 1.21 in medium and 1.25 in risk index (Fig. 36.16). These recent data are convincing
low volume centers (P < 0.001). Although pancreas graft evidence that center volume has an effect on outcomes after
survival at 1 year did not differ significantly between vol- pancreas transplantation.
ume categories, high center volume was protective for graft
failure (HR, 0.70; P = 0.037) compared with low center
volume. The results favor the notion that patient and graft Postoperative Management of
survival after pancreas transplantation are superior in Common Complications
higher volume centers and that high-volume centers have
good results, even though they transplant organs with the IMMEDIATE POSTOPERATIVE CARE
highest PDRI (Fig. 36.15).72
In another analysis of OPTN data, the effect of transplant Intraoperatively, blood glucose is monitored every 30 min-
center volume on pancreas allograft survival in 11,568 utes and is controlled with an insulin drip until reperfusion
SPK and 4308 solitary pancreas (PTA/PAK) transplants of the pancreas or longer if necessary. At the time of organ
between 2000 and 2013 was examined.73 For SPK trans- reperfusion, adequate volume status and blood pressure
plantation, low (HR 1.55, 95% confidence interval [CI] are imperative to avoid graft hypoperfusion. We routinely
1.34–1.8) and medium (HR 1.24, 95% CI 1.07–1.44) discontinue the insulin drip immediately after organ reper-
center volumes were associated with a higher risk of early fusion, and after a short (1 hour) observation in the postop-
pancreas graft failure at 3 months. The increased risk erative care unit. The vast majority of patients should be off
associated with low center volume extended to 1, 5, and insulin by the time they leave the postoperative care unit.
10 years. For solitary pancreas transplants, low, but not Close monitoring of glucose (every 1 hour) is continued
medium, center volume was associated with a higher risk of for the first 24 hours, and persistent or acute elevation of
early pancreas graft failure at 3 months (HR 1.56, 95% CI serum glucose above 200 mg/dL warrants immediate eval-
1.23–1.97) and this risk persisted over 10 years. Patients uation with Doppler ultrasonography to assess graft perfu-
transplanted at high-volume centers had better pancreas sion and function. Similarly, serum amylase and lipase are
survival rates across all categories of the pancreas donor monitored daily to assess pancreas graft function.
624 Kidney Transplantation: Principles and Practice

metabolism of the immunosuppressant and resultant


1 year pancreas allograft survival in SPK for center volume higher systemic concentrations. Fungal infections result in
groups by PDRI
the highest rates of graft loss and patient mortality. Cyto-
megalovirus (CMV) prophylaxis is recommended for any
positive combination of a donor–recipient pair.79–82 Ganci-
PDRI <0.85
clovir and, more recently, valganciclovir are presently the
PDRI 0.86–
antiviral agents of choice in pancreas transplantation and
1.15 can be initiated intravenously or per oral in the immediate
postoperative period. Most centers begin trimethoprim/sul-
PDRI 1.16–
1.56
famethoxazole immediately postoperatively and continue
14–34 cases/yr long-term prophylaxis against Pneumocystis jirovecii and
PDRI 1.57– Nocardia infections.
2.11 1–6 cases/yr

PDRI >2.12 Surgical, Medical, and Urologic


65% 70% 75% 80% 85% 90% 95% Complications
Fig. 36.16 The 1-year pancreas allograft survival in low- and high-­ Pancreatic grafts are susceptible to a unique set of surgical
volume centers in the SPK setting in the US stratified according to PDRI complications related to the exocrine secretions and the low
group. PDRI, pancreas donor risk index; SPK, simultaneous pancreas and microcirculatory blood flow to the gland. Surgical compli-
kidney. (Reproduced from Alhamad T, et al. Transplant center volume and
the risk of pancreas allograft failure. Transplantation 2017;101(11):2757–64.) cations are more common after pancreas transplantation
compared with kidney transplantation. Nonimmunologic
complications of pancreas transplantation account for graft
ANTICOAGULATION
losses in 5% to 10% of cases. These occur commonly within
Some centers advocate low-dose intravenous or subcutane- 6 months of transplant and are as important an etiology
ous heparin owing to the risk of vascular thrombosis lead- of pancreas graft loss in SPK transplantation as is acute
ing to technical failure. With the exception of aspirin (Bayer rejection.
Corporation, Pittsburgh, PA), the University of Wisconsin
transplant program does not use anticoagulation intra- VASCULAR THROMBOSIS
operatively or postoperatively in uremic diabetic patients
undergoing SPK transplant. A small series of patients in According to the 2015 SRTR report, graft thrombosis
the past received Plavix (clopidogrel bisulfate, Bristol-Myers remains the most common cause of early graft technical
Squibb/Sanofi Pharmaceuticals Partnership, Bridgewater, failure at a rate of 4.1% and 6.4% in SPK and PAK recipi-
NJ); however we routinely avoid use of systemic anticoagu- ents.26 It typically occurs within 48 hours to a few days of
lation with heparin or dextran, which has resulted in a low the transplant. This is generally due to venous thrombo-
rate of bleeding requiring reexploration while maintaining sis of the pancreas portal vein. The etiology is not entirely
a very low rate of pancreatic graft thrombosis (<4%).31,74 defined but is believed to be associated with reperfusion
Solitary pancreas transplants selectively receive a single pancreatitis and the relatively low-flow state of the pan-
intraoperative dose of 2000 U intravenous (IV) heparin creas graft. Obviously, technical misadventure is also possi-
without any postoperative continuation. Most patients ble but, in our experience, it is almost entirely due to severe
receive deep vein thrombosis prophylaxis with sequential ischemia reperfusion injury. To minimize graft thrombosis,
compression devices and selectively with 5000 U subcuta- prudent selection of donor pancreas grafts, short cold and
neous heparin at the time of induction and reinitiated post- warm ischemia times, and meticulous surgical technique to
operatively depending on clinical indication. minimize tension on the PV are necessary. The quality of
the pancreas graft, the age of the donor, and the cold isch-
emia time also influence graft thrombosis rates.56 Arterial
ANTIMICROBIAL PROPHYLAXIS
thrombosis is less common and is usually associated with
Early infection results in a high incidence of graft loss and in anastomosis to atherosclerotic vessels. Because of this,
serious patient morbidity and mortality.75–78 Various single many centers exclude patients with severe aortoiliac dis-
agents or combinations are available and should be given ease. However, mild to moderate disease is not necessarily
over the first 24 to 48 hours after transplantation. Recipi- a contraindication as long as two safe arterial implantation
ents with positive urine cultures (from preoperative speci- sites can be approached.
mens) or positive intraoperative duodenal stump cultures, if
cultured, should have antibiotic coverage for 3 to 7 days. A TRANSPLANT PANCREATITIS
detailed microbial history of an individual transplant can-
didate is imperative so that appropriate antibiotic coverage Pancreatitis of the allograft occurs to some degree in all
can be initiated intraoperatively. Because of the duodenal patients postoperatively. It is common to see a temporary
anastomosis in pancreas transplantation and the potential elevation in serum amylase and lipase levels for 48 to 96
contamination of the operative field with small bowel con- hours posttransplant. These episodes are transient and mild
tents, many centers also recommend antifungal prophy- without significant clinical consequence. Although most of
laxis with fluconazole; CNI serum levels must be monitored the time these episodes are self-limited and benign, occa-
closely when azoles are administered because of decreased sionally severe acute respiratory distress syndrome and
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 625

systemic inflammatory response syndrome can result in to 8% of cases with a higher incidence in uremic patients
cardiopulmonary instability which needs to be managed in than nonuremic patients and the majority of cases occur
the intensive care unit with standard measures. within the first 45 days (personal unpublished data).
This is generally self-limiting and will manifest as dimin-
ished hemoglobin level associated with hematochezia
HEMORRHAGE
or melanotic stool. Initial conservative management is
Bleeding postoperatively accounts for less than 1% of causes appropriate with IV octreotide drip (50–100 μcg/h) and
of graft losses.26 Blood transfusions postoperatively are not correction of abnormal anticoagulation, which is highly
uncommon, and although conservative measures usually effective in stopping the bleeding in the majority of cases.
suffice, the rate of relaparotomy for intraabdominal bleed- It is unusual for reoperative exploration to be required,
ing can be as high 16%.83 but if necessary an enterotomy and intraoperative endos-
copy can usually pinpoint the source and allow thera-
peutic measures.
ENTERIC CONVERSION
In a recent comprehensive review of the literature, urologic
complications account for more than 90% of the causes Induction and Maintenance
for enteric conversion.69 In our experience, in an attempt
to restore quality of life and correct serious urologic com-
Immunosuppression
plications as well as intractable metabolic acidosis, more
INDUCTION OPTIONS AND OUTCOMES
than half of the surviving patients underwent conversion
to enteric drainage. Within 5 years after transplanta- As pancreas transplant recipients experience higher rates of
tion, approximately 30% of the total number of surviving rejection than either kidney or liver recipients, the majority
patients underwent enteric conversion and within 15 years of pancreas transplants are performed using some form of
approximately 50% had undergone conversion.31 induction immunosuppression.25,85 Patients who appear to
have a higher risk of early rejection and appear to benefit
COMPLICATIONS OF THE ENTERIC-DRAINED most from induction therapy are those who are sensitized,
PANCREAS TRANSPLANT receiving solitary pancreas transplants, repeat transplants,
African American patients, or patients receiving positive
The most serious complication of the enteric-drained pan- crossmatch organs or those with positive pretransplant
creas transplant is that of an enteric leak and intraab- donor specific antibody.86
dominal abscess. This feared problem usually occurs 1 to 6 Based on the available evidence on comparisons of induc-
months posttransplant, yet is not necessarily the most com- tion therapy in SPK transplantation, one can conclude that
mon leak complication. Additional types of leaks are often alemtuzumab and antithymocyte globulin (ATG) are asso-
termed parenchymal leaks that do not involve the duode- ciated with the lowest rates of acute kidney, not pancreas,
nal segment. Patients present with fever, abdominal dis- rejection, at the cost of a higher rate of viral complications,
comfort, and leukocytosis, increased serum enzymes, and mainly in the form of CMV viremia. Despite a weak argu-
occasionally mild hyperglycemia. A high index of suspicion ment for induction to prevent early pancreas rejection,
is required to make a swift and accurate diagnosis. Imaging annual data reports from the OPTN/SRTR demonstrate that
studies involving a CT scan are very helpful. Surgical explo- most pancreas transplant recipients continue to receive
ration and repair of the enteric leak are felt to be necessary some form of induction therapy.25,26,87 The latest OPTN/
in our experience but abscess and parenchymal enzyme SRTR data report demonstrates that T cell depleting anti-
leaks can often be managed without reexploration depend- body use remained at approximately 80%, whereas the use
ing on clinical status. A decision must be made whether the of IL-2 receptor antagonists is declining. PAK transplants
infection can be eradicated without removing the pancreas generally have higher rates of rejection than SPK patients
allograft. Anastomotic leak and intraabdominal infection and T cell depleting agents are frequently used in this set-
account for less than 1% of graft losses according to national ting as well.
data.25,26 Incomplete eradication of the infection will result Immunosuppression in the UK follows similar lines to
in progression to sepsis and multiple organ system failure. practice in North America. Antibody induction is univer-
Peripancreatic infections can result in development of a sal, with the majority of UK units using alemtuzumab and
mycotic aneurysm, potentially giving rise to intraabdomi- greater use of polyclonal antithymocyte globulin in other
nal or intraluminal bleeding. Transplant pancreatectomy European countries; others use basiliximab.
is indicated if mycotic aneurysm is diagnosed. Several risk
factors have been identified to associate with a higher risk
MAINTENANCE IMMUNOSUPPRESSION
of intraabdominal infections including: older donor age,
longer preservation time, older recipient age, enteric drain- Multiple studies have demonstrated that the risk of acute
age versus bladder drainage, vascular graft thrombosis, and rejection in SPK transplant recipients is reduced using
retransplants.84 MMF instead of azathioprine.88–90 A multicenter prospec-
Early gastrointestinal bleeding occurs after the enteric- tive randomized study of tacrolimus vs. cyclosporine in SPK
drained pancreas from a combination of perioperative transplant recipients receiving MMF, steroids, and induc-
anticoagulation and bleeding from the suture line of the tion therapy demonstrated improved pancreas allograft
duodenoenteric anastomosis. Despite a running suture functional survival and lower rates of rejection for recipi-
line on the duodenojejunostomy, we observed this in up ents receiving tacrolimus.91 Hence, the combination of
626 Kidney Transplantation: Principles and Practice

MMF and tacrolimus has become the preferred modality of


1
maintenance immunotherapy in all pancreas transplant Reperfusion Injury
scenarios,92–95 and this is standard practice in Europe, too.

Relative probability
Ent/Pa Leak ACR/AMR
The use of sirolimus as a solo agent, or as a replacement
to tacrolimus, has fallen out of favor over the past decade to
<10% of transplants receiving it currently. This decrease is
arguably due to less well tolerated side effects (and wound- Ileus SBO
Other
related complications) without the advantage of better
outcomes.96
0 1 2 3 6 12 18 24
Months posttransplant
RESULTS OF EARLY STEROID WITHDRAWAL Fig. 36.17 Temporal relationship of elevated pancreas enzymes to eti-
Steroid withdrawing/rapid tapering has shown promis- ology. Surgical complications typically present early in the postopera-
tive course, whereas rejection most commonly presents later. “Other”
ing results compared with conventional steroid mainte- includes possible causes, such as transplant pancreatic duct stricture,
nance regimens. Most recent studies of induction therapy native pancreatitis, intraductal papillary mucinous neoplasms or can-
in pancreas transplantation have focused on alemtuzumab cer in pancreas transplant or native pancreas, and penetrating ulcer.
induction and early steroid elimination. In all of the stud- Of note, although complete graft thrombosis can present as increased
pancreatic enzymes, in our experience, this is a very uncommon pre-
ies evaluated on this topic, patient survival exceeds 95% sentation, and when most grafts thrombose, very limited increases in
at 1 year, and 1-year allograft survival of the pancreas is enzymes are seen, if at all. Relative probability on the y-axis does not
greater than 85%.97–100 In a small randomized controlled represent the overall probability or incidence of this complication;
trial comparing alemtuzumab versus ATG induction in instead, it strives to convey the relative probability that these diag-
SPK recipients with the intent of steroid-sparing tacroli- noses are associated with elevated pancreatic enzymes. ACR, acute
cellular rejection; Ent/Pa Leak, enteric or pancreatic leak; SBO, small
mus monotherapy in the alemtuzumab group, graft sur- bowel obstruction. (Reproduced from Redfield RR, Kaufman DB, Odorico
vival and rejection rates were similar between groups, JS. Diagnosis and treatment of pancreas rejection. Curr Transplant Rep
and most patients receiving alemtuzumab and tacrolimus 2015;2:169.)
monotherapy remained steroid free at 1-year posttrans-
plant.101,102 Two further studies comparing alemtuzumab
Elevated Amylase/Lipase
versus ATG found no difference in patient- or graft-related
outcomes. However, there was a higher incidence of CMV CT Abdomen/C·peptide/Hb A1C
infections in the thymoglobulin-treated group,99 and gly-
cated hemoglobin levels were lower in the alemtuzumab •Negative for Intraabdominal Pathology •Hyperglycemia
•Normal pancreas size •Exogenous insulin requirement
group compared with the ATG group.98 Although the evi- •Functional pancreas •Small pancreas
dence supports good outcomes in pancreas transplantation
with early steroid elimination/withdrawal especially in the •Ultrasound Guided Core Needle biopsy
•DSA
Consider No Therapy

case of alemtuzumab induction, late steroid withdrawal •C4d


(>1 year) is associated with a high rate of chronic rejection
ACMR MIXED aAMR cAMR
in kidney transplant recipients and should only be under-
taken in specific circumstances in the setting of pancreas
transplantation. •Grade 1 – Steroids, if no
response ATG (1.5 mg/kg)
Treatment of ACMR
add IVIG/PP
IVIG/PP ?
•Grade 2 – Steroids and
ATG (5–7doses)

Rejection Rates, Diagnosis, and •Grade 3 – Steroids and


ATG (7doses)
If plateau in improvement or
refractory → re-biopsy
Need for anti-B Cell
Therapies

Management Fig. 36.18 The University of Wisconsin Diagnosis and Treatment


Algorithm. aAMR, acute antibody-mediated rejection; ACMR, acute
DIFFERENTIAL DIAGNOSIS OF INCREASED cell-mediated rejection; ATG, antithymocyte globulin; cAMR, chronic
antibody-mediated rejection; CT, computed tomography; DSA, donor-
PANCREATIC ENZYMES specific antibody; IVIG, intravenous immunoglobulin; PP, plasma-
pheresis. (Reproduced from Redfield RR, McCune KR, Sollinger HW, et al.
Symptomatic increases in pancreatic enzymes can point to Pancreas allograft antibody mediated rejection: current concepts and
either transplant or native gland disease. Because signs and future therapies. Trends Transplant 2014.)
symptoms of pancreatic graft dysfunction can arise from
multiple causes in the setting of an enteric-drained pancreas pancreatic allograft rejection have relatively few symptoms
graft, it is helpful to consider both the timing of presenta- or are completely asymptomatic.
tion and the differential diagnosis when evaluating these Duodenal or parenchymal enzyme leak, pseudocyst,
patients. Fig. 36.17 demonstrates the temporal relationship mycotic aneurysm, small bowel obstruction (SBO), intrinsic
of elevated pancreas enzymes related to possible underlying pancreatic abnormalities such as duct strictures, and tumors
etiologies.22 Clinically, pancreas allograft function is com- are included in the differential diagnosis of elevated enzymes
monly monitored by serum glucose, serum amylase, serum and are usually diagnosed with imaging studies. Table 36.2
lipase, C-peptide level, HbA1c, or, if bladder-drained, uri- depicts the common diagnoses contributing to increased
nary amylase. In our practice (Fig. 36.18), the approach to pancreatic enzymes after pancreas transplantation. Regard-
the patient with elevated enzymes is history and physical to less of the time frame posttransplant, a pancreas allograft
determine whether the patient is asymptomatic or has ten- biopsy and CT scan will efficiently identify the cause for ele-
derness over the graft. The majority of patients with acute vated pancreatic enzymes in the vast majority of cases.22 In
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 627

TABLE 36.2 Common Diagnoses Contributing Another approach is laparoscopic biopsy. In a series of
to Increased Pancreatic Enzymes After Pancreas more than 100 patients, the authors have shown that the
Transplantation laparoscopic approach to kidney and pancreas allograft
biopsies can be safely performed with high tissue yields.103
Frequency the
Overall Entity Presents as
Other methods that have been described include percuta-
Diagnosis Frequency (%) Increased Enzymes neous CT-guided biopsy, endoscopic biopsy of the donor
duodenum for enteric-drained pancreata, or, if bladder-
PERIOPERATIVE (<45 DAYS)
drained, transcystoscopic needle biopsy of pancreatic head
Enzyme leaka ∼5 High and/or mucosal biopsy of the donor duodenum.
Infected fluid/abscess ∼5 Moderate
Thrombosis 2–5 Low
Ileus 5–10, transient Moderate PANCREAS REJECTION: BANFF CRITERIA,
Acute rejection 1–2 High
DEFINITION OF ANTIBODY MEDIATED
MID-POSTOPERATIVE (>45 DAYS–1 YEAR) REJECTION, AND TREATMENT
Acute rejectionb 15–20 High
SBO 5 Low-moderate Rejection is most often heralded by an increase in pancre-
Pseudocyst 2–5 High atic enzymes, with lipase being more sensitive than amy-
Constipation 2–5 Low-moderate lase; most patients are asymptomatic on presentation.
Abscess 2–5 Low-moderate
CMV pancreatitis ∼1 High When asymptomatic, the source of the increased enzymes
is often from the insensate transplanted pancreas, but
LATE POSTOPERATIVE (>1 YEAR)
unfortunately increased enzymes are not a specific marker
Acute rejection 5–10 High
Chronic rejection ∼5 Moderate-high
for rejection. The 2007 Banff guidelines for the diagnosis of
SBO/ventral hernia ∼10 Low-moderate pancreas rejection solidified prior histopathologic descrip-
Intrinsic pancreatic ∼5 Moderate-high tions and focused on acute cellular rejection (ACR).104–106
abnormalityc However, in response to reports in the literature document-
Native pancreatitis 1 High ing pancreas antibody-mediated rejection (AMR), a 2011
CMV pancreatitis <1 High
Banff update established comprehensive guidelines for diag-
aEnzyme leak—enteric or parenchymal, including pseudocyst or pancreatic nosis of acute and chronic AMR23,107 of the pancreas.
ascites
bFrequency depends on risk factors such as type of transplant, primary vs.

retransplant, sensitized status, donor-specific antibody, race mismatch, IMMUNOLOGIC CHALLENGES: DONOR SPECIFIC
and donor age ANTIBODIES, DISCORDANT KIDNEY AND
cIncludes graft trauma, chemical pancreatitis, pancreatic duct stricture, Intra-
PANCREAS BIOPSIES
ductal papillary mucinous neoplasm (IPMN), carcinoma, pseudocyst
CMV, cytomegalovirus; SBO, small bowel obstruction. The deleterious effects of both preformed donor-specific anti-
Reproduced from Redfield RR, Kaufman DB, Odorico JS. Diagnosis and treat- bodies (DSA) and de novo DSA are well established in the
ment of pancreas rejection. Curr Transplant Rep. 2015;2(2):169-75.
kidney transplant literature108; this is less well studied in pan-
creas grafts. The rate of de novo DSA in pancreas transplants
our experience, transplant pancreatitis that is not attribut- may be as high as 20% to 40%, but the rate may be influ-
able to an anatomic or immunologic cause is uncommon; enced by immunosuppressive management and recipient
beyond the first few days posttransplant, transplant pancre- characteristics.109,110 As for the kidney transplant popula-
atitis should be considered a diagnosis of exclusion. tion, most centers will now monitor DSA levels postopera-
tively with the frequency of monitoring either by protocol or
PANCREAS TRANSPLANT BIOPSY determined by a preoperative risk stratification. However, it
is currently unknown what to do with de novo DSA in the
Pancreas allograft biopsies are typically performed for spe- setting of normal pancreas allograft function and normal
cific indications, such as elevated pancreatic enzymes, fever pancreatic enzymes, and whether de novo DSA, either in the
of unknown origin, or mild hyperglycemia. Percutaneous setting of rejection or in the absence of rejection, portends
biopsies are most commonly done using real-time ultra- a worse outcome in the pancreas allograft, similar to what
sound or CT fluoroscopy guidance. With experience, biop- studies have shown for kidney and heart de novo DSA.
sies can be performed with low risk. We have performed well
over 400 percutaneous biopsies with a low complication
rate and no graft losses.22 Only two patients to date have Pancreas Transplant Outcomes
required reoperation: one for bleeding and one for control
of pancreatic ascites, which ultimately resolved, and both Patient survival remains exceedingly high for all three cat-
patients currently still retain excellent graft function. We pre- egories of pancreas transplants, with 1- and 3-year patient
fer ultrasound-guided biopsy with an 18-gauge automatic survival rates at 96.3% and 94.9% for SPK, 97.9% and
biopsy device. Our trajectory for biopsy is ideally toward the 94.5% for PAK, and 96.3% and 94.9% for PTA (Fig. 36.19)
tail, avoiding the splenic artery and vein. Because of the nest in the latest era (2010–2014).26 For uremic diabetes, data
of vessels and larger duct in the head region, we prefer to from the United States Renal Data System (USRDS) has
biopsy the body/tail of the graft, aiming either transversely shown a 10-year survival rate of 9.6% to 11.2% at best
or preferably longitudinally with respect to the axis of the for patients on hemodialysis without a kidney transplant.
pancreas. The following video illustrates a real-time ultra- Whether a pancreas transplant confers an effect on survival
sound guided pancreas allograft biopsy (video 1). probabilities for patients with diabetes was initially clouded.
628 Kidney Transplantation: Principles and Practice

Tx Type=SPK 5
100
1966–1987 PAK
1988–1994
1995–2001 4 PTA
80
2002–2008

Relative hazard ratio


SPK
Patient Survival [%]

2009–2016
60
3

40
2
20
Wait-listed patients
1
0
0 5 10 15 20 25
Years posttransplant
0
Fig. 36.19 Patient survival rates for primary deceased donor US 0 50 100 150 200 250 300 350
simultaneous pancreas and kidney (SPK) transplants performed from Days since transplantation
January 1, 1966 to December 31, 2016. (Reproduced with permission of
professor Angelika Grussner (data presented at IPITA 2017 meeting) Fig. 36.21 Mortality hazard ratios by recipient category. PAK, pancreas
after kidney transplant; PTA, pancreas transplant alone; SPK, simultane-
ous pancreas and kidney. (Reproduced from Gruessner RW, Sutherland
DE, Gruessner AC. Mortality assessment for pancreas transplants. Am J
100 Transplant 2004;4(12):2018–26.)

80
100

60
Percent

80

40 Survival
Survival [%]

Categories n 1 yr 4 yr 60
SPK 6995 97.5% 90.7%
20 Wait 5536 87.2% 46.0% 40

0 20
KTA
0 12 24 36 48
PTA & PAK
Months waiting SPK
0
Fig. 36.20 Patient survival after simultaneous pancreas and kidney 0 1 2 3 4 5
(SPK) transplantation versus those waiting for an SPK transplant.
(Reproduced from Gruessner RW, Sutherland DE, Gruessner AC. Mortality Time on waitlist (years)
assessment for pancreas transplants. Am J Transplant 2004;4(12):2018–26.)
Fig. 36.22 Waitlist survival for diabetic patients on the waitlist for
transplantation for US primary DD pancreas and kidney transplants,
January 1, 2005–December 31, 2016. DD, KTA, PAK, pancreas after kid-
In two separate analyses of UNOS data looking at two differ- ney; PTA, pancreas transplant alone; SPK, simultaneous pancreas and
ent eras, the initial analysis111 (1995–2000) concluded at kidney. (Reproduced from professor Angelika Gruessner with permission.)
4 years; SPK recipients were found to have a significantly
higher probability of survival than patients who remained transplantation (n = 256) or LDK transplantation alone
on the waitlist, but the opposite was true for PTA and PAK (n = 230) between 1983 and 2012.113 With a median
recipients, drawing the conclusion that survival for those follow-up of 7.9 years, and after adjustment for donor and
with diabetes and preserved kidney function and receiving recipient risk factors, the HR for cardiovascular-related
a solitary pancreas transplant was significantly worse com- death in SPK recipients compared with LDK recipients was
pared with the survival of waitlist patients receiving con- 0.63 (P = 0.047). The Oslo group has also provided evi-
ventional therapy. However, after eliminating patients who dence for long-term benefit of pancreas transplant function
were listed at multiple centers and patients who changed with respect to renal function and the ultrastructure of the
transplant centers in the previous analysis (at least 12% kidney. Twenty-five SPK and 17 LDK recipients, similar in
of patients, inherently a substantial bias), a second analy- age and duration of diabetes, were followed for a median
sis112 (1995–2003) demonstrated that overall mortality in of 10.1 years. As expected, mean HbA1c levels were lower
all three categories was not increased after transplantation, in the SPK patients (5.5% vs. 8.3%). Biochemical studies
and for SPK recipients it was significantly decreased com- showed less GFR decline from baseline (11 ± 21 vs. 23 ±
pared with waitlist mortality. Therefore pancreas trans- 15 mL/min, P = 0.060). Kidney biopsies showed that the
plantation is justified for patient survival, especially for SPK glomerular basement membrane was significantly wider
transplants (Figs. 36.20–36.22). (P = 0.008) and the mesangial volume fraction was signifi-
A single-center analysis from one of Europe’s most active cantly higher (P = 0.007) in the kidney-alone recipients.
pancreas transplant centers, Oslo University Hospital, The (negative) slope of eGFR was also significantly reduced
compared cardiovascular mortality in 486 patients with in the SPK recipients (–1.1 mL/min/year) compared with
renal failure and type 1 diabetes, undergoing either SPK kidney-alone recipients (–2.6 mL/min/year).114
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 629

Effect on Quality of Life and the effect of reversal of autonomic neuropathy in pan-
creas transplant recipients has been associated with bet-
End-Organ Complications ter patient survival rates than in patients with failed or no
transplantation.120,121
QUALITY OF LIFE STUDIES
Retinopathy
The quality of life of pancreas transplant recipients has
been well-studied historically.115–117 Patients with a func- The data on retinopathy reversal after pancreas transplan-
tioning pancreas graft describe their quality of life and rate tation are mixed.122–125 Pancreas transplantation does not
their health significantly more favorably than those with appear to have an immediate, dramatic beneficial effect on
nonfunctioning pancreas grafts. Satisfaction encompasses preestablished diabetic retinopathy. Retinopathy appears to
not only the physical capacities, but also relate to psychoso- progress for at least 2 years after transplantation of the pan-
cial and vocational aspects. The functioning pancreas graft creas, but begins to stabilize in 3 to 4 years compared with
leads to even better quality of life compared with recipients diabetic recipients of a kidney transplant only. Longer-term
of a kidney transplant alone.115 studies of 5 to 10 years have not been reported. For many
patients with well-treated and stabilized retinopathy, nor-
malization of blood glucose levels might only be expected
END-ORGAN COMPLICATIONS to have a neutral effect, whereas a beneficial long-term
Recipients of a successful pancreas transplant maintain effect might be observed in very early retinopathic changes.
normal plasma glucose levels without the need of exog- Other causes of visual disturbances in this patient popula-
enous insulin therapy. This results in normalization of gly- tion, such as cataracts, may confound visual acuity studies.
cosylated hemoglobin levels and a beneficial effect on many
secondary complications of diabetes. The durability of the
transplanted endocrine pancreas has been established, Summary
with the current expected half-life for an SPK pancreatic
graft being 15 years.25 The implications of prolonged nor- Pancreas transplantation is a proven therapeutic treat-
malization of glycemia and glycosylated hemoglobin levels ment option for diabetes, especially simultaneously with
are significant and may benefit in the long-term retinal, or after kidney transplantation. It is superior to insulin
nephrologic, neurologic, and macrovascular complications therapies with regard to the efficacy of achieving glycemic
of diabetes. control. It can enhance quality of life and numerous stud-
ies have demonstrated beneficial effects on diabetic second-
Nephropathy ary complications, including prevention of recurrent DM
The development of diabetic nephropathy in transplanted nephropathy in the renal allograft. A pancreas transplant
kidneys residing in patients with T1D has been well estab- routinely produces an immediate normoglycemic and
lished. There is marked variability in the rate of acquisition insulin-independent state that normalizes HbA1c levels
of renal pathology in patients with T1D and a kidney trans- with significant anticipated long-term benefits. As immu-
plant alone. The onset of pathologic lesions can be detected nologic and technical graft failures have declined in recent
as early as a few years after kidney transplantation. Clinical years and overall graft outcomes have improved, pancreas
deterioration of renal allograft function can lead to loss 10 transplantation can be offered to insulin-dependent dia-
to 15 years posttransplant. Pancreas transplantation is the betics with broadening indications and eligibility criteria
only available treatment which has restored long-term (10 while balancing risk versus benefit. Pancreas transplan-
or more years) normoglycemia without the risks of severe tation should be in the armamentarium of every trans-
hypoglycemia, allowing testing of the reversibility or pre- plant center for the treatment of diabetic patients, yet it is
vention of recurrent diabetic nephropathy lesions. Diabetic increasingly being practiced at specialized centers where
glomerular lesions were not significantly changed at 5 experience is associated with superior outcomes. Despite
years but were dramatically improved after 10 years, with advances in technology and improved insulin delivery
most patients’ glomerular structure returning to normal at devices, no current option mimics the nondiabetic state as
the 10-year follow-up.118 These studies also showed that well as pancreas transplantation does. In the future, pan-
tubulointerstitial remodeling, including decreased intersti- creas transplantation will likely continue to have a well-
tial collagen, was possible.118,119 defined clinical role for diabetic patients and will remain an
important option in the treatment of diabetes and diabetic
Neuropathy nephropathy.
Successful pancreas transplantation has been shown to halt,
and in many cases, reverse motor-sensory and autonomic References
neuropathy as early as 12 to 24 months posttransplant.
1. Miller KM, et al. Current state of type 1 diabetes treatment in the
This has been studied most extensively in recipients of SPK U.S.: updated data from the T1D exchange clinic registry. Diabetes
transplants. This raises the possibility that improvement of Care 2015;38(6):971–8.
diabetic neuropathy occurs, in part, due to improvement 2. Lasker RD. The diabetes control and complications trial: implications
of uremic neuropathy. However, pancreas transplantation for policy and practice. N Engl J Med 1993;329(14):1035–6.
3. Karamanou M, et al. Milestones in the history of diabetes mellitus:
alone in preuremic patients has also been shown to result the main contributors. World J Diabetes 2016;7(1):1–7.
in improvement in diabetic neuropathy. Many patients 4. Sutherland DE. In: Gruessner W, editor. History of pancreas trans-
express subjective improvements of peripheral sensation 6 plantation in transplantation of the pancreas. New York: Springer-
to 12 months after pancreas transplantation. Interestingly, Verlag; 2004.
630 Kidney Transplantation: Principles and Practice

5. Banting FG, Best CH. The internal secretion of the pancreas: univer- 36. Wolfe RA, et al. Comparison of mortality in all patients on dialysis,
sity of toronto studies physiological series. Toronto: The University patients on dialysis awaiting transplantation, and recipients of a first
Library; 1922. p. 16. cadaveric transplant. N Engl J Med 1999;341(23):1725–30.
6. Banting FG, et al. The effect produced on diabetes by extracts of pan- 37. Sampaio MS, et al. Transplantation with pancreas after living donor
creas. Toronto: The University Library; 1923. p. 11. kidney vs. living donor kidney alone in type 1 diabetes mellitus recip-
7. Bergan JJ, et al. Total pancreatic allografts in pancreatectomized ients. Clin Transplant 2010;24(6):812–20.
dogs. Arch Surg 1965;90:521–6. 38. Browne S, et al. The impact of pancreas transplantation on kidney
8. Brooks JR, Gifford GH. Pancreatic homotransplantation. Transplant allograft survival. Am J Transplant 2011;11(9):1951–8.
Bull 1959;6(1):100–3. 39. Cryer PE. Mechanisms of hypoglycemia-associated autonomic fail-
9. Coppola ED, Dejode LR, Howard JM. Studies of homotransplantation ure in diabetes. N Engl J Med 2013;369(4):362–72.
of the kidney in dogs treated with an experimental cancer chemo- 40. Cryer PE. Death during intensive glycemic therapy of diabetes:
therapeutic agent (AB-103). Surg Gynecol Obstet 1962;115:80–8. mechanisms and implications. Am J Med 2011;124(11):993–6.
10. Dejode LR, Howard JM. Studies in pancreaticoduodenal homotrans- 41. Cryer PE. Hypoglycemia in diabetes: pathophysiological mecha-
plantation. Surg Gynecol Obstet 1962;114:553–8. nisms and diurnal variation. Prog Brain Res 2006;153:361–5.
11. Largiader F, et al. Orthotopic allotransplantation of the pancreas. 42. Gruessner RW, et al. Over 500 solitary pancreas transplants in
Am J Surg 1967;113(1):70–6. nonuremic patients with brittle diabetes mellitus. Transplantation
12. Idezuki Y, et al. Experimental pancreaticoduodenal preservation and 2008;85(1):42–7.
transplantation. Surg Gynecol Obstet 1968;126(5):1002–14. 43. Scalea JR, et al. Pancreas transplant alone as an independent risk
13. Sutherland DE. Pancreas and islet transplantation. II. Clinical trials. factor for the development of renal failure: a retrospective study.
Diabetologia 1981;20(4):435–50. Transplantation 2008;86(12):1789–94.
14. Sutherland DE. Pancreas and islet transplantation. I. Experimental 44. Mehrabi A, et al. Expanding the indications of pancreas transplanta-
studies. Diabetologia 1981;20(3):161–85. tion alone. Pancreas 2014;43(8):1190–3.
15. Kelly WD, et al. Allotransplantation of the pancreas and duo- 45. Ris F, et al. Islet autotransplantation after extended pancreatec-
denum along with the kidney in diabetic nephropathy. Surgery tomy for focal benign disease of the pancreas. Transplantation
1967;61(6):827–37. 2011;91(8):895–901.
16. Lillehei RC, et al. Pancreatico-duodenal allotransplantation: experi- 46. Bramis K, et al. Systematic review of total pancreatectomy and
mental and clinical experience. Ann Surg 1970;172(3):405–36. islet autotransplantation for chronic pancreatitis. Br J Surg
17. Lillehei RC, et al. Transplantation of the pancreas. Acta Endocrinol 2012;99(6):761–6.
Suppl (Copenh) 1976;205:303–20. 47. Bedat B, et al. Impact of recipient body mass index on short-term
18. Sollinger HW, et al. Clinical and experimental experience with pan- and long-term survival of pancreatic grafts. Transplantation
creaticocystostomy for exocrine pancreatic drainage in pancreas 2015;99(1):94–9.
transplantation. Transplant Proc 1984;16(3):749–51. 48. Fridell JA, et al. Growth of a nation part II: impact of recipient obe-
19. Calne RY. Paratopic segmental pancreas grafting: a technique with sity on whole-organ pancreas transplantation. Clin Transplant
portal venous drainage. Lancet 1984;1(8377):595–7. 2011;25(4):E366–74.
20. Niederhaus SV, Kaufman DB, Odorico JS. Induction therapy in pan- 49. Hanish SI, et al. Obesity predicts increased overall complica-
creas transplantation. Transpl Int 2013;26(7):704–14. tions following pancreas transplantation. Transplant Proc
21. Ellis TM. Interpretation of HLA single antigen bead assays. Trans- 2005;37(8):3564–6.
plant Rev (Orlando) 2013;27(4):108–11. 50. Rogers J, et al. Influence of mild obesity on outcome of simulta-
22. Redfield RR, Kaufman DB, Odorico JS. Diagnosis and treatment of neous pancreas and kidney transplantation. J Gastrointest Surg
pancreas rejection. Curr Transplant Rep 2015;2(2):169–75. 2003;7(8):1096–101.
23. Drachenberg CB, et al. Guidelines for the diagnosis of antibody-medi- 51. Laurence JM, et al. Optimizing pancreas transplantation outcomes in
ated rejection in pancreas allografts-updated Banff grading schema. obese recipients. Transplantation 2015;99(6):1282–7.
Am J Transplant 2011;11(9):1792–802. 52. Porubsky M, et al. Pancreas transplantation after bariatric surgery.
24. Squifflet JP, Gruessner RW, Sutherland DE. The history of pan- Clin Transplant 2012;26(1):E1–6.
creas transplantation: past, present and future. Acta Chir Belg 53. Burke III GW, et al. Recurrence of autoimmunity following pancreas
2008;108(3):367–78. transplantation. Curr Diab Rep 2011;11(5):413–9.
25. Kandaswamy R, et al. OPTN/SRTR. 2015 Annual data report: pan- 54. Clarke WL, et al. Reduced awareness of hypoglycemia in adults with
creas. Am J Transplant 2017;17(Suppl. 1):117–73. IDDM: a prospective study of hypoglycemic frequency and associated
26. Gruessner AC, Gruessner RW. Pancreas transplantation of US and symptoms. Diabetes Care 1995;18(4):517–22.
Non-US cases from 2005 to 2014 as reported to the United Network 55. Fernandez LA, et al. Superior long-term results of simultaneous
for Organ Sharing (UNOS) and the International Pancreas Trans- pancreas-kidney transplantation from pediatric donors. Am J Trans-
plant Registry (IPTR). Rev Diabet Stud 2016;13(1):35–58. plant 2004;4(12):2093–101.
27. (EDQM), E.D.f.t.Q.o.M.a.H. 2017; Available online at: https://www. 56. Finger EB, et al. A composite risk model for predicting technical
edqm.eu/en/organ-transplantation-reports-73.html. Accessed May failure in pancreas transplantation. Am J Transplant 2013;13(7):
10, 2017. 1840–9.
28. NHSBT, A.r.o.p.a.i.t. 2017. Accessed May 10, 2017. 57. Axelrod DA, et al. Systematic evaluation of pancreas allograft qual-
29. Redfield RR, et al. Pancreas transplantation in the modern era. Gas- ity, outcomes and geographic variation in utilization. Am J Trans-
troenterol Clin North Am 2016;45(1):145–66. plant 2010;10(4):837–45.
30. Stratta RJ. The future of pancreas transplantation: ‘ain’t what it used 58. Arpali E, et al. The importance and utility of hemoglobin A1c Lev-
to be’. Curr Opin Organ Transplant 2016;21(4):375–6. els in the assessment of donor pancreas allografts. Transplantation
31. Sollinger HW, et al. One thousand simultaneous pancreas-kidney 2017;101(10):2508–19.
transplants at a single center with 22-year follow-up. Ann Surg 59. Fernandez LA, et al. Simultaneous pancreas-kidney transplantation
2009;250(4):618–30. from donation after cardiac death: successful long-term outcomes.
32. Light J, Tucker M. Simultaneous pancreas kidney transplants in dia- Ann Surg 2005;242(5):716–23.
betic patients with end-stage renal disease: the. 20-yr experience. 60. Muthusamy AS, et al. Pancreas transplantation from donors after
Clin Transplant 2013;27(3):E256–63. circulatory death from the united kingdom. Am J Transplant
33. Light JA, Barhyte DY. Simultaneous pancreas-kidney transplants in 2013;13(3):824.
type I and type II diabetic patients with end-stage renal disease: simi- 61. Stewart ZA, et al. Histidine-tryptophan-ketoglutarate (HTK) is asso-
lar 10-year outcomes. Transplant Proc 2005;37(2):1283–4. ciated with reduced graft survival in pancreas transplantation. Am J
34. Stratta RJ, et al. Pancreas transplantation in C-peptide positive Transplant 2009;9(1):217–21.
patients: does “type” of diabetes really matter? J Am Coll Surg 62. Rao PS, et al. A comprehensive risk quantification score for deceased
2015;220(4):716–27. donor kidneys: the kidney donor risk index. Transplantation
35. Kaufman DB, Sutherland DE. Simultaneous pancreas-kidney trans- 2009;88(2):231–6.
plants are appropriate in insulin-treated candidates with uremia 63. Feng S, et al. Characteristics associated with liver graft failure: the
regardless of diabetes type. Clin J Am Soc Nephrol 2011;6(5):957–9. concept of a donor risk index. Am J Transplant 2006;6(4):783–90.
36 • Pancreas and Kidney Transplantation for Diabetic Nephropathy 631

64. Mittal S, et al. Validation of the pancreas donor risk index for use in a 91. Bechstein WO, et al. Efficacy and safety of tacrolimus compared with
UK population. Transpl Int 2015;28(9):1028–33. cyclosporine microemulsion in primary simultaneous pancreas-
65. Muthusamy AS, et al. Pancreas transplantation from donors after kidney transplantation: 1-year results of a large multicenter trial.
circulatory death from the united kingdom. Am J Transplant Transplantation 2004;77(8):1221–8.
2012;12(8):2150–6. 92. Stratta RJ. Simultaneous use of tacrolimus and mycophenolate
66. Ausania F, et al. A registry analysis of damage to the deceased donor pan- mofetil in combined pancreas-kidney transplant recipients: a multi-
creas during procurement. Am J Transplant 2015;15(11):2955–62. center report. the fk/mmf multi-center study group. Transplant Proc
67. White SA, Shaw JA, Sutherland DE. Pancreas transplantation. Lan- 1997;29(1-2):654–5.
cet 2009;373(9677):1808–17. 93. Gruessner RW, et al. A multicenter analysis of the first experience
68. Vrakas G, et al. Solitary pancreas transplantation: a review with FK506 for induction and rescue therapy after pancreas trans-
of the UK experience over a period of 10 yr. Clin Transplant plantation. Transplantation 1996;61(2):261–73.
2015;29(12):1195–202. 94. Bruce DS, et al. Tacrolimus/mycophenolate provides superior immu-
69. El-Hennawy H, Stratta RJ, Smith F. Exocrine drainage in vascu- nosuppression relative to neoral/mycophenolate in synchronous pan-
larized pancreas transplantation in the new millennium. World J creas-kidney transplantation. Transplant Proc 1998;30(4):1538–40.
Transplant 2016;6(2):255–71. 95. Kaufman DB, et al. Mycophenolate mofetil and tacrolimus as pri-
70. De Roover A, et al. Pancreas graft drainage in recipient duodenum: mary maintenance immunosuppression in simultaneous pancreas-
preliminary experience. Transplantation 2007;84(6):795–7. kidney transplantation: initial experience in 50 consecutive cases.
71. Lindahl JP, et al. Outcomes in pancreas transplantation with exo- Transplantation 1999;67(4):586–93.
crine drainage through a duodenoduodenostomy versus duodenoje- 96. Berney T, Andres A, Toso C, Majno P, Squifflet JP. mTOR inhibi-
junostomy. Am J Transplant 2018;18(1):154–62. tion & clinical transplantation: pancreas & islet. Transplantation
72. Kopp W, et al. Center volume is associated with outcome after pan- 2018;102(2S Suppl. 1):S30–1. Available online at: https://doi.org/
creas transplantation within the eurotransplant region. Transplan- 10.1097/TP.0000000000001700.
tation 2017;101(6):1247–53. 97. Sundberg AK, et al. Pilot study of rapid steroid elimination with
73. Alhamad T, et al. Transplant center volume and the risk of pancreas alemtuzumab induction therapy in kidney and pancreas transplan-
allograft failure. Transplantation 2017;101(11):2757–64. tation. Transplant Proc 2005;37(2):1294–6.
74. Sollinger HW. Pancreatic transplantation and vascular graft throm- 98. Reddy KS, et al. Alemtuzumab with rapid steroid taper in simultane-
bosis. J Am Coll Surg 1996;182(4):362–3. ous kidney and pancreas transplantation: comparison to induction
75. Benedetti E, et al. Intra-abdominal fungal infections after pancreatic with antithymocyte globulin. Transplant Proc 2010;42(6):2006–8.
transplantation: incidence, treatment, and outcome. J Am Coll Surg 99. Kaufman DB, et al. Alemtuzumab induction and prednisone-free
1996;183(4):307–16. maintenance immunotherapy in simultaneous pancreas–kidney
76. Papalois BE, et al. Long-term peritoneal dialysis before transplan- transplantation comparison with rabbit antithymocyte globulin
tation and intra-abdominal infection after simultaneous pancreas- induction – long-term results. Am J Transplant 2006;6(2):331–9.
kidney transplantations. Arch Surg 1996;131(7):761–6. 100. Burke GW, et al. Use of tacrolimus and mycophenolate mofetil for
77. Pirsch JD, et al. Posttransplant infection in enteric versus bladder- pancreas–kidney transplantation with or without OKT3 induction.
drained simultaneous pancreas-kidney transplant recipients. Trans- Transplant Proc 1998;30(4):1544–5.
plantation 1998;66(12):1746–50. 101. Margreiter R, et al. Alemtuzumab (Campath-1H) and tacrolimus
78. Everett JE, et al. Characterization and impact of wound infection monotherapy after renal transplantation: results of a prospective
after pancreas transplantation. Arch Surg 1994;129(12):1310–6. randomized trial. Am J Transplant 2008;8(7):1480–5.
discussion 1316-7. 102. Bösmüller C, Ollinger R, Sieb M, et al. Tacrolimus monotherapy fol-
79. Lowance D, et al. Valacyclovir for the prevention of cytomegalovirus lowing alemtuzumab induction in combined kidney–pancreas trans-
disease after renal transplantation. international valacyclovir cyto- plantation: results of a prospective randomized trial. Ann Transplant
megalovirus prophylaxis transplantation study group. N Engl J Med 2012;17(4):45–51.
1999;340(19):1462–70. 103. Uva PD, et al. Laparoscopic biopsies in pancreas transplantation. Am
80. Fishman JA. Infection in solid-organ transplant recipients. N Engl J J Transplant 2017;17(8):2173–7.
Med 2007;357(25):2601–14. 104. Drachenberg CB, et al. Evaluation of pancreas transplant needle
81. Fishman JA, AST Infectious Diseases Community of Practice. Intro- biopsy: reproducibility and revision of histologic grading system.
duction: infection in solid organ transplant recipients. Am J Trans- Transplantation 1997;63(11):1579–86.
plant 2009;9(Suppl. 4):S3–6. 105. Drachenberg CB, et al. Banff schema for grading pancreas allograft
82. Fishman JA, Rubin RH. Infection in organ-transplant recipients. N rejection: working proposal by a multi-disciplinary international
Engl J Med 1998;338(24):1741–51. consensus panel. Am J Transplant 2008;8(6):1237–49.
83. Troppmann C, et al. Surgical complications requiring early relapa- 106. Nakhleh RE, Sutherland DE. Pancreas rejection: significance of his-
rotomy after pancreas transplantation: a multivariate risk factor topathologic findings with implications for classification of rejection.
and economic impact analysis of the cyclosporine era. Ann Surg Am J Surg Pathol 1992;16(11):1098–107.
1998;227(2):255–68. 107. Niederhaus SV, et al. Acute cellular and antibody-mediated rejection
84. Gruessner RW, et al. The surgical risk of pancreas transplantation of the pancreas allograft: incidence, risk factors and outcomes. Am J
in the cyclosporine era: an overview. J Am Coll Surg 1997;185(2): Transplant 2013;13(11):2945–55.
128–44. 108. Mohan S, et al. Donor-specific antibodies adversely affect kidney
85. Kandaswamy R, et al. Reply to comment on the article “OPTN/ allograft outcomes. J Am Soc Nephrol 2012;23(12):2061–71.
SRTR.” 2015 annual data report: pancreas. Am J Transplant 109. Mittal S, et al. De novo donor-specific HLA antibodies: biomarkers of
2017;17(7):1954–5. pancreas transplant failure. Am J Transplant 2014;14(7):1664–71.
86. Sutherland DE, Gruessner A. In: Gruessner A, editor. Transplanta- 110. Cantarovich D, et al. Posttransplant donor-specific anti-HLA anti-
tion of the Pancreas. New York: Springer-Verlag; 2004. bodies negatively impact pancreas transplantation outcome. Am J
87. Gruessner AC. 2011 update on pancreas transplantation: compre- Transplant 2011;11(12):2737–46.
hensive trend analysis of 25,000 cases followed up over the course of 111. Venstrom JM, et al. Survival after pancreas transplantation in
twenty-four years at the International Pancreas Transplant Registry patients with diabetes and preserved kidney function. JAMA
(IPTR). Rev Diabet Stud 2011;8(1):6–16. 2003;290(21):2817–23.
88. Odorico JS, et al. A study comparing mycophenolate mofetil to aza- 112. Gruessner RW, Sutherland DE, Gruessner AC. Mortality assessment
thioprine in simultaneous pancreas-kidney transplantation. Trans- for pancreas transplants. Am J Transplant 2004;4(12):2018–26.
plantation 1998;66(12):1751–9. 113. Lindahl JP, et al. Long-term cardiovascular outcomes in type 1
89. Elkhammas EA, et al. Simultaneous pancreas/kidney transplanta- diabetic patients after simultaneous pancreas and kidney trans-
tion: comparison of mycophenolate mofetil versus azathioprine. plantation compared with living donor kidney transplantation. Dia-
Transplant Proc 1998;30(2):512. betologia 2016;59(4):844–52.
90. Merion RM, et al. Randomized, prospective trial of mycophenolate 114. Lindahl JP, et al. In patients with type 1 diabetes simultaneous pan-
mofetil versus azathioprine for prevention of acute renal allograft creas and kidney transplantation preserves long-term kidney graft
rejection after simultaneous kidney-pancreas transplantation. ultrastructure and function better than transplantation of kidney
Transplantation 2000;70(1):105–11. alone. Diabetologia 2014;57(11):2357–65.
632 Kidney Transplantation: Principles and Practice

115. Suzuki A, et al. Evaluation of quality of life after simultaneous pan- 121. Navarro X, Sutherland DE, Kennedy WR. Long-term effects of
creas and kidney transplantation from living donors using short pancreatic transplantation on diabetic neuropathy. Ann Neurol
form 36. Transplant Proc 2008;40(8):2565–7. 1997;42(5):727–36.
116. Sutherland DE, et al. Lessons learned from more than 1,000 pan- 122. Koznarova R, et al. Beneficial effect of pancreas and kidney trans-
creas transplants at a single institution. Ann Surg 2001;233(4): plantation on advanced diabetic retinopathy. Cell Transplant
463–501. 2000;9(6):903–8.
117. Ziaja J, et al. Impact of pancreas transplantation on the qual- 123. Ramsay RC, et al. Progression of diabetic retinopathy after pancreas
ity of life of diabetic renal transplant recipients. Transplant Proc transplantation for insulin-dependent diabetes mellitus. N Engl J
2009;41(8):3156–8. Med 1988;318(4):208–14.
118. Mauer M, Fioretto. Pancreas transplantation and reversal of diabetic 124. Wang Q, et al. The influence of combined kidney-pancreas trans-
nephropathy lesions. Med Clin North Am 2013;97(1):109–14. plantation on the progression of diabetic retinopathy. A case series.
119. Fioretto, et al. Reversal of lesions of diabetic nephropathy after pan- Ophthalmology 1994;101(6):1071–6.
creas transplantation. N Engl J Med 1998;339(2):69–75. 125. Pearce IA, et al. Stabilisation of diabetic retinopathy following
120. Kennedy WR, et al. Effects of pancreatic transplantation on diabetic simultaneous pancreas and kidney transplant. Br J Ophthalmol
neuropathy. N Engl J Med 1990;322(15):1031–7. 2000;84(7):736–40.
37 Kidney Transplantation in
Children
PAMELA D. WINTERBERG and ROUBA GARRO

CHAPTER OUTLINE Introduction Surgical Evaluation


Epidemiology of End-Stage Renal Disease Neurologic Development
in Children Psychosocial Issues
Incidence Evaluation Updates
Etiology Perioperative Management of Pediatric
Access to Transplantation Renal Transplant Recipients
Timing of Transplantation Preoperative Management
Patient and Graft Survival Intraoperative Management
Prognostic Factors Influencing Graft Survival Postoperative Management
Donor Source Immunosuppressive Protocols and Drugs
Recipient Age Induction Therapy Agents
Donor Age and Size Lymphocyte-Depleting Agents
Recipient Race Nondepleting Agents
HLA Matching Maintenance Therapy
Presensitization Corticosteroids
Delayed Graft Function and Thrombosis Calcineurin Inhibitors
Contraindications to Transplantation mTOR Inhibitors
Recurrence of Original Disease Antimetabolites
Primary Glomerulonephritis Acute Rejection in Pediatric
Focal Segmental Glomerulosclerosis Transplantation
Congenital Nephrotic Syndrome Diagnosis of Acute Rejection
Alport Syndrome Treatment of Acute Rejection
MPGN/C3 Glomerulopathy Acute Cellular Rejection
Secondary Glomerulonephritis Antibody-Mediated Rejection
IgA and Henoch-Schonlein Purpura Long-Term Management Posttransplant
Hemolytic-Uremic Syndrome Hypertension and Cardiovascular Disease
Membranous Nephropathy Infections After Transplantation
Systemic Lupus Erythematosis Viral Infections
c-ANCA and p-ANCA-Positive Bacterial Infections
Glomerulonephritis Lower Urinary Tract Symptoms
Metabolic Disease Growth and Bone Health
Primary Hyperoxaluria Type I (Oxalosis) Nonadherence in Pediatric Transplantation
Nephropathic Cystinosis Adolescence and Emerging Adulthood
Pretransplant Evaluation Psychosocial Development
Evaluation of Potential Living Donor Puberty
Evaluation of Recipient Sexuality and Body Image
Medical Evaluation of Issues Related to Transitional Care
ESRD
Evaluation of Extrarenal Disease

Introduction The medical and surgical care of ESRD and kidney trans-
plantation in children poses unique challenges. Growth and
Kidney transplantation is the preferred treatment for end- neurocognitive development are impaired during chronic
stage renal disease (ESRD) in children and confers improved kidney disease (CKD) and are a unique focus of pediatric
survival, skeletal growth, health-related quality of life, and nephrology care. The diagnosis of ESRD in children cre-
neuropsychological development compared with dialysis. ates an extra burden for caretakers and siblings. Therefore
633
634 Kidney Transplantation: Principles and Practice

treatment of ESRD and kidney transplant in the pediatric TABLE 37.1 Expected Remaining Lifetime (in years) of
population focuses on family-centered care and often uti- Prevalent Patients by Initial ESRD Modality as Reported in
lizes a multidisciplinary approach. Psychological develop- the 2017 USRDS Annual Report
ment is also addressed as children acquire the skills and
Dialysis Transplant General
attitudes needed to live an independent life as an adult with Age Group Patients Patients Population
a chronic medical condition.
Children who receive kidney transplants have longer 0–4 23.6 56.9 77.1
5–9 24.3 56.3 72.3
expected remaining lifetimes than adults at the time of kid- 10–13 24.1 52.2 67.8
ney transplant (Table 37.1).1 Therefore it is particularly 14–17 20.9 48.8 63.9
important to maximize graft function and graft survival in 18–21 17.7 45.2 60.0
this population. Children are also undergoing immune sys- 22–29 16.0 42.0 54.2
tem development and maturation at the time of transplant.
Data from: US Renal Data System. 2017 USRDS Annual Data Report: Epidemiol-
This, coupled with longer survival time, underscores the ogy of Kidney Disease in the United States. Bethesda, MD: National Institutes
importance of optimizing control of alloimmunity while min- of Health, National Institute of Diabetes and Digestive Kidney Diseases; 2017.
imizing side effects during long-term immunosuppression.
The number of children receiving kidney transplants
every year is small, and even the largest centers in the US (CoBrazPed-RTx) was established and has collected out-
rarely transplant more than 30 children per year. Therefore come data from 1751 pediatric kidney transplant recipients
it has been extremely important to maintain national and performed at 13 centers through December 2013.6
international databases to identify areas for research and Most figures and statistical references in this chapter are
improvement in outcomes among pediatric kidney trans- from the SRTR and NAPRTCS databases, but we also cite
plant recipients. literature from studies of other registries around the world.
There are two databases for pediatric kidney transplan-
tation in wide use in North America. The United Network
for Organ Sharing (UNOS) collects information for every Epidemiology of End-Stage Renal
kidney transplant in the US within the Organ Procure-
ment and Transplantation Network (OPTN). Waitlist,
Disease in Children
demographic, and survival statistics from this database are
INCIDENCE
reported annually through the Scientific Registry of Trans-
plant Recipients (SRTR) report. Although this registry con- Among adults, especially in developed countries, the prev-
tains pediatric recipients, there are several pediatric-specific alence of ESRD continues to increase, resulting in over-
variables (growth, for example) that are not reported in this whelming demand for kidney transplantation. For example,
registry. Furthermore, the definition of pediatric recipients in the US, there were 120,688 newly reported cases of ESRD
in SRTR is age less than 18 years, whereas at the time of in 2014 and 678,383 prevalent ESRD patients, including
writing, the US Renal Data System (USRDS), which reports those with a functioning graft. The incidence and preva-
on all forms of renal replacement therapy for ESRD in the lence of ESRD in children represents a very small fraction,
US, including kidney transplant, started including patients less than 2%, of the overall ESRD population in the US.
up to age 21 years in pediatric statistics. In 1987, the North Although the incidence of ESRD among adults has stabi-
American Pediatric Renal Trials and Collaborative Studies lized in recent years, the incidence among children has
(NAPRTCS) began a voluntary registry that included up decreased annually between 2008 and 2014.1
to 159 medical centers in the US, Canada, and Mexico. By
2010, the registry contained information for 11,603 kidney
ETIOLOGY
transplants in 10,632 children.2 More recently, the Improv-
ing Renal Outcomes Collaborative (www.irocnow.org) has Although diabetic nephropathy and hypertensive nephrop-
been established as a learning network focused on quality athy are the most common causes of ESRD in adults, these
improvement and research using chronic care models in diseases are rare in childhood. Rather, the most common
pediatric kidney transplant centers in North America. causes of ESRD in children are congenital, cystic, and
Other databases around the world have also been used to hereditary diseases, which combined account for 38% of
study risk factors and outcomes in pediatric kidney trans- incident cases.1 The most common hereditary disorders are
plantation. The Canadian Pediatric End-Stage Renal Dis- congenital obstructive uropathies (9.5%) and renal hypo-
ease database has been constructed by linking registry data plasia/dysplasia (10%). Primary glomerular disease is the
with administrative data from their universal health care second most common etiology, accounting for 25% of new
delivery system to study outcomes for children with ESRD cases, predominantly due to focal segmental glomeruloscle-
in Canada.3 The European Renal Association – European rosis (FSGS). Secondary glomerulonephritis and vasculitis
Dialysis and Transplant Association (ERA-EDTA) regis- account for 12% of new cases, of which lupus nephritis is the
try collects outcomes data for adult and pediatric ESRD most common. The underlying etiology of ESRD also varies
patients from national and regional registries of 35 Euro- by age of presentation. As expected, congenital anomalies
pean and Mediterranean countries.4 The Australia and of the kidney and urologic tract (CAKUT) and hereditary/
New Zealand Dialysis and Transplant Registry (ANZDATA) cystic disorders are the most common underlying causes of
collects data on all dialysis and transplant patients, includ- ESRD among young age groups, whereas primary glomeru-
ing children, in Australia and New Zealand.5 In 2004 the lar diseases and secondary glomerulonephritis are the lead-
Collaborative Brazilian Pediatric Renal Transplant Registry ing causes in adolescents (Fig. 37.1).1
37 • Kidney Transplantation in Children 635

Causes of ESRD in Children Annual Pediatric Kidney Transplant Counts


(USRDS 2011–2015) 1500
Total

Number transplanted
100
1250 Deceased donor
Other causes
80 1000 Living donor
Percentage (%)

C/H/C
750
60 Secondary GN
500
40 CAKUT
250
Primary GN
20 0
A 2000 2005 2010 2015
0
0–4 5–9 10–13 14–17 18–21 Median Wait Time (by age)
Age group 50
0–4
Fig. 37.1 Causes of end-stage renal disease (ESRD) in children stratified 40 5–9
by age as reported by the US Renal Data System (USRDS) 2017 report. 10–13

Months
Primary glomerulonephritis (GN) diseases include focal glomerulo- 30
sclerosis, membranous nephropathy, membranoproliferative GN, IgA 14–17
20 18–21
nephropathy, and other proliferative GN. Congenital anomalies of
the kidney and urologic tract (CAKUT) include congenital obstructive 10
uropathy, renal hypoplasia/dysplasia, and reflux nephropathy. Second-
ary GN includes lupus nephritis, hemolytic uremic syndrome (HUS), 0
Henoch-Schonlein (HSP), and vasculitis. Cystic, hereditary, and con- B 2000 2005 2010 2015
genital (C/H/C) disorders include polycystic kidney diseases (dominant
or recessive), Alport syndrome, congenital nephrotic syndrome, med- Median Wait Time (by race)
ullary cystic kidney diseases, including nephronophthisis, cystinosis, 50
primary oxalosis, sickle cell disease, and other congenital malformation White
syndromes. Other causes include 7% of total patients in the reporting 40 African
period with unknown etiology and 6.7% with missing data on etiology. American

Months
(US Renal Data System. 2017 USRDS Annual Data Report: Epidemiology of 30
Kidney Disease in the United States. Bethesda, MD: National Institutes of 20
Health, National Institute of Diabetes and Digestive Kidney Diseases; 2017.)
10
Overall, the male-to-female ratio for incident ESRD is 1.3 0
(56% of new cases are male), but is highest in the young- C 2000 2005 2010 2015
est patients, who predominantly suffer from congenital
Median Wait Time (by donor type)
disorders (roughly 3:1 male–female ratio), some of which 50
only occur in males (i.e., obstruction due to posterior ure- Deceased
thral valves). In contrast, secondary glomerulonephritis, 40 Living
especially lupus nephritis, is more common in females (1:4
Months

30
male–female ratio). Overall, 66% of children with ESRD in
20
the US identify as white race.1
The industrialized nations (North America, Europe, 10
Japan, Australia, New Zealand) have similar distribution 0
of etiologies as USRDS data presented previously.6 In the D 2000 2005 2010 2015
developing world, single-center studies and survey reports
suggest CAKUT and hereditary/cystic disorders are more Year
common, whereas acquired disorders are less common Fig. 37.2 Pediatric kidney transplant statistics over time for the US
than in industrialized nations.7 as reported by the US Renal Data System (USRDS). Panel A shows the
number of kidney transplant performed each year in the US for pedi-
atric patients aged 0 to 21 years. Panels B–D show median wait times
from incident dialysis by age, race, and donor type. (US Renal Data Sys-
Access to Transplantation tem. 2017 USRDS Annual Data Report: Epidemiology of Kidney Disease in
the United States. Bethesda, MD: National Institutes of Health, National
In the US, roughly 800 children receive kidney transplants Institute of Diabetes and Digestive Kidney Diseases; 2017.)
each year, comprising less than 5% of the total number of
kidney transplants performed each year. Children under transplantation for all age groups (98.3 per 100 active wait-
the age of 18 years represent 1.5% of total patients listed list years, compared with 18.8 per 100 for adults).8 Data
for kidney transplant in the US (1509 children vs. 97,680 from the USRDS report that median waiting times from
adults listed as of the end of 2015). Of the pediatric patients initiation of dialysis until transplant for pediatric patients
on the waiting list in 2015, 57% were over the age of 11 (0–21 years) range from 10 to 17 months depending on age
years.8 Data from the SRTR indicate that the total num- (Fig. 37.2).1
ber of kidney transplants in children (<18 years) in the US Historically, living-related donor transplants were more
decreased from a peak of 899 in 2005 to 718 in 2015.8 common in children than deceased donor transplants. This
The transplant community has consistently supported was likely driven by parents’ understanding of the benefit
timely access of deceased donor kidneys to pediatric recipi- of living donation for their child. However, the rate of liv-
ents. As a result, children have the highest rates of kidney ing donor transplants in children has been declining since
636 Kidney Transplantation: Principles and Practice

2002 (see Fig. 37.2). For example, in 2015, only 33.7% allocation system (e.g., 65.4% in 2015) received a kidney
of pediatric recipients received living-donor kidney trans- from a donor with KPDI less than 20%. The number of HLA
plants, compared with 50.1% in 2004. SRTR data from mismatches was higher among deceased-donor recipients
transplants performed in 2015 showed that, for the first than among living-donor recipients; 83.5% of deceased-
time, children under the age of 6 years were more likely donor recipients and 24.1% of living-donor recipients had
to receive a deceased-donor transplant (53%) instead of a more than three HLA mismatches in 2013 to 2015.8
living-donor transplant (47%), when historically, this age
group had the highest rates of living-donor transplant.8
In October 2005, OPTN implemented a new allocation Timing of Transplantation
policy for kidney transplants from deceased donors under
the age of 35 years to increase access to transplantation Transplantation is initially considered when renal replace-
from young donors for pediatric (ages <18 years) recipients, ment therapy is imminent. Due to increased risk of graft loss
which is known as Share-35. This kidney allocation policy and mortality in infants and children under 2 years of age,
emphasized the importance of younger donors and shorter most pediatric centers perform transplants in children once
waiting times for children over human leukocyte anti- they achieve a weight above 10 to 15 kg. Reports from a
gen (HLA) matching to minimize dialysis time. Although few centers have described successful transplant outcomes
Share-35 shortened pediatric wait times across races9 and in children under 15 kg.14,15,16 Infants and young children
decreased average donor age, the policy also resulted in pedi- with ESRD frequently have delayed growth, so often a child
atric recipients receiving kidneys with high HLA mismatch- will be older than 2 years of age before achieving the thresh-
ing.10 The long-term consequences of these trends on overall old size and weight for their transplant center.
graft survival in children will need future study. Similar On average, 30% of pediatric kidney transplant recipi-
trends in reduced wait times and increased rate of deceased- ents in the US from 2013 to 2015 received transplantation
donor transplants with concomitant decrease in living- before dialysis (e.g., preemptive transplants). An additional
donor transplant in pediatric recipients have been observed 24% of pediatric kidney transplant recipients in the same
in Europe when pediatric prioritization was implemented.11 time period received dialysis treatment for less than 1 year
The absolute number and proportion of deceased-donor at the time of transplant.8
kidney transplants in pediatric recipients increased after There is conflicting evidence for a benefit of preemptive
institution of the Share-35 policy from 40% to 50% during transplant for patient and graft survival in pediatric trans-
1998 to 2005 to 61% to 65% during 2006 to 2009 and plant recipients.17–19 Time on dialysis before transplant
was accompanied by a decrease in the absolute number of continues to be a risk factor for decreased graft survival,20,19
living-donor transplants.10 It is unclear at this time if the although some recent analyses suggest short times on dial-
trend in fewer living-donor transplants is a direct conse- ysis (less than 2 years) in children may not have as big an
quence of the policy change or due to the increasing preva- effect as longer periods.18,21 Increasing time with ESRD
lence of comorbidities in parents (obesity and diabetes in during childhood is also associated with impaired growth
particular) that preclude them from donating, as the down- and development and can result in disruption of education.
ward trend in living-donor transplants predates the policy Accordingly, preemptive transplant may have quality of life
(see Fig. 37.2).12 benefits to children beyond graft survival.
In December 2014, a new Kidney Allocation System (KAS)
in the US was employed with the goal to improve utility and
longevity of allografts, increase transplant access to sensi- Patient and Graft Survival
tized patients, and improve racial/ethnic disparities.13 The
new system replaced the previous kidney donor classification Patient survival for pediatric kidney transplant is excellent,
of standard- versus expanded-criteria donors with a more with overall 5-year patient survival of 97.7% among pedi-
refined metric known as the Kidney Donor Profile Index, or atric recipients in 2006 to 2010 as reported by SRTR.8 Data
KDPI, to predict the quality and longevity of the deceased- from the USRDS consistently show lower mortality rates for
donor kidneys and to appropriately match predicted graft children receiving kidney transplants compared with chil-
survival with expected recipient survival. Pediatric recipients dren on dialysis.1 The adjusted relative risk for mortality
under this new allocation policy receive priority for the high- for children receiving renal replacement therapy decreases
est quality donor kidneys (with KDPI less than 35%), owing with increasing age. The highest mortality for both dialy-
to the fact that young recipients have the longest remaining sis and transplant was in the 0 to 4-year-old age group,
life-expectancy posttransplant (see Table 37.1). However, but transplant still provided extra survival benefit. In an
there have been some concerns that the KDPI metric does analysis of the USRDS database, children who had received
not accurately predict graft survival in pediatric recipients a kidney transplant had a lower mortality rate (13.1
compared with other pediatric-specific predictive models.13 deaths/1000 patient years) than children remaining on the
The actual effect of the new allocation system on chil- waitlist (17.6 deaths/1000 patient years).22 Unlike similar
dren is still under evaluation, and more studies are needed. studies in adults, there was no significant excess mortality
There was initially a noticeable decrease in transplantation within the first 6 months posttransplant.
rate in children <6 years of age. Similar to adult recipients, Historically, graft survival rates in children were inferior
transplant rates increased for the highly sensitized pediatric to adults. However, in the past 20 years, graft survival in
candidates (with cPRA greater than 98%) and significantly children of all ages now rivals the rates seen in adults. Graft
declined for moderately sensitized candidates with cPRA survival improved dramatically for children during the
of 80% to 97%. Most pediatric recipients under the new 1980s and 1990s, but little progress has been made since
37 • Kidney Transplantation in Children 637

Death-Censored Graft Failure


(living-donor recipients) Graft Failure by Age and Donor Type
60
100
6-month <11, DD
<11, LD
Percentage (%)
1-year

Percentage (%)
90
40 3-year
5-year 11–17, DD
80
10-year 11–17, LD
20
70

0 60
2000 2005 2010 2015 0 10 20 30 40 50 60
A D
Death-Censored Graft Failure
(deceased-donor recipients) Patient Survival by Age and Donor Type
60
100
6-month <11, DD
99 <11, LD
Percentage (%)

1-year

Percentage (%)
40 3-year
98 11–17, DD
5-year
10-year 11–17, LD
97
20
96

0 95
2000 2005 2010 2015 0 10 20 30 40 50 60
B Year E

Incidence of Acute Rejection by 1 Year


(by age and era)
15
2009–10
2011–12
Percentage (%)

10 2013–14

0
<6 6–10 11–17 All
C Age group
Fig. 37.3 Outcomes for pediatric kidney transplant recipients in the US as reported by the Scientific Registry of Transplant Recipients (SRTR) 2015
report. Death-censored graft survival for pediatric recipients of living donor (A) and deceased donor (B) kidney transplants. Graft failure (D) and patient
survival (E) among pediatric recipients from 2006–2010 stratified by age and donor type. Incidence of acute rejection by 1 year for pediatric recipients
over three eras, stratified by age group (in years) (C). DD, deceased donor; LD, living donor. (Hart A, Smith JM, Skeans MA, et al. OPTN/SRTR 2015 Annual
Data Report: Kidney. Am J Transplant 2017;17(Suppl 1):21–116.)

2000 (Fig. 37.3). The adolescent age group (11–17) has a (>36 h; odds ratio [OR] = 3.38 vs. <36 h) and recipient age
worse 5-year graft survival compared with children receiv- 2 to 5 years (OR = 2.02 vs. 6–12 years). Long-term graft
ing a transplant under the age of 11 years (see Fig. 37.3).8 survival was most affected by race (relative risk [RR] = 1.93
for African American vs. others), adolescent recipients (RR
PROGNOSTIC FACTORS INFLUENCING GRAFT = 1.50 for 13–20 years vs. 6–12 years), and FSGS as pri-
mary diagnosis (RR = 1.27 vs. others).23
SURVIVAL
The following factors have been found to be important Donor Source
determinants of short-term and long-term graft survival in Children in all age groups receiving kidney transplants
pediatric kidney transplants. Some of these factors are more from living donors have superior short-term and long-
predictive of short-term survival, whereas others influence term graft survival (see Fig. 37.3) compared with children
long-term graft survival. For example, an analysis of the receiving deceased-donor grafts.8 The improved outcomes
UNOS database from 1995 until 2002 demonstrated that for living-donor kidney transplants are thought to be due
the most significant risk factors for early graft loss (by 3 to reduced cold ischemia time, improved HLA-matching,
months posttransplant) in children who received deceased- and improved preoperative preparation of the recipi-
donor kidney transplants were prolonged ischemia time ent.24 Survival of living unrelated donor grafts has even
638 Kidney Transplantation: Principles and Practice

been found to be superior to that of grafts from younger transplant, receive fewer living-donor transplants, and have
deceased donors.25 In addition, living donation from older longer duration of dialysis before transplant, all of which
relatives (e.g., a grandparent) has been found to have supe- are associated factors contributing to worse long-term out-
rior long-term outcomes in pediatric recipients compared comes.41 After controlling for 19 variables, including age,
with deceased-donor transplant.26 The most poorly HLA- primary diagnosis, preemptive transplant, rejection experi-
matched grafts from the oldest living donor had survival ence, HLA-B mismatch, immunosuppression, and gender,
similar to or better than any deceased donor in an analysis black patients still had higher risk for graft failure (Hazard
of USRDS.27 Finally, for deceased-donor kidneys, outcomes Ratio [HR] = 1.6, 95% confidence interval [CI] 1.46–1.86).42
are superior for pediatric recipients of grafts from donation
after brain death than donation after circulatory death due HLA Matching
to increased risk of graft loss by 4 years in the latter.28 Degree of HLA mismatch is an important prognostic factor
for long-term graft survival for both deceased- and living-
Recipient Age donor transplants.43 Although the ideal situation would be
An analysis of OPTN data has shown the best long-term donation from an HLA-identical sibling, this has occurred
graft survival occurred in middle-aged adult recipients and in only 3.4% of pediatric living-donor transplants reported
children younger than 12 years, whereas the worst out- in NAPRTCS. Most live kidney donations come from haplo-
comes were seen in adolescent and young adult recipients.29 identical parents. An analysis of NAPRTCS in 2000 sug-
Several studies have confirmed that recipients aged 17 to 24 gested that a six-antigen matched deceased-donor kidney
years have the highest risk of graft loss,30–32 prompting fur- had equivalent graft survival and rejection rates as haplo-
ther research in how to best support this age group to opti- identical living-donor transplants.44 However, long-term
mize outcomes. graft survival (5 years) was actually 10% better for the
children who received six-antigen matched deceased-donor
Donor Age and Size kidneys.
Multiple reports in the literature indicate that donation There is some evidence that donation from a sibling with
from young adults improves graft survival.33,34 The “ideal” noninherited maternal antigens confers survival benefit for
donor age derived from these analyses has been determined the graft and may be due to bidirectional immune regula-
to be more than 6 years old and less than 45 to 50 years tion between donor and recipient immune cells.45 Grafts
old. Donor age appears to be more important in determin- from mothers in these studies had poorer outcomes. This
ing graft survival for deceased-donor transplants than for has particular relevance for pediatrics because mothers
living-donor transplants. represent the majority of parental donors.
Interestingly, children receiving grafts from deceased There is debate as to the effect of degree of HLA mismatch
donors younger than age 18 years have improved relative on acute rejection and graft survival. An analysis of 9029
glomerular filtration rate (GFR) over time compared with chil- pediatric recipients in the Collaborative Transplant Study
dren receiving kidneys from deceased or living adult donors.35 in Europe found a hierarchical relationship for an effect of
Senescence of somatic kidney cells has been postulated to increasing mismatches at the A-, B-, and DR-loci on graft
be the underlying cellular mechanism responsible for this survival.34 Similarly, there are inconsistent findings on the
effect.36 In other words, renal allografts from young donors are role of HLA-DR matching and graft survival.46 In 2008
able to adapt to the increasing metabolic demand of a growing Gritsch, et al. reported that children in the US receiving zero
recipient, whereas kidneys from older donors have decreased HLA-DR mismatched deceased-donor kidneys had compa-
capacity for cell survival and regeneration and undergo accel- rable 5-year graft survival to children who had received
erated aging under the stress of transplantation. 1- or 2-DR mismatched kidneys.47 A recent analysis of Euro-
Transplants from very young donors (younger than age 5 pean pediatric kidney transplants48 found that whereas two
years) to pediatric recipients have historically been avoided HLA-DR mismatches associated with lower graft survival
due to reports of increased risk of graft loss. In a recent analy- in children receiving transplants from 1988 to 1997, this
sis of the NAPRTCS database, primary graft nonfunction was effect was not seen in a more recent era (1998–2007). They
found to be more frequent in kidney transplants from very did, however, describe a disturbing association of HLA-DR
young donors (3.7% compared with 0.3% in transplants from double-mismatch with increased risk of developing non-
donors aged 6 to 35 years or “ideal donors”). However, lon- Hodgkin lymphoma.48 This interesting finding needs to be
ger term, the 3-year graft survival and estimated glomerular validated in additional cohorts of pediatric kidney trans-
filtration rate (eGFR) of functioning grafts from very young plant recipients.
donors was equivalent to ideal donors.37 In a more recent Sensitization occurs with increasing HLA-mismatch49
study, graft survival rate was almost 90% for recipients of and specifically the degree of HLA-DR mismatch.50 As stated
infant donor kidneys implanted either en bloc or singly.38 earlier, children have longer expected remaining lifetimes
after transplantation and increased expectancy for multiple
Recipient Race transplants. Therefore development of HLA-sensitization
African American race has been demonstrated in multiple from mismatched primary transplants has been demon-
studies to associate with worse long-term graft survival, strated to be a barrier to subsequent transplant.50,49
including increased risk for delayed graft function, acute
rejection, and graft loss.39,40 It is unclear whether the rela- Presensitization
tive role of genetics (e.g., higher rates of FSGS) versus racial NAPRTCS reported that >5 transfusions significantly
disparities explains these findings. For example, African increased the frequency of delayed graft function (defined
American children are more likely to be older at the time of as dialysis in the first transplant week) and graft failure
37 • Kidney Transplantation in Children 639

(HR = 1.22, p = 0.016 for living-donor transplants and HR PRIMARY GLOMERULONEPHRITIS


= 1.25, p < 0.001 for deceased-donor transplants). This
effect was presumed to be due to increased sensitization. Focal Segmental Glomerulosclerosis
The use of blood transfusion has decreased with increasing FSGS recurs in approximately 15% to 60% of children
use of erythropoietin to treat anemia during ESRD, and the with nephrotic syndrome undergoing renal transplant
largest risk for sensitization in pediatric transplant candi- and is the most common cause of graft loss due to recur-
dates is prior solid-organ transplantation. rence.53–55 Although several small single-center studies in
the 1990s suggested clinical features of young age of diag-
Delayed Graft Function and Thrombosis nosis, rapid progression to ESRD, and mesangial prolifera-
Delayed graft function (DGF) is uncommon (approximately tion on native biopsy to be associated with posttransplant
6%) in pediatric kidney transplant recipients, as reported recurrence, many of these predictive factors have failed to
in the 2015 SRTR report. A single-center retrospective be durably replicated.53 A recent analysis of UNOS data
study of 126 pediatric kidney transplant recipients from showed an overall recurrence rate of 15% in children
2004 to 2011 showed similar rate of 9% DGF and identi- receiving kidney transplant between 1988 and 2008.56
fied vascular cause of brain death and donor age >15 years FSGS recurrence occurred more frequently in Caucasian
as possible risk factors for DGF. Overall, technical causes of and younger recipients. Although receipt of a living-
graft loss (vascular thrombosis, primary nonfunction, and donor kidney was previously described as a risk factor
others) contributed to 13% of all graft failures reported in for FSGS recurrence, in this analysis, donor type did not
NAPRTCS since 1987, suggesting that these events occur remain significant in multivariate analysis.56 This may be
in about 3% of pediatric transplants. Several studies of pedi- explained by the confounding effect of younger recipient
atric transplant registries in the UK, Ireland, and the Neth- age because rates of living-donor transplantation increase
erlands reported similar rates of thrombosis. Risk factors for with decreasing recipient age. The significance of young
graft thrombosis in children include recipient age <6 years, recipient age in this study may also indirectly support ear-
donor age <6 years, cold ischemia time >24 hours, and his- lier associations between younger age at FSGS diagnosis
tory of peritoneal dialysis.51 and/or rapid progression to ESRD and FSGS recurrence.
Another clinical feature recently associated with FSGS
recurrence is the response to steroid therapy during native
Contraindications to FSGS disease. In an analysis of 150 pediatric patients
Transplantation transplanted in Europe for ESRD due to steroid-resistant
nephrotic syndrome, it was found that the patients with
Children with ESRD have fewer comorbid conditions that initial steroid sensitivity who developed steroid resistance
might mitigate against major surgery or use of immuno- during treatment of their native kidney disease had the
suppressive medications than adults, so the risks of kidney highest rate of recurrence (92.9%) posttransplant com-
transplantation are often greatly outweighed by the ben- pared with children who had steroid-resistance from the
efits. Therefore most children with ESRD are eventually outset (30%).57 These findings suggest that the develop-
referred for transplant. ment of secondary steroid resistance during treatment
There are few absolute contraindications to kidney trans- of idiopathic nephrotic syndrome may predict posttrans-
plant in children. Situations that might not be appropriate plant recurrence.
for referral or listing include active or untreated malig- Patients with FSGS recurrence have inferior graft survival
nancy, active or untreated infection, and multiple or pro- compared with other ESRD etiologies.53,54 Furthermore,
gressive medical conditions with overall poor prognosis for the effect of FSGS recurrence on graft loss is more significant
recovery (e.g., severe brain injury or multiorgan failure). in children compared with adults.58 Higher rates of acute
Transplant is considered after a reasonable disease-free tubular necrosis (ATN) in both living-donor and deceased-
period for children with prior malignancies. donor transplants have been reported in children with FSGS
Mild, isolated mental retardation is not a contraindica- compared with children with other causes of ESRD.59 It has
tion to transplant, per se, as improvement in neurocogni- been suggested that the increased rate of ATN, possibly due
tive development has been seen after transplant.52 Children to early FSGS recurrence, plays a role in the decreased graft
with devastating neurologic dysfunction may not benefit survival for children with FSGS.60 This observation influ-
appreciably from transplant, but the potential for reha- enced some centers to offer living-donor transplant (which
bilitation, self-care, and parental preferences should be has lower rates of ATN in general) with pretransplant plas-
considered. Finally, issues with medical compliance or mapheresis in an attempt to prevent rapid FSGS recurrence
unstable family situations can delay consideration for kid- and improve graft survival; however, the use of plasma-
ney transplant. pheresis pretransplant has not proven consistently benefi-
cial in small trials.61
Recurrence of FSGS often presents early after kidney
Recurrence of Original Disease transplantation in children, with a reported median time
to recurrence of 6 to 14 days, although heavy proteinuria
Recurrent disease is a significant cause of graft loss in chil- often can be detected within hours after transplant. It is
dren, accounting for 6.9% of graft losses in the NAPRTCS usually characterized by nephrotic-range proteinuria (pro-
cohort.2 Recurrence in the transplanted kidney can occur tein/creatinine ratio >2 mg/mg) and hypoalbuminemia,
with primary glomerulonephritis, secondary glomerulone- but can present as complete nephrotic syndrome, including
phritis, and metabolic diseases. anasarca and hypercholesterolemia. Whereas recurrent
640 Kidney Transplantation: Principles and Practice

disease typically presents within the first 2 years after trans- others. In general, it is recommended that children at risk
plant, the presentation of nephrotic syndrome after 2 years should have daily monitoring of protein/creatinine ratios in
is generally considered to be secondary to calcineurin inhib- the early posttransplant period to allow rapid detection of
itor (CNI) toxicity, chronic rejection, or de novo disease. FSGS recurrence and early initiation of treatment.
Biopsies early after recurrence often demonstrate normal Some centers perform plasmapheresis either before a
histology on light microscopy with effacement of podocyte planned living donor transplant or in the perioperative
foot processes on electron microscopy. Later biopsies have period of a deceased-donor transplant. It is unclear if this
characteristic segmental lesions of FSGS with endocapillary practice offers any benefit over early detection and treat-
proliferation and foam cell accumulation and can progress ment of established recurrence.71,61
to glomerular sclerosis and interstitial fibrosis. Unfortu- High-dose cyclosporine with or without adjunctive plas-
nately, histology classification has not been shown to be mapheresis and/or high-dose steroids has shown efficacy in
predictive of treatment response in recurrent FSGS.62 achieving complete or partial remission of posttransplant
The pathophysiology of primary idiopathic FSGS in FSGS recurrence in children.72,73,74 The antiproteinuric
native kidneys and recurrent FSGS after transplant remains effect of CNIs has been postulated to be due to T cell sup-
unclear. It is likely a multifactorial process involving pression and inhibition of cytokine secretion thought to be
cytokines secreted by T cells, a humoral factor that alters injurious to podocytes, as well as to a direct effect on the
podocyte cytoskeletal structure, and a balance between stabilization of the podocyte cytoskeleton.67 The alkylating
circulating permeability factors and inhibitors of such agent cyclophosphamide has also been reported to be effica-
factors.63 cious in some children with recurrent FSGS, usually in com-
Genetic forms of FSGS have low rates of recurrence, esti- bination with plasmapheresis.75 Reduction in proteinuria
mated at around 3% for patients with homozygous64 and has also been reported with the use of angiotensin block-
compound heterozygous podocin (NPHS2) mutations.65 ade76 either alone or in conjunction with plasmapheresis.
Recurrence of nephrotic syndrome in children with NPHS2 Finally, there have been anecdotal cases reporting pro-
mutations does not appear to be due to antipodocin anti- longed remission of proteinuria in children with recurrent
bodies.66 Further study is needed to clarify the pathogenesis FSGS after plasmapheresis and B cell depletion with ritux-
of recurrence in this population. imab.77–79 This was first reported for a child who inciden-
A circulating factor has been proposed to cause increased tally achieved remission of FSGS recurrence after rituximab
albumin permeability of the slit diaphragm during FSGS therapy to treat posttransplant lymphoproliferative disease
recurrence, but the identity, source, and pathologic effects (PTLD).80 More recently, there have been small case reports
of such a factor are yet to be fully elucidated.67 Bioassays demonstrating efficacy of another humanized anti-CD20
of albumin permeability have had conflicting results in pre- antibody, ofatumumab, in inducing remission of native kid-
dicting FSGS recurrence and have not proven to be specific ney FSGS or FSGS recurrence in patients resistant to ritux-
or highly predictive of response to therapy or long-term imab therapy.81 The true value of this approach remains to
renal outcome in patients with nephrotic syndrome. Fur- be established.
thermore, demonstration of a neutralizing effect of normal
serum or urine from nephrotic patients on albumin perme- Congenital Nephrotic Syndrome
ability suggests that loss or deficiency of a natural inhibi- Congenital nephrotic syndrome (CNS), by definition, occurs
tor may play a role.63 Serum soluble urokinase receptor within the first 3 months of life and is most commonly due
(suPAR) has been proposed as a circulating factor capable to mutations in the NPHS1 gene, encoding nephrin, a
of causing FSGS.68 Two-thirds of the patients with FSGS major structural component of the slit diaphragm. Infants
in this study had elevated serum suPAR concentrations with CNS are frequently born prematurely, have enlarged
compared with healthy controls and patients with other placentas, and present with nephrotic-range proteinuria,
glomerular diseases. Furthermore, patients who had FSGS anasarca, and hypoalbuminemia. Secondary causes (i.e.,
recurrence after transplant had the highest serum concen- neonatal cytomegalovirus [CMV], congenital rubella,
trations of suPAR, providing hope for a clinically predictive human immunodeficiency virus [HIV], hepatitis B, toxo-
test. However, a small prospective study found no correla- plasmosis, syphilis, and infantile lupus) should be excluded.
tion with suPAR levels and remission of proteinuria in adult The genetics of CNS are proving more complex in recent
patients treated for recurrent FSGS.69 Further research is years with the identification of additional genetic mutations
needed in understanding the pathogenesis of FSGS recur- (including genes previously only associated with FSGS).82
rence and to identify predictive clinical tests. Recurrence of nephrotic syndrome has been reported in
In light of the limited understanding of the underlying 25% of children with CNS due to homozygous Fin-major
pathophysiology, the treatment of recurrent FSGS is not mutations in nephrin (NPHS1) with mean time to recur-
well established. The most commonly reported therapies for rence of 12 months posttransplant (range of 5 days to 2
recurrent FSGS are plasmapheresis or protein A immuno- years). Antinephrin antibodies have been implicated in a
adsorption therapy.70 A review of the literature found that majority of these children.77,83
49 of 70 children (70%) receiving plasmapheresis for FSGS Vascular thrombosis and death from infection (with a
recurrence achieved partial or full remission.63 Early detec- functional graft) have been described more often in children
tion of recurrence and initiation of plasmapheresis appears with CNS after transplant compared with children with
to provide the best results. However, these studies likely other primary diseases.84 Hypercoagulability due to urinary
overrepresent the benefit of plasmapheresis given small losses of antithrombin III (ATIII) along with younger age at
sample sizes and the use of only historical groups for com- the time of transplant likely contribute to the increased risk
parison in a few of the studies or no comparison group in of these complications.
37 • Kidney Transplantation in Children 641

Diffuse mesangial sclerosis (DMS) can also present as basement membrane (anti-GBM) has been reported to
nephrotic syndrome in early infancy and is associated with occur in approximately 3% to 5% of AS males within the
mutations of the Wilm tumor suppressor gene 1 (WT1). first year after transplant.88,92 Late occurrence has also
Denys-Drash syndrome consists of progressive glomerulop- been described.93
athy (DMS) and male pseudohermaphroditism (although Antibodies are generated against the type IV collagen α5
genotypic females have also been described) and is due to or α3 chains.94,95 Patients with mutations resulting in com-
heterozygous mutations within exon 8 or 9 of WT1. Chil- plete absence or severe truncation of these proteins appear
dren with Denys-Drash syndrome are at increased risk of to be at highest risk of developing anti-GBM antibodies.
developing Wilm tumor; therefore bilateral nephrectomy is It should be noted that the anti-GBM antibodies in these
often performed once they develop ESRD. Frasier syndrome patients are specific for different epitopes of the noncollage-
presents with normal female genitalia with streak gonads, nous domains of the α3α4α5(IV) collagen network than the
XY karyotype, and progressive glomerulopathy (DMS on Goodpasture allo-epitope, and therefore may not be detect-
histology) and is due to splice mutations within exon 9 of able in the serum using the clinically available enzyme-
WT1. Children with Frasier syndrome are at risk of develop- linked immunosorbent assay (ELISA) used to diagnose
ing gonadoblastoma and therefore should undergo oopho- spontaneous anti-GBM nephritis in Goodpasture disease.95
rectomy. There have also been reports of isolated DMS in Early reports described rapidly progressive anti-GBM
children with WT1 mutations without other syndromic fea- disease with nearly 90% to 100% graft loss. With better
tures who may not be at risk for malignancy.85 recognition of this entity, reports of subclinical anti-GBM
Other than one report of membranoproliferative glo- (i.e., demonstration of linear IgG deposits along the GBM
merulonephritis (MPGN) in a child with Denys-Drash,86 in transplant biopsies without graft dysfunction) suggest
no other studies report recurrence of nephrotic syndrome a broader spectrum of clinical disease.92 Despite the poor
in children with WT1 mutations undergoing kidney trans- graft outcome described during anti-GBM nephritis in AS,
plant.87 Overall, children with Denys-Drash have compara- the severe form does not occur frequently enough to affect
ble patient and graft survival as children with other causes overall graft survival statistics for AS patients.88 Chronic
of ESRD.2 rejection is the most common cause of graft loss in this
population. Patients who have lost a prior graft to anti-
Alport Syndrome GBM disease are at higher risk of recurrence after a subse-
Alport syndrome (AS) is a clinically and genetically hetero- quent transplant, although some have reported successful
geneous nephropathy characterized by glomerular base- retransplantation.96
ment membrane (GBM) defects due to alterations in the
type IV collagen matrix. AS presents with persistent micro- MPGN/C3 Glomerulopathy
scopic hematuria and proteinuria that can progress to renal MPGN describes a pattern of glomerular injury with com-
failure and is often associated with sensorineural hearing mon histologic features of glomerular capillary wall thick-
loss and ocular abnormalities. It is the prototype of inher- ening (membrano-) and hypercellularity in the glomerular
ited nephritis and accounts for 2% of children with ESRD in tufts (-proliferative). MPGN was historically classified into
the US. The most common and severe form is inherited in three morphologic types based on electron microscopy find-
an X-linked recessive pattern and is associated with early ings: type I with presence of immune deposits in the suben-
progression to ESRD (50% of boys by age 25 years). Auto- dothelial and mesangial areas, type II with electron-dense
somal recessive and autosomal dominant forms have also deposits within the basement membrane, and type III with
been described. complex GBM formation with subendothelial and subepi-
The genetics of X-linked AS were defined more than 20 thelial electron-dense deposits that are bridged by intra-
years ago as a mutation in the gene encoding the α5 chain membranous deposits. A unifying characteristic of all types
of type IV collagen (COL4A5). Patients with mutations of MPGN is hypocomplementemia (low C3). This traditional
resulting in a truncated protein (i.e., large rearrangement, system resulted in overlap among MPGN type I and type III
premature stop, or frame-shift mutations) tend to progress while considering MPGN type II, known as dense deposit
to ESRD earlier (50% by age 19 years).88,89 Female car- disease (DDD), as a separate entity because it has unique
riers of COL4A5 mutation are considered to have a less pathogenic and clinical features.
severe course, but some do develop ESRD in late adulthood. Recent advances in our understanding of MPGN patho-
Development of hearing loss and progression of proteinuria genesis has led to a new classification system for the diagno-
appear predictive of a more severe course in female carri- sis and management of these disorders. In the new system,
ers. Evaluation of potential living donors, especially female MPGN is classified as either mediated by immune complexes
relatives, should include evaluation for hematuria and pro- (immune complex mediated MPGN) or by complement dys-
teinuria, as the heterozygous carrier state carries a risk of regulation and activation of the alternative complement
developing ESRD after donation.90,91 pathway, now termed C3 glomerulopathy. This differen-
Autosomal recessive forms involving mutations in the tiation is made based on immunofluorescence microscopy.
α3 (COL4A3) and α5 (COL4A4) chains have also been Immune complex mediated MPGN is defined by the pres-
described. Rare autosomal dominant mutations in COL4A3 ence of immunoglobulin staining on immunofluorescence
and COL4A4 have also been reported with less severe renal microscopy and is most commonly associated with under-
disease, fewer cases of hearing loss, and absence of reported lying autoimmune disease or infections. Therefore recur-
ocular changes.89 rence and management are dependent on the underlying
Although Alport syndrome itself does not recur, devel- systemic disease resulting in renal pathology. In contrast,
opment of de novo antibodies directed to the glomerular complement-mediated MPGN (i.e., C3 glomerulopathy)
642 Kidney Transplantation: Principles and Practice

results from dysregulation and persistent activation of the There is minimal data about transplant outcomes for
alternative complement pathway with deposition of com- patients with C3GN. Zand et al. evaluated 21 adult and pedi-
plement products in the glomeruli and the mesangium atric renal transplant recipients with a history of C3GN as a
with predominantly bright C3 and minimal immunoglobu- cause of the ESRD. C3GN recurred in 66.7% of the patients
lin staining on immunofluorescence microscopy. Genetic with graft failure in 50% of recurrent C3GN at median time
defects in the proteins involved in the alternative comple- 28 months. The median time to graft failure posttransplant
ment cascade and/or autoantibodies have been identified in was longer in patients with C3GN compared to DDD (6–7
patients with C3 glomerulopathy, which is further classi- years vs. 2.5 years, respectively).102
fied into DDD, C3 glomerulonephritis (C3GN), and CFHR5 Recurrent MPGN presents with hematuria, progres-
nephropathy. DDD is distinguished from C3GN on electron sive proteinuria, and deteriorating graft function. Low
microscopy by the presence of large, sausage-shaped, osmo- or low-normal C3 levels have been reported. There are
philic dense deposits within the GBMs. In contrast, in C3GN no randomized trials to determine efficacy of treatment of
the deposits tend to be mesangial and subendothelial. posttransplant recurrence of C3GN and DDD. Nonspecific
The major defect in DDD and C3GN is excessive activa- treatments, including the use of angiotensin blockade, ste-
tion of the alternative complement pathway. The alter- roids, anticoagulation, or antiplatelet therapy, have been
native complement pathway is normally autoactivated reported in the literature with variable success. In patients
by spontaneous cleavage of the C3 to C3b which leads to with genetic deficiency in factor H, plasma infusion can be
the formation of the C3 convertase by the binding factor used to correct the deficiency. Plasma exchange has been
B and properdin. Normally the activity of this pathway is reported in small case series for removal of circulating auto-
tightly regulated by the activity of the C3 convertase, which antibodies like C3 nephritic factor or antifactor H a
­ ntibodies
is inhibited by factor H. In patients with DDD and C3GN, and to replace defective proteins in the case of activating
increased activity of C3 convertase is most commonly mutations in C3.102–104 Rituximab has been reported in
caused by (1) stabilization and reduced degradation of C3 cases involving autoantibodies.104 More recently, the use
convertase enzyme by autoantibodies (called C3 nephritic of an anti-C5 antibody (eculizumab) has been proposed to
factor; C3NeF) or (2) deficiency or autoantibody directed block the downstream effects of uncontrolled C3 convertase
against the natural inhibitor of C3bBb, factor H. Low lev- activity,105 but the clinical utility, long-term implications,
els or defective factor B, factor I, and C3 have also been and identification of patients who would benefit most from
described in DDD and C3GN. this therapy still need to be defined.106
C3 glomerulopathies have a progressive course with
development of ESRD in up to 50% of patients and high SECONDARY GLOMERULONEPHRITIS
recurrence after transplantation.
Historic reports of outcomes for patients with MPGN IgA and Henoch-Schonlein Purpura
type I and III are challenging to apply to patients diagnosed IgA nephropathy accounts for 3% of incident cases of ESRD
under the new classification system. in children in the US.1 Histologic recurrence in adult trans-
An analysis of UNOS data showed MPGN type I has plant recipients with IgA nephropathy has been reported to
a significant negative effect on graft survival compared be 30% to 35%. Transplant patients with abnormal urinal-
with other forms of glomerulonephritis.97 Disease recur- ysis (hematuria or proteinuria) are more likely to show his-
rence was the most common cause of graft loss (14.5%) for tologic evidence of recurrence.107 The role of recurrence in
patients with MPGN type I in this study. Graft loss has been graft loss is variable in these reports. In some single-center
reported in about 50% of patients with recurrent MPGN studies, as many as 40% to 50% of patients with recurrence
type I. Low complement levels (C3) at the time of transplant have been reported to have graft failure; however, a recent
and younger age at time of transplant have been associated analysis of patients in East Asia found that chronic rejec-
with increased risk of recurrence. Another report of serial tion had a larger effect on long-term graft survival rate in
protocol biopsies identified that some patients have asymp- patients with IgA nephropathy.108,109
tomatic recurrence of MPGN type I.98,99 Henoch-Schonlein purpura (HSP) nephritis is a rare
An analysis of the NAPRTCS database showed signifi- cause of ESRD in children, accounting for only 0.4% of
cantly worse 5-year graft survival (50.0%) for children incident cases in the US.1 Histologic recurrence of HSP110
with MPGN type II (DDD) compared with the database as a has been reported in as high as 70% of patients within 2
whole (74.3%).100 The most common cause of graft failure years after transplant, with clinically evident recurrence
in children with DDD was recurrent disease (14.7% of graft (hematuria, moderate proteinuria, and hypertension) in
losses). Of the children with DDD who had posttransplant 15% to 35%.111 Long-term graft survival, however, does
biopsies (n = 18) in the registry, 67% had recurrent DDD. not appear to be greatly affected by recurrence.108,112 An
No correlation was found between pretransplant presenta- analysis of adolescents and young adults in the UNOS data-
tion or C3 levels with the risk of recurrence or graft loss.100 base revealed graft loss due to recurrent disease in 13.6% of
In a single-center retrospective study, DDD was reported patients with HSP.113
to recur in 60% of children, whereas the rate of graft loss
was similar to other children, transplanted at that cen- Hemolytic-Uremic Syndrome
ter.101 Finally, an analysis of the UNOS database, including Hemolytic-uremic syndrome (HUS) accounts for approxi-
children and adults, reported graft loss due to recurrence mately 1.5% of children with ESRD in the US.1 HUS is char-
in 30% of patients with DDD and significantly worse acterized by the clinical triad of microangiopathic hemolytic
graft survival compared with patients with other forms of anemia, thrombocytopenia, and renal failure, and it can
glomerulonephritis.97 be due to secondary causes (infectious, drug-induced,
37 • Kidney Transplantation in Children 643

auto-antibodies) or primary genetic defects in complement patients with aHUS, but up to 40% of these patients also
regulatory components that lead to persistent activation of carry a mutation in CFH, CFI, MCP, or C3.121
the alternative pathway of the complement cascade. Atypical HUS is also increasingly recognized as a com-
The most common cause of HUS in children (account- plex, polygenic disease. First, incomplete penetrance is
ing for approximately 90% of cases) is associated with coli- common in most of these mutations, suggesting that “mul-
tis due to shiga-toxin producing bacteria (E. coli, Shigella tiple hits” contribute to a predisposition to aHUS. Second,
dysenteriae, others). Epidemiologic studies estimate that various polymorphisms in genes encoding CFH, MCP, CFH-
5% to 10% of children with shiga-toxin producing E. coli related protein (CFHR1), and C4b-binding protein (C4b-BP)
(STEC) infections develop HUS, with children under the have been associated with aHUS. Furthermore, bigenic
age of 5 years carrying the highest risk. Of the children abnormalities have been described in about 10% of patients;
that develop HUS, about two-thirds develop oliguric renal therefore mutational analysis of all complement compo-
failure.114 ESRD occurred in 10% to 15% of children with nents is recommended during the workup of patients sus-
HUS in a meta-analysis of long-term prognosis (>1 year) pected to have aHUS. Finally, cases of de novo thrombotic
after diarrhea-associated HUS.115 The presence of anuria or microangiopathy (TMA) after kidney transplant in patients
prolonged oliguria has been associated with higher risk for with non-HUS causes of ESRD have also been found retro-
long-term sequelae, including CKD, proteinuria, and hyper- spectively to have CFH or CFI mutations.120
tension in as many as 30% of HUS survivors.116 The risk of Historically, studies on the outcomes of children with
disease recurrence after transplant in this particular group HUS have used simple delineation of cases based on the
appears to be very low (<1%), and graft survival is similar to presence or absence of prodromal diarrhea. This delin-
children with nonglomerular primary disease.117,118 eation is not always clinically straightforward because
Infections with bacteria other than STEC that produce shiga-toxin-related cases may present without diarrhea,
shiga- or shiga-like toxins, including Shigella dysenteriae and 20% to 30% of cases of aHUS due to genetic comple-
and Citrobacter sp, have also been implicated in the etiology ment disorders are preceded by a diarrheal illness (includ-
of HUS in children. Furthermore, urinary tract infections ing STEC infection). Furthermore, as mentioned previously,
with STEC have also been found in children with HUS pre- there are nondiarrheal bacterial infections associated with
senting without diarrhea. HUS (STEC urinary tract infection [UTI] and Pneumococcus).
Finally, invasive Streptococcus pneumoniae infections have A recent guideline has been published in an effort to stan-
also been linked with a rare, but severe, form of HUS that dardize diagnostic workup and treatment for children with
has higher rates of progression to ESRD than the classi- atypical HUS.122 Children without diarrheal prodrome or
cal diarrhea-associated disease. Cases typically have large pneumococcal infection or those with recent diarrhea and
bacterial burden with empyema and bacteremia, but cases certain clinical characteristics associated with increased
with meningitis or pericarditis have also been described.119 risk of genetic predisposition (Box 37.1) should undergo a
Bacterial neuraminidase exposes a crypt-antigen known as full diagnostic evaluation for the cause of HUS, including
the Thomsen-Freidenrich (or T-) antigen on erythrocytes, investigation for shiga-toxin-producing bacteria as well as
platelets, and endothelium, and this is thought to play a role mutational analysis.
in endothelial activation and subsequent microvascular
thrombus formation. T-antigen exposure can be confirmed BOX 37.1 Suggested Evaluation of
on patient erythrocytes using the lectin Arachis hypogea. Children with Hemolytic Uremic Syndrome
Recurrence of pneumococcal HUS after transplantation has
not been reported for these patients. Risk factors that should prompt a diagnostic workup for atypical
HUS that cannot be associated with infection or other sec- HUS, even if diarrhea is present, include:
ondary causes (often referred to as atypical HUS) accounts □ Age of onset under 6 months old

for 5% to 10% of all cases in children and carries a higher □ Insidious onset
□ Relapse of HUS or a suspected previous case of HUS
risk to progress to ESRD without treatment. Genetic or
□ Previous unexplained anemia
acquired disorders of complement regulation are identified
□ Nonsynchronous family history of HUS
in about 60% to 70% of these cases. They also have higher □ Presentation with severe hypertension
risk of recurrence and graft loss posttransplant. Therefore □ Presentation of HUS posttransplant (for any organ)
identifying these cases is paramount for (1) preemptive Diagnostic workup includes:
treatment to prevent progression to ESRD and (2) planning □ Serum/plasma C3 level (although normal values do not exclude
for successful transplantation. inherited disorders of complement regulation)
In recent years, a clear link has been established between □ Plasma/serum concentration of factor H and factor I

disordered regulation of the alternative pathway of the □ Antifactor H antibody titers

complement system and atypical HUS (aHUS). Mutations □ Membrane cofactor protein (MCP: CD46) surface expression on

have been described in three important regulatory pro- mononuclear leukocytes by flow cytometry
□ Gene mutation analysis for factor H, factor I, MCP, factor B and
teins of the alternative pathway: complement factor H
(CFH; 20%–30% of aHUS registry cases), complement fac- C3
□ Plasma vWF protease activity (ADAMTS13)
tor I (CFI; 2%–12%), and membrane cofactor protein (MCP; □ Homocysteine and methylmalonic acid levels (plasma and
10%–15%).120 Gain-of-function mutations in genes encod- urine) to evaluate for defects in cobalamin metabolism
ing complement factor B (CFB; 1%–2%) and complement C3
(10%) and loss-of-function mutations in thrombomodulin Adapted from Ariceta G, Besbas N, Johnson S, et al. Guideline for the in-
(THBD gene) have also been associated with aHUS. Finally, vestigation and initial therapy of diarrhea-negative hemolytic uremic
autoantibodies to factor H are detectable in 5% to 10% of syndrome. Pediatr Nephrol 2009;24:687–96.
644 Kidney Transplantation: Principles and Practice

TABLE 37.2 Risk of ESRD and Transplant Recurrence for of therapy or during infections (especially CMV infection).
Hereditary Forms of Atypical HUS Furthermore, several patients with CFH or CFI mutations
have been reported to have graft loss after HUS recurrence
Frequency of Risk of despite PE, although these cases did not receive preemp-
Gene Mutations Risk of ESRD Recurrence
tive PE. Finally, patients with MCP mutation do not appear
CFH 24%–27% 52%–65% 80%–90% to benefit from plasma therapy. Liver transplant alone or in
MCP 5%–9% 6%–63% 15%–20% combination with kidney transplant has been reported for
C3 2%–8% 43%–67% 40%–50%
CFI 4%–8% 17%–83% 70%–80% children with factor H mutation with the rationale that the
CFB <1%–4% 70% Unknown transplanted liver would provide wild-type factor H.129–131
THBD 5% 23% Unknown Although early attempts resulted in acute thrombotic events
CFHR5 <1%–4% Unknown Unknown and high mortality rates, several centers in Europe have
DGKE <1% Unknown Unknown
CFHR 3%–10% Unknown Unknown
reported improved outcomes with ancillary plasma exchange
CFH antibody 3%–6% Unknown Unknown and anticoagulation therapies. However, the risk of morbid-
ity and mortality has limited the use of this approach. Suc-
C3, complement 3; CFB, complement factor B; CFH, complement factor H; CFHR, cessful transplantation in patients with complement factor H
complement factor H-related protein; CFI, complement factor I; DGKE, autoantibodies have been reported with the use of preemp-
diacylglycerol kinase epsilon; MCP, membrane cofactor protein; THBD,
­thrombomodulin. tive plasma exchange with or without rituximab.132,133
Adapted from Nester, CM, Barbour T, de Cordoba SR, Dragon-Durey MA, Although CNIs have been associated with de novo HUS
Fremeaux-Bacchi V, Goodship TH, Kavanagh D, Noris M, Pickering M, after solid-organ transplant, avoidance of CNIs has not been
Sanchez-Corral P, Skerka C, Zipfel P, Smith RJ. Atypical aHUS: State of the shown to affect the risk of recurrence in genotyped patients
Art. Mol Immunol. 2015;67:31–42.
with aHUS.124
Finally, the use of the anti-C5 monoclonal antibody,
Recurrence after transplant can present as graft throm- eculizumab, to prevent membrane attack complex forma-
bosis or graft failure with hematologic signs of HUS (micro- tion holds promise for both the prevention and treatment of
angiopathic hemolytic anemia and thrombocytopenia). recurrence in children with aHUS undergoing renal trans-
The risk of recurrence after transplant varies depending plantation.134–136 Moreover, the use of eculizumab may
on the genetic mutation identified (Table 37.2), and before prevent progression to ESRD and obviate the need for renal
the development of anticomplement therapy was approxi- transplantation.137 Optimal dosage and interval between
mately 60% overall. The Global aHUS Registry (NIH clinical eculizumab infusions remain to be fully defined, and the
trials identifier NCT01522183), of which approximately high cost of this agent may limit access. Although studies
20% have received kidney transplant, may provide addi- of long-term outcomes are not available, a requirement for
tional information on outcomes with current therapeutic life-long therapy is anticipated.
approaches.123 High rates of recurrence (80%) and graft
loss (80%–100%) have been reported for patients with fac- Membranous Nephropathy
tor H or factor I mutations.124,125 In theory, patients with Membranous nephropathy is rare in children at only 0.5%
isolated mutations in the membrane-bound MCP protein of incident ESRD in the US,1 so risk of recurrence in children
should not develop recurrent HUS because the allograft after transplant is not clear.
would have wild-type, functional MCP protein. An analy-
sis of the International Registry of Recurrent and Famil- Systemic Lupus Erythematosis
ial HUS/Thrombotic Thrombocytopenic Purpura (TTP) Data on lupus nephritis recurrence in children are scarce.
included three patients with documented MCP mutations, This is likely owing to the late presentation of recurrence,
all with excellent graft function at 3 to 13 years posttrans- which for most patients who are transplanted as adolescents
plant.126 However, HUS recurrence has been reported in would occur in adulthood. An analysis of the NAPRTCS
a few patients with MCP mutation. Possible explanations registry reported similar patient and graft survival for chil-
include endothelial microchimerism, in which endothelial dren with systemic lupus erythematosis (SLE) compared
cells of recipient origin (expressing the mutated form of with matched controls.138 There was an increased inci-
MCP) repopulate the transplanted kidney,127 or additional, dence of recurrent rejection episodes in SLE patients receiv-
undiagnosed, genetic susceptibility of the recipient (in cir- ing living donor transplants for which there is no current
culating or fluid phase complement components). Living explanation.
donation is not advised for children with potential aHUS
given the high risk of recurrence and the uncertain effects c-ANCA and p-ANCA-Positive Glomerulonephritis
of gene-gene interactions even from related donors found Pauci-immune glomerulonephritis associated with antineu-
not to carry the same mutation as the recipient. trophil cytoplasmic antibody with cytoplasmic (c-ANCA) or
Plasma therapy has historically been the cornerstone perinuclear (p-ANCA) staining patterns are a rare cause of
of treatment for children with aHUS with CFH mutations. ESRD in children, estimated at 2% of incident ESRD cases
Infusion of fresh frozen plasma (FFP) can provide functional in the US.1 Recurrence rates of the small vessel vasculitides
factor H, factor I, and C3 for patients with deficiencies in (SVV) in the adult literature are low at about 5% to 6%,111
these factors, whereas plasma exchange withdraws anti- and recurrence of granulomatosis with polyangiitis (or GPA;
CFH antibodies and mutated forms of factor H. Preemptive formerly known as Wegener granulomatosis) is rare. Aver-
plasma exchange (PE) initiated before and continuing for age time to recurrence is 31 months, but it can occur within
some time after transplant has been successful in prevent- weeks to many years after transplant. Graft loss due to recur-
ing aHUS recurrence in a small number of patients with CFH rence has been reported in 2% to 7% of adults transplanted
mutation,128 but delayed recurrence can occur with tapering with diagnosis of SVV.
37 • Kidney Transplantation in Children 645

Renal recurrence is often heralded by microscopic hema- visual impairment, hypothyroidism, endocrine pancreatic
turia and proteinuria with focal or diffuse pauci-immune insufficiency, and myopathy) have become more appar-
necrotizing glomerulonephritis seen on biopsy. The ANCA ent as the lifespan of patients with cystinosis increases and
pattern or titers at the time of transplant do not appear to likely are postponed by continued cysteamine treatment
be predictive of disease recurrence. Similar to SLE, a waiting posttransplant.
period of 6 to 12 months of inactive disease is recommended
before transplant. Persistently positive ANCA serologies,
however, should not preclude transplant because they are
not an accurate marker of disease activity. There are anec-
Pretransplant Evaluation
dotal reports of the use of cyclophosphamide, corticoste- EVALUATION OF POTENTIAL LIVING DONOR
roids, mycophenolate, and plasmapheresis for treatment
of recurrence in patients with GPA and p-ANCA associated Evaluation of a potential living donor (see Chapter 7) for
glomerulonephritis.111 a pediatric recipient is similar to that for adults, with the
exception of preference for younger aged donors. Living
METABOLIC DISEASE donors are evaluated for comorbid conditions that would
either increase their own risk for developing ESRD or affect
Primary Hyperoxaluria Type I (Oxalosis) the recipient (i.e., certain viral infections). Adult-sized kid-
Primary hyperoxaluria type I (PH1, also known as oxalo- neys from living donors have excellent potential for long-
sis) is a rare autosomal recessive disorder caused by a defect term graft survival in pediatric recipients. Although the
in hepatic alanine:glyoxylate aminotransferase (AGT), ideal situation would be donation from an HLA-identical
which catalyzes the conversion of glyoxylate to glycine. sibling, most live-kidney donations for pediatric recipients
AGT deficiency results in overproduction of oxalate, result- come from haplo-identical parents because siblings are
ing in massive renal excretion of insoluble calcium oxa- often not able to provide informed consent due to their age.
late leading to nephrolithiasis and nephrocalcinosis. As
GFR declines with progressive renal involvement, oxalate EVALUATION OF RECIPIENT
accumulates and results in systemic oxalosis. More than
100 different mutations have been described, and there is Many similarities exist in the medical evaluation of poten-
considerable phenotypic heterogeneity, even within family tial pediatric and adult transplant recipients (see Chapter 4).
members with identical mutations. However, certain conditions occur more frequently in chil-
The most severe form presents in infancy with renal fail- dren, so the medical evaluation of pediatric recipients has a
ure necessitating dialysis. In children with less severe pre- slightly different emphasis. The following section describes
sentation and early diagnosis, conservative management the common medical, surgical, and psychological issues
with pyridoxine (thought to reduce oxalate production in taken into consideration during the pretransplant evalua-
a subset of B6-responsive patients), increased fluid intake, tion of a pediatric patient.
and citrate treatment might delay progression of kidney
disease.139 Some centers advocate for preemptive liver Medical Evaluation of Issues Related to ESRD
transplant or combined liver–kidney transplant because Cardiovascular Disease. Hypertension is a common prob-
liver transplantation corrects the underlying metabolic lem in children with CKD. Although chronic fluid overload
disorder.140–142 The use of hemodialysis before transplant can result in left ventricular hypertrophy and dilated car-
has also been advocated to reduce systemic oxalate levels diomyopathy, florid heart failure is uncommon in pediatric
to mitigate injury to the allograft. Data from the NAPRTCS CKD patients.146–148 Evaluation of heart function should
registry indicate poor patient and graft survival for children be performed before transplant to identify risk for impaired
with oxalosis after kidney-only transplant with high rates of cardiac output that can negatively affect perioperative fluid
recurrence and death from sepsis. Recent longitudinal stud- allograft perfusion.
ies from Europe suggest improved outcomes in recent years, Pediatric recipients are at risk for exacerbated hyperten-
with the best outcomes in children who were diagnosed sion posttransplant, including malignant hypertension,
early and underwent combined kidney–liver transplant.143 due to the combination of volume expansion, corticosteroid
therapy, and CNI exposure in the postoperative period. Opti-
Nephropathic Cystinosis mized blood pressure control in children with ESRD, includ-
Cystinosis is a rare, autosomal recessive disease due to a ing the use of bilateral nephrectomy149 in cases refractory
defect in the lysosomal cystine transporter (encoded by the to medical management, is paramount to successful trans-
cystinosin gene), resulting in intracellular accumulation of plant outcomes.
cystine, proximal tubule dysfunction (renal Fanconi syn-
drome), and progressive kidney disease. Since the develop- GN of Unknown Etiology. The underlying cause of ESRD
ment of the cystine-depleting drug, cysteamine, progression should be identified in preparation for transplant to antici-
to ESRD may be delayed. Although nephropathic cystinosis pate the risk of recurrence. Complement levels (C3 and C4),
does not recur posttransplant, protocol biopsies have shown antinuclear antibody (ANA), antineutrophil cytoplasmic
interstitial deposition of cystine crystals without appar- antibody (ANCA), and antidouble-stranded DNA (antids-
ent clinical consequences. Graft survival rates for children DNA) titers should be performed in children with suspected
with cystinosis are comparable or even improved compared glomerulonephritis as a cause of ESRD. As mentioned in
with peers with other ESRD etiologies.144,145 Extrarenal the previous section, children suspected of having HUS as
manifestations of continued cystine accumulation (i.e., a cause of ESRD should be evaluated for atypical forms of
646 Kidney Transplantation: Principles and Practice

HUS, including mutational analysis and complement lev- in young children. Gastrostomy tubes can also ensure
els. Finally, identifying a hereditary disease as the cause of adequate hydration in small children who may be unable
ESRD also aids in the evaluation of potential living related to meet the increased posttransplant fluid requirements
donors. orally. Adequate nutrition is also important to optimize
healing from transplant surgery. Therefore pediatric clini-
Nephrotic Syndrome. CNS poses a unique challenge cal dieticians play a vital role in pretransplant evaluation.
in preparation for kidney transplant due to prolonged
hypogammaglobulinemia, malnutrition, chronic ana- Evaluation of Extrarenal Disease
sarca, recurrent infections, and high-risk thrombosis that Infections. Pediatric transplant candidates, like adults,
arise from chronic, uncontrolled urinary losses of various should be free from active infection to minimize complica-
proteins. Some centers perform preemptive unilateral or tions posttransplant. There are several infectious disease
bilateral nephrectomy in these children and interim peri- considerations that should be considered in pediatric trans-
toneal dialysis before transplantation.150,151 This allows plant recipients.
normalization of serum albumin and IgG levels, resolution urinary tract infections. Many children with ESRD
of hypercoagulability, and optimization of nutrition with have abnormal urinary tract anatomy, bladder outlet
concomitant improvement in growth before transplant. obstruction, or vesicoureteral reflux (VUR) that place them
Another approach includes “medical nephrectomy” via the at increased risk for recurrent UTI posttransplant. Indeed,
use of renin-angiotensin system blockade and prostaglan- UTI is the most common bacterial infection in children
din inhibitors to effectively reduce GFR and minimize pro- awaiting kidney transplant. Aggressive antibiotic treat-
teinuria with or without unilateral nephrectomy.152,153 ment and prophylaxis is helpful in suppressing UTI in most
In children with idiopathic nephrotic syndrome, protein- children. Preemptive native nephrectomy149 has been per-
uria typically diminishes as they approach ESRD. Active formed in children with a history of recurrent UTI, severe
nephrotic syndrome is a prothrombotic state due to urinary VUR, and hydronephrosis to reduce the risk of urosepsis
losses of antithrombotic factors leading to increased risk of posttransplant.
perioperative thromboembolic events. Furthermore, hypo- cytomegalovirus. Children are more likely to be CMV
albuminemia can complicate postoperative fluid manage- naïve at the time of transplant than adults. Anti-CMV IgM and
ment because of increased third spacing, thereby increasing IgG titers as well as polymerase chain reaction (PCR) detec-
the risk of electrolyte derangement and graft hypoperfu- tion of CMV viral nucleic acid should be obtained at the time
sion. Continued proteinuria from native kidneys may also of pretransplant evaluation to plan for postoperative CMV
mask the detection of early FSGS recurrence posttransplant. prophylaxis. If initial serologic studies are negative, repeat
Therefore active nephrotic syndrome is the most common studies at the time of transplant can confirm immunologic
indication for native nephrectomies before transplant in naiveté in the recipient. Donor CMV titers are also indicated
children.149 to stratify the risk for developing posttransplant CMV disease.
epstein-barr virus. Similar to CMV, many children
Renal Osteodystrophy. Aggressive treatment of sec- undergoing evaluation for kidney transplant are naïve
ondary hyperparathyroidism, renal osteodystrophy, and to EBV exposure. Primary EBV infection posttransplant
adynamic bone disease with vitamin D analogs and calci- increases the risk of PTLD. Although the use of costimula-
mimetics is vitally important for children with ESRD to opti- tion blockade (e.g., belatacept) is not currently approved
mize growth and anemia management before transplant. in children, the concern for increased risk of PTLD154 for
In general, secondary hyperparathyroidism improves after seronegative recipients has profound implications for the
transplant, but it can often take several months to a year for use of belatacept in the pediatric kidney transplant popula-
parathyroid hormone (PTH) levels to completely normalize. tion; see Chapter 19). Anti-EBV IgM and IgG titers as well
Persistent hyperparathyroidism after kidney transplant can as EBV PCR should be assessed at the time of evaluation
result in hypercalcemia and/or hypophosphatemia, and it and, if initially negative, repeated at the time of transplant
affects growth potential. Ideally, pediatric transplant can- to confirm seronegativity and absence of infection. As with
didates should have intact PTH levels within the National CMV, donor EBV serologies are helpful in stratifying the risk
Kidney Foundation’s Kidney Disease Outcomes Quality of early PTLD as a naïve recipient is exposed to infected leu-
Initiative (KDOQI) or Kidney Disease Improving Global Out- kocytes at the time of transplant.
comes (KDIGO) target range for CKD stage 5 (200–300 pg/ hepatitis b and c. Annual screening for hepatitis B
mL or 2–3 times the upper limit of normal). However, trans- (HBV) remains the standard of care for children on dialy-
plant may proceed safely despite higher PTH levels provided sis. HBV infection in children is uncommon due to the suc-
serum calcium and phosphorus levels are under acceptable cess of newborn vaccination programs. Although hepatitis
control. C (HCV) infection is rarely reported in the general pediat-
ric population, there are small studies suggesting that up
Nutrition and Growth. Poor feeding and linear growth to 20% of children receiving long-term hemodialysis have
delay are prominent features of chronic renal failure in detectable HCV antibodies155 or viral antigens and nucleic
young children. As mentioned previously, most centers acid156 on screening tests. The clinical significance of these
prefer pediatric patients to achieve a weight of 10 to 15 kg results is not clear as the children in these reports were
before kidney transplant, and infants on dialysis may not asymptomatic. Nevertheless, children undergoing evalu-
reach this goal before 2 years of age. Often gastrostomy tube ation for transplant should have HBV and HCV serologic
placement and recombinant growth hormone (rGH) ther- testing along with serum aminotransferase levels to exclude
apy are needed to optimize growth before kidney transplant the presence of active infection before transplant.
37 • Kidney Transplantation in Children 647

immunization status. Routine childhood immuni- Prior Malignancy. In general, children with ESRD do not
zations should be completed, if possible, before kidney need screening for preexisting malignancy, but a prior his-
transplant.157,158 Live attenuated virus vaccines (i.e., tory of malignancy warrants additional evaluation. Wilm
measles-mumps-rubella and varicella vaccines) are gener- tumor is the most common malignancy resulting in ESRD
ally contraindicated in immunosuppressed patients due to in children. A waiting period of 2 years after treatment for
the increased risk of disseminated disease by the vaccine Wilm tumor is recommended and has resulted in excel-
virus strain. Therefore children awaiting transplant should lent outcomes with a low risk of recurrence after trans-
receive live virus vaccination at least 1 to 2 months before plant.165,166 However, reevaluation of the recommended
transplant. Other, nonlive-virus vaccinations (i.e., hepatitis waiting time is being considered due to the high morbidity
A, Tetanus-diptheria-acellular pertussis or Tdap, expanded and mortality associated with dialysis, and improvements
13-valent pneumococcal, meningococcal, and human pap- in the prevention and treatment of acute rejection since
illoma virus or HPV) should also be administered before the original recommendations were established.167 Kidney
transplant because the immunosuppressant therapy can transplant in children with a history of other extrarenal
impair immunologic response to these vaccines. Children malignancies is generally considered after a recurrence-free
awaiting transplant should also receive annual influenza period of 2 to 5 years.
vaccination. Antibody titers against hepatitis B, hepatitis
A, measles, mumps, rubella, and varicella should be evalu- Surgical Evaluation
ated to determine whether a booster vaccination is needed Children undergoing kidney transplant evaluation often
within 6 months of transplant.158–160 Further research is need multiple surgical interventions, including bladder
needed to develop optimal vaccine protocols to minimize augmentation, placement of Mitrofanoff continent urinary
waning of protective immunity posttransplant.161 diversion, vesicostomy closure, gastrostomy tube place-
ment, or native nephrectomy before or at the time of trans-
Hemostasis. As mentioned earlier, thrombosis is a signifi- plant. Therefore it is vital that a surgical plan be established
cant cause of graft loss in very young recipients. However, among pediatric surgeons, transplant surgeons, and urolo-
identifying children at highest risk for this complication gists before kidney transplant in children to coordinate
has been difficult. Retrospective studies have identified surgical approaches and spare or maintain the vascular
the following risk factors for graft thrombosis in pediatric supplies that are unique for each procedure.
recipients: recipient age under 5 years, history of peritoneal
dialysis,162 high urine output pretransplant, young donor Vascular Evaluation. The abdominal vasculature should
age (<5 years old), and prolonged cold ischemia times (>24 be assessed for patency in preparation for transplant sur-
h).163,164 Central venous lines are the most common vas- gery. Children with prior history of femoral lines (includ-
cular access for children receiving hemodialysis, and line- ing dialysis catheters) or inflammatory conditions of the
associated thrombosis is a common event in this patient abdomen (such as multiple abdominal surgeries or recur-
population. Data linking prior history of catheter-associ- rent peritonitis) are at increased risk of thrombosis of the
ated thrombosis in children on dialysis with risk of graft inferior vena cava (IVC) or iliac vessels, thereby complicat-
thrombosis after transplant are not established. ing vascular anastomoses of the graft. Magnetic resonance
There are few data on the prevalence of inherited hyper- venogram (MRV) or computed tomography angiography
coagulability that may predispose some children to graft (CTA) are sensitive techniques for assessing IVC patency as
thrombosis. In adults, inheritance of factor V Leiden or well as providing a detailed anatomic survey of abdominal
prothrombin (G20210A) mutations significantly increase vasculature. In patients at lower risk of thrombosis, Doppler
the risk of graft thrombosis. Adults with SLE, especially in ultrasound is useful to screen for IVC and iliac vein patency,
the presence of detectable antiphospholipid antibody or β2- but may be dependent on operator expertise, especially in
glycoprotein-1, are at particularly high risk of thromboem- small children.
bolic events.163 There are insufficient data to determine the
risk of posttransplant thrombosis in children with hyperho- Urologic Evaluation. As mentioned earlier, nearly 25%
mocysteinemia or 5,10-methylene tetrahydrofolate reduc- of children receiving kidney transplants have underlying
tase (MTHFR) polymorphisms. urologic abnormalities, including lower tract obstruction,
Children with a history of recurrent thrombotic events, vesicoureteral reflux, or bladder dysfunction. Therefore
a strong family history of thrombophilia, or significant pediatric urologists play an important role in the manage-
proteinuria should undergo coagulation workup by a ment of these patients before and after transplantation.
specialist to determine whether chronic anticoagulation All children under consideration for kidney transplant
therapy is warranted. Evaluation of hypercoagulability should have a renal ultrasound to evaluate for hydro-
includes measurement of prothrombin time (PT), partial nephrosis, hydroureter, and bladder wall thickening. A
thromboplastin time (PTT), platelet count, fibrinogen, voiding cystourethrogram (VCUG) should be performed in
ATIII level, protein C and protein S levels, and activated selected children, including those with a history of urologic
protein C resistance (to monitor factor V Leiden). Ado- causes of ESRD, history of UTIs, hydronephrosis on ultra-
lescents with a history of SLE should be screened for sound, or signs (e.g., thickened bladder wall on ultrasound)
antiphospholipid antibody, anticardiolipin antibody, and or symptoms suggestive of dysfunctional voiding.
β2-glycoprotein-1. Further workup, including muta- Urodynamic studies are indicated for selected children
tional analysis in other genes associated with inherited with urinary tract abnormalities to assess bladder capacity,
thrombophilia should be undertaken under the advice of compliance, voiding pressure, leak point pressure, and post-
a pediatric hematologist. void residual (see Chapter 12). Early, aggressive treatment
648 Kidney Transplantation: Principles and Practice

of neurogenic or dysfunctional bladder is critical as elevated syndrome, are at increased risk for developing Wilm tumor
bladder pressures have been associated with increased risk over time. Therefore bilateral nephrectomy is considered
of developing reflux into the transplanted kidney and worse once they approach ESRD.177
graft survival.168–170 Medical management of neurogenic Many centers, including ours, perform pretransplant
bladder often includes anticholinergic therapy combined native nephrectomies for patients on dialysis with recalci-
with intermittent catheterization and should be contin- trant hypertension with good short-term results, including
ued after transplant. Children with a history of posterior achievement of age-normative blood pressures and a reduc-
urethral valves should be assessed for persistent anatomic tion in the number and dosage of antihypertensive medi-
obstruction after valve ablation or have a neourethra (e.g., cations. The efficacy of bilateral nephrectomy in improving
Mitrofanoff) that can be intermittently catheterized after blood pressure control for children on dialysis or prevent-
transplant to prevent lower urinary tract obstruction. ing posttransplant hypertension has not been extensively
In some cases, poor bladder compliance persists despite studied. Results reported from our center involving uni-
appropriate medical management, necessitating blad- lateral or bilateral nephrectomy of diseased native kidneys
der augmentation171 with small bowel or colon segments have shown good results during short-term follow-up.178
before transplant. However, otherwise healthy but small Similarly, children with multicystic dysplastic kidney dis-
capacity, defunctionalized bladders in children with oligu- ease or reflux nephropathy resulting in a unilateral poorly
ric ESRD do not require augmentation to achieve excellent functioning kidney have been reported to have resolution
graft outcomes.172,173 Typically, urologic correction should of hypertension or diminution of antihypertensive medica-
be undertaken 3 to 6 months before transplant to allow for tions after nephrectomy.179 However, a recent retrospective
adequate healing before immunosuppression.174 study did not demonstrate an effect of bilateral nephrec-
Because many children with urologic disorders have tomy on the risk of posttransplant hypertension or left ven-
nonoliguric renal failure, careful assessment of daily urine tricular hypertrophy.180 Prospective longitudinal studies
volume by history and 24-hour urine collection is war- are needed to evaluate whether bilateral nephrectomy for
ranted to plan for posttransplant fluid management and hypertension improves long-term cardiovascular morbidity
need for native nephrectomy. in children receiving kidney transplant.
The surgical approach to nephrectomy (retroperitoneal
Native Nephrectomy. The most common indications for vs. transperitoneal; open vs. laparoscopic) should be based
native nephrectomy in children undergoing kidney trans- on the center’s expertise and the needs of the patient (e.g.,
plant are excessive urine output, high-grade vesicoureteral receiving peritoneal dialysis).181 Although a recent report
reflux with recurrent or recalcitrant pyelonephritis, con- from Italy suggests that renal embolization in children
tinued heavy proteinuria (see Primary Glomerulonephritis may be a minimally invasive alternative to nephrectomy
section for risk for recurrent diseases with nephrotic-range for indications other than risk of malignancy,182 we have
proteinuria section), uncontrolled hypertension, and risk of observed unacceptable morbidity from this procedure and
renal malignancy.149 would not recommend this approach.
Unique to pediatrics, excessive urine output (>20 mL/kg/
24 h) occurs more often in small children receiving an adult- Neurologic Development
size graft and has been associated with risk of renal transplant Developmental Delay. It is increasingly recognized that
thrombosis.164 Furthermore, high urine output after the children with CKD have higher rates of neurocognitive
immediate posttransplant period (>4 L/day) can complicate delays. Factors that have been associated with increased
fluid management for small children for whom it would be risk for neurocognitive deficits include longer duration of
difficult to consume such large volumes to avoid hypovolemia. CKD, increased severity (i.e., advanced stages of CKD), and
Nephrectomy of a native kidney with severe reflux (grade younger onset of disease.183
3 or 4) should be considered to reduce the risk for early UTI Children with ESRD during infancy have significant
or urosepsis in the setting of immunosuppression. Preemp- developmental delay due to uremia. In the absence of struc-
tive native nephrectomy has been advocated for certain tural brain abnormalities, psychomotor delay can improve
patients for which the underlying renal disease results in after transplant, with many infants regaining normal
continued urinary losses of electrolytes (i.e., for Bartter developmental milestones.52 Although overall neurode-
syndrome)175 or heavy proteinuria (i.e., FSGS or CNS)176 velopmental outcomes are favorable after transplant, in a
complicating the optimal nutritional and medical man- prospective study of children undergoing kidney transplant
agement of children before transplant. Reduction in heavy before the age of 5 years, lower IQ and learning disabilities
proteinuria due to FSGS after nephrectomy may reduce were most common in children born prematurely and those
the risk of thrombotic events by correcting the hyperco- who had multiple hypertensive crises and/or seizures during
agulable state and allow for earlier recognition of recur- dialysis.184 This study suggests that prevention of hyperten-
rent FSGS after transplant. As mentioned earlier, children sion-related morbidity may improve neurodevelopmental
with CNS may benefit from improved nutrition and reduced outcomes for young children after kidney transplant.
risk for infection after nephrectomy. Finally, children with Other infants needing dialysis in the first year of life may
autosomal recessive polycystic kidney disease (ARPKD) have structural neurologic abnormalities resulting from
often require unilateral or bilateral nephrectomy of rapidly insults associated with premature birth or anoxic/ischemic
enlarging kidneys to allow for both improved respiratory injury (e.g., hypoxic ischemic encephalopathy, periventric-
status and adequate space in the abdomen for a future kid- ular leukomalacia after intraventricular hemorrhage, or
ney graft. Children with disorders related to mutations in microcephaly). Children with structural neurologic abnor-
the WT1 gene, such as Denys-Drash syndrome and Frasier malities can present with hypotonia, spasticity, myoclonus,
37 • Kidney Transplantation in Children 649

severe cognitive delays, and seizures. Children with severe Evaluation Updates
mental retardation may not respond well to the constraints Pediatric candidates for kidney transplant should be reeval-
of ESRD care, where the need for multiple, and often pain- uated at regular intervals (generally every 6–12 months) to
ful, procedures can be confusing and uncomfortable. In identify any changes in their medical condition or psycho-
such situations, the potential for rehabilitation, self-care, social status that might alter the risk of commencing with
and parental preferences should be considered during joint transplantation. A simplified version of the initial medical
decision making between the medical team and family/ evaluation is appropriate for update visits.
caregivers in determining whether long-term dialysis or
transplantation should be pursued.

Seizures. Up to 20% of children with ESRD are treated for


Perioperative Management of
seizure disorder typically related to hypertensive crises, Pediatric Renal Transplant
and about 5% require ongoing anticonvulsant therapy Recipients
posttransplant.185,186 Adequate seizure control should be
obtained before transplantation, preferably with anticon- PREOPERATIVE MANAGEMENT
vulsants that do not interfere with the metabolism of com-
monly used immunosuppressant medications. Phenytoin Children presenting for imminent kidney transplant sur-
(Dilantin), barbiturates, and carbamazepine can signifi- gery should be clinically stable without signs of active infec-
cantly reduce serum levels of CNIs and prednisone. Newer tion. A final set of laboratory studies is obtained to evaluate
anticonvulsants may not interfere with immunosuppres- for any electrolyte or metabolic derangements that require
sant drug levels, but it is prudent to check for updates in dialysis or medical treatment before anesthesia induction.
drug-drug interactions when planning for transplant. Intravascular volume status is important before trans-
plant surgery as children with hypovolemia (especially
Psychosocial Issues those with high urine output) are at increased risk of graft
Psychoemotional Status. A multidisciplinary approach thrombosis164 and graft hypoperfusion leading to ATN.
with involvement of physicians, child psychologists, and Therefore, if dialysis treatment is indicated before surgery,
child life specialists is important for the preparation of chil- excessive fluid removal should be avoided. Similarly, chil-
dren and their families for kidney transplantation. Children dren with residual urine output should receive intravenous
with emotional or psychiatric disorders often require addi- (IV) fluids to maintain intravascular volume while oral
tional mental health resources including psychiatric care. intake is restricted awaiting surgery.
Acquisition of coping skills, problem-solving skills, and Subclinical infections of skin, dialysis access site, peri-
behavior modification can improve a child’s experience with toneal fluid, and urinary tract should be evaluated with a
the inherent complexity of dialysis or transplantation medi- thorough history, physical examination, and laboratory
cal care. Pharmacotherapy for depression, bipolar disorder, studies, including urinalysis and urine culture, peripheral
and attention deficit hyperactivity disorder are important white blood cell count with differential blood cultures for
adjunctive therapies. Reduced clearance with impaired those with indwelling venous catheters, and peritoneal
renal function, clearance by dialysis, and interference with cell count and culture for those maintained on peritoneal
the metabolism of immunosuppressive medications should dialysis. A recent episode of peritonitis or peritoneal dialy-
be considered when selecting psychotropic medications sis (PD) catheter exit-site infection does not preclude trans-
in children with ESRD. Most selective serotonin reuptake plantation, but the child should complete 10 to 14 days of
inhibitors (SSRIs) do not interfere with immunosuppressive antibiotics and have a negative peritoneal fluid culture off
medications. antibiotics before transplant.
As mentioned earlier, CMV and EBV serologies should be
Nonadherence. Suspected nonadherence contributes to repeated if previous results revealed immunologic naiveté.
approximately 44% of graft losses and 23% of late acute Final HLA crossmatch should be performed within 1 week
rejection episodes reported in the literature for adolescent before living-donor kidney transplant or in the hours pre-
kidney transplant recipients.187 Patterns of medication and ceding deceased-donor transplant.
dialysis treatment compliance should be assessed for every
child undergoing evaluation for kidney transplant to iden- INTRAOPERATIVE MANAGEMENT
tify patients at high risk for nonadherence posttransplant.
Social, behavioral, and psychiatric interventions should be Small children can present operative challenges given
initiated before transplant for those patients with identified the relatively large size of an adult kidney graft compared
or anticipated issues with noncompliance. Identification with recipient size. Children weighing more than 30 kg
and nurturing of psychosocial support systems and fre- are often treated surgically as small adults with graft
quent medical and social work follow-up are often required placement in the standard extraperitoneal pelvic location
to prepare the pediatric candidate for transplantation. and vascular anastomoses to the common iliac artery and
Again, the best chance of rehabilitation and preparation vein. However, in small children intraabdominal place-
for transplant is achieved when there is close coordination ment may be preferable with vascular anastomoses to the
between medical and mental health providers. It is also of infrarenal aorta and IVC. The surgical approach should
particular importance for the transplant and dialysis medi- be individualized with appropriate matching of blood
cal teams to maintain close communication as the recipient vessel size and attention to expected circulatory volume
prepares for transplant. requirements.
650 Kidney Transplantation: Principles and Practice

Intraoperative management of the pediatric kidney trans- crystalloid. Hypokalemia and hypophosphatemia may
plant recipient is focused on achieving optimal graft perfu- develop in the first few postoperative days. Potassium and/
sion and preventing complications arising from underlying or phosphate salts can be added to the replacement fluids as
ESRD. In small children, a central venous catheter is often appropriate.
inserted for close monitoring of central venous pressures. As the kidney graft regains urinary concentrating abil-
Central venous pressures (CVP) should be maintained at ity, urine output declines to levels that are more reason-
12 to 18 cmH2O and mean arterial pressures (MAP) above ably achievable as daily oral intake. Urine replacement
70 mmHg via infusion of crystalloid or 5% albumin before with IV crystalloid can be discontinued at that time,
clamp release to ensure adequate perfusion to the adult- and intake goals of 150% to 200% of calculated main-
sized transplanted kidney.188 tenance needs should be started by mouth. Children
It is important to recognize the challenge that this can with intraabdominal graft placement are susceptible to
pose in children under 30 kg, as a kidney graft from an prolonged postoperative ileus due to displacement of the
adult can sequester 150 mL to 250 mL of blood, which colon and intestines with an adult-sized graft occupying
can represent well over 10% of a small child’s total circu- almost the entire right side of the abdomen. These chil-
lating volume. In infants, up to 50% of cardiac output is dren often require continuation of maintenance IV flu-
directed to perfusion of an adult-sized kidney graft. Transfu- ids beyond the first few days until they can tolerate oral
sion with packed red blood cells (pRBC) is often necessary fluids and nutrition. Small recipients (<15 kg) of large
in the smallest recipients to avoid severe anemia. In addi- adult kidneys are at risk for developing the uncommon
tion, continuous dopamine infusion at 2 to 3 μg/kg/min is complication of abdominal compartment syndrome with
often necessary, especially in infants, to maintain higher increased intraabdominal pressure complicated by oligu-
MAP and is continued for 24 to 48 hours postoperatively ria and hypotension.191
to allow the graft to slowly accommodate to lower MAP in Hypertension is commonly observed in children after
the recipient.188 Finally, mannitol (1 g/kg) with or without transplant. In some instances, elevated blood pressures
furosemide (1 mg/kg) is often administered before clamp improve with adequate analgesia. Fluid overload can
removal to facilitate diuresis. Mannitol infusion may also also contribute to postoperative hypertension and often
prevent sudden shifts in serum sodium and acute decreases improves once spontaneous mobilization of fluid occurs.
in serum osmolality, which increases the risk for postopera- Aggressive treatment of mild to moderate hypertension is
tive neurologic complications.189 typically not recommended early posttransplant to avoid
Blood gases and lactate levels should be monitored intra- sudden changes in MAP and decreased graft perfusion. This
operatively because clamping of the aorta or iliac artery can being said, children who were on multiple antihypertensive
result in lactic acid accumulation, metabolic acidosis, and medications before transplant may need reinstitution of
vasoconstriction. Intraarterial injection of calcium chan- at least some of their previous medications to avoid severe
nel antagonists (e.g., verapamil or papaverine) or nitro- rebound hypertension and the accompanying adverse
glycerin190 have been used to overcome arterial spasm that effects.
impairs graft perfusion. Postoperative allograft ultrasound is performed routinely
For most immunosuppression protocols, intravenous in many pediatric transplants centers, although there is
methylprednisolone sodium succinate (Solu-Medrol) is no consensus on the timing or necessity of the imaging.
administered at the beginning of the surgical case. In Information gained by ultrasound includes assessment of
addition, many pediatric transplant centers use intrave- vascular patency, increased resistive indices suggestive of
nous biologic agents for induction therapy, which are also risk of delayed graft function, identification of surgical com-
administered intraoperatively. plications (e.g., lymphocele), and detection of collecting
duct dilatation to identify those at risk for future urologic
complications.192,193
POSTOPERATIVE MANAGEMENT
Goals for hospital discharge of the pediatric transplant
Children are monitored in the intensive care unit setting recipient include adequate oral fluid intake to prevent
for the immediate postoperative period. In the first 2 to 3 hypovolemia (and subsequent graft hypoperfusion), stable
days, the postoperative management is focused on optimiz- immunosuppression regimen, completion of family and
ing graft perfusion and mitigating the effects of fluid over- caregiver education, access to medications, and arrange-
load (e.g., electrolyte derangements and hypertension). As ment of outpatient follow-up.194
mentioned earlier, small children often require dopamine
infusion for 24 to 48 hours posttransplant to maintain graft
perfusion and allow gradual accommodation of the graft to
Immunosuppressive Protocols and
the lower mean arterial pressures of the recipient. Drugs
Fluid management in small children requires fastidious
care due to their small size and potentially very large post- Several other chapters in this book are devoted to a detailed
transplant urine volumes. Urinary losses are replaced in discussion of the various classes of immunosuppressant
equal volumes with intravenous 0.45% or 0.9% sodium medications used for induction, maintenance, and treat-
chloride infusion for the first 24 to 48 hours. Dextrose ment of rejection after kidney transplantation (see Chapters
should be withheld from the initial IV fluids given for urine 15–20). Therefore this chapter will discuss pediatric-spe-
replacement to avoid hyperglycemia and osmotic diuresis. cific issues related to various immunosuppressant medi-
Replacement of insensible water losses should be admin- cations and trends in immunosuppressant protocols after
istered as a separate infusion with dextrose-containing kidney transplantation in children.
37 • Kidney Transplantation in Children 651

Infants and small children cannot swallow pills; there- administered via central venous catheter. Although ATG
fore they frequently require special formulations of immu- formulations are better tolerated than OKT3, they still carry
nosuppressive medications (i.e., compounded solutions). the risk of severe cytokine release and anaphylaxis; accord-
Furthermore, it has become increasingly recognized that ingly, premedication with corticosteroids, acetaminophen,
the metabolism of these medications undergoes maturation and antihistamine 30 to 60 minutes before infusion is
in children, so pharmacokinetics can vary by age. Therefore recommended.
children with kidney transplants may require a different Thymoglobulin induction is typically administered at 1 to
dosing schedule than that published in the adult literature. 2 mg/kg/day for a cumulative dose of 7 to 10 mg/kg. There
There are often limited data on the safety, efficacy, and is limited information about the average dosing used at pedi-
pharmacokinetics of immunosuppressant medications atric centers. However, the NAPRTCS registry reports that
in children due, in part, to limited inclusion of children in median duration of ATG treatment in participating centers
clinical trials. Consequently, most of the immunosuppres- is 5 days. A recent retrospective study suggests that a lower
sant medications used to prevent or treat rejection in pedi- cumulative dose of 6 mg/kg was noninferior to historical
atric kidney transplant recipients are not approved by the doses of 10 mg/kg in preventing acute rejection in pediatric
US Food and Drug Administration (FDA) for use in children kidney transplant patients.197 Intermittent therapy based
and therefore are administered as “off-label” use. on peripheral CD3 + lymphocyte counts has been suggested
to reduce the cumulative dose with a low rate of acute rejec-
tion in high-risk adult kidney transplant recipients,198 but
INDUCTION THERAPY AGENTS
this practice has not been validated in children. Single-cen-
Fig. 37.4 illustrates the trends in the use of induction anti- ter retrospective studies report adequate graft survival rates
bodies for pediatric kidney transplants reported in the 2015 in children after rATG and triple immunosuppression with
SRTR annual report. Induction therapy of some form was 10-year follow-up.199,200
reported for approximately 91% of pediatric kidney trans- A small study in pediatric kidney transplant recipients
plant recipients. The use of T cell depleting agents has con- suggested greater T cell depletion after 5 days of Thymo-
tinued to increase over the past decade, reaching 61% in globulin compared with 10 days of Atgam.201 However,
2015, whereas the use of IL-2 receptor antagonist (IL2RA) a direct comparison of patient outcomes between Thymo-
has remained steady at 33.3%, and fewer patients are globulin and Atgam induction has not been reported in
receiving no induction therapy. Although adult trials have pediatric recipients. Atgam is typically dosed at 10 to 15
demonstrated decreased rates of rejection using induction mg/kg daily for 5 to 10 days.
therapy, there are no controlled trials in pediatrics to verify Concerns have been raised regarding an increased risk
the benefits of this practice. A retrospective analysis of pedi- of PTLD associated with depletional induction therapy,
atric recipients in the UNOS registry found no difference in especially with high doses of OKT3 or Thymoglobulin.202
the number of rejection episodes or 3-year graft survival for However, long-term exposure to CNIs and mammalian tar-
children receiving polyclonal rabbit antithymocyte globu- get of rapamycin (mTOR) inhibitors or possibly the balance
lin (rATG) or monoclonal anti-IL2RA antibody compared between B cell and T cell depletion may be more relevant
with no induction (all were discharged with triple immu- determinants of PTLD risk.203,204
nosuppressive maintenance therapy, including a CNI, anti-
metabolite, and steroids).195 A retrospective study of SRTR Alemtuzumab. Alemtuzumab (or Campath-1H) is a
data suggests that lymphocyte-depleting agents reduced monoclonal antibody directed at CD52-positive T cells and
the risk of acute rejection for African American pediatric B cells. The use of alemtuzumab induction in adult kidney
recipients compared with IL2RA induction.196 transplant recipients, along with reduced doses of CNIs and
mycophenolate (MMF) and without long-term steroids,
Lymphocyte-Depleting Agents have had encouraging results in small series.205 However,
OKT3. The first monoclonal drug available in this class pharmacokinetic data and long-term outcomes in children
was the lymphocyte-depleting drug, muromunab-CD3 (or receiving alemtuzumab induction are still limited. Bartosh
OKT3). Administration of OKT3 was complicated by severe et al. first described the use of alemtuzumab in four high-
symptoms of cytokine release, often referred to as the “first- risk pediatric kidney transplant recipients.206 They reported
dose effect.” Use of OKT3 for children undergoing kidney similar prolonged lymphocyte depletion and increased risk
transplant fell out of favor in the late 1990s with the avail- of developing anti-HLA antibodies in the absence of CNI,
ability of alternative depleting agents that were better tol- as had been reported in adults. Since then, alemtuzumab
erated. The manufacturer of OKT3 voluntarily withdrew it induction in pediatric kidney transplant recipients has
from the US market due to decreased demand. allowed for steroid-free immunosuppression207,208 and
reduced CNI exposure.209 The optimal dose of alemtuzumab
ATG. Polyclonal antithymocyte globulins (or ATG) are in pediatric recipients is still unclear. The Pittsburgh group
the most commonly used induction therapy for pediatric reports a single dose 0.4 to 0.5 mg/kg for induction,210 and
kidney transplant recipients in the US.195 There are two the prospective phase II trial (PC-01) utilizes two doses (on
preparations in use: one derived from the immunization of day −1 and day +1) of 0.3 mg/kg (with a maximum dose of
rabbits (rATG or Thymoglobulin, Genzyme) and one from 20 mg).209
the immunization of horses (hATG or Atgam, Upjohn). In Concerns regarding development of de novo anti-HLA
Europe, a similar polyclonal antibody derived from rabbit antibody production after alemtuzumab have been raised. A
serum is available (ATG-Fresenius S, Fresenius AG). Due longitudinal study of the immune profile and development
to the sclerosing nature of these formulations, they are of anti-HLA antibodies in children receiving alemtuzumab
652 Kidney Transplantation: Principles and Practice

induction was published as part of a multicenter clinical for avascular necrosis of the femoral head, has prompted
trial (PC-01) with initial maintenance of tacrolimus and increased use of regimens minimizing steroid exposure in
MMF followed by conversion to sirolimus and MMF at 2 to 3 children. In addition, the undesirable cosmetic effects of
months for low-risk children receiving living-donor kidney long-term corticosteroid use, including cushingoid facial
transplants (n = 35).209 The cumulative hazard of develop- features and acne, also likely contribute to nonadherence,
ing circulating anti-HLA antibodies in children (roughly particularly in adolescent patients.
33% at 24 months posttransplant) was higher than that
reported in the adult literature (15%–20% at 1–5 years).
However, development of anti-HLA antibodies in this study Induction Therapy Use
was not associated with a decline in renal function or his- 100
tologic changes on 2-year surveillance biopsy. Long-term T-cell depleting
follow-up of these patients will be needed to assess the 80 IL2-RA

Percentage (%)
potential benefits of this immunosuppressive regimen in None
children. 60
A small single-center study of alemtuzumab induction in
40
highly sensitized pediatric patients resulted in graft survival
comparable to patients with low PRA receiving nondeplet- 20
ing induction with IL2RA.211
0
Nondepleting Agents 2006 2008 2010 2012 2014 2016
Anti-IL2RA Antibodies. There have been two high-affin-
ity monoclonal antibodies against the inducible α-chain of Maintenance Immunosuppression
the IL-2 receptor (IL2RA or CD25) in use for the preven- 100
Tacrolimus
tion of allograft rejection in children. Basiliximab (Simulect, Mycophenolate
80
Novartis) is a chimeric antibody approved for the preven-
Percentage (%)
Cyclosporine
tion of acute rejection in pediatric kidney transplant recipi- 60 Azathioprine
ents and is generally given in two doses (10 mg for children
<35 kg and 20 mg for children >35kg) on days 0 and 4 40
posttransplant. Daclizumab (Zenapax, Roche), which was
withdrawn from the US market in 2009, is a humanized 20
antibody and is administered at a dose of 1 mg/kg every 14 0
days for five doses beginning within 24 hours of transplant. 2006 2008 2010 2012 2014 2016
Daclizumab was favored in steroid-free protocols in chil-
dren212 due to the prolonged saturation of the IL-2 receptor Corticosteroid Use
for approximately 90 days posttransplant.213 100
Anti-IL2RA antibody induction is overall well-tolerated At transplant
in children. There is one report, however, of two children 80 At 1 year
Percentage (%)

who developed noncardiogenic pulmonary edema in the


absence of delayed graft function.214 Basiliximab is com- 60
monly combined with triple therapy immunosuppression
40
with CNI, MMF, and prednisone.215
20
MAINTENANCE THERAPY
0
Fig. 37.4 illustrates the trends in maintenance immuno- 2006 2008 2010 2012 2014 2016
suppression for children receiving kidney transplants in the
US during the past decade as reported by the SRTR. There mTOR Inhibitor Use
are several different immunosuppression protocols in use in 20
At transplant
children. More than 90% of children in the US receive tacro- At 1 year
limus and mycophenolate as initial maintenance immu- 15
Percentage (%)

nosuppression. Overall trends include decreased usage of


cyclosporine and azathioprine and increased use of tacro- 10
limus and mycophenolate. Approximately 60% of pediatric
recipients in the US receive corticosteroids posttransplant, a 5
trend that has remained stable since 2006.
Corticosteroids 0
2006 2008 2010 2012 2014 2016
Corticosteroids have long played a key role in the prevention
of rejection after solid-organ transplant. However, focus on Fig. 37.4 Trends in immunosuppression use among pediatric trans-
plant recipients as reported by the 2015 Scientific Registry of Trans-
reducing the off-target side effects of long-term glucocorti- plant Recipients (SRTR) report. (Hart A, Smith JM, Skeans MA, et al. OPTN/
coid exposure, including impaired linear growth, reduced SRTR 2015 Annual Data Report: Kidney. Am J Transplant 2017;17(Suppl 1):
bone density, hypertension, glucose intolerance, and risk 21–116.)
37 • Kidney Transplantation in Children 653

Steroid-Based Regimens. Steroids continue to be uti- differences in acute rejection episodes in the two groups, and
lized as a maintenance immunosuppression therapy in the steroid withdrawal group had superior 3-year allograft
approximately 60% of children receiving kidney trans- survival.223 However, the high rate of complications (most
plantation in the US. The use of more potent immunosup- notably of PTLD) led to the early termination of this study
pressant medications, namely tacrolimus and MMF, have and recommendations against the routine use of this potent
allowed for overall reduction of the prednisone dose used immunosuppressant protocol in children.
for prevention of rejection and the mitigation of steroid
side effects. Although reduction of the corticosteroid dose Steroid Avoidance/Early Withdrawal. Steroid avoid-
has been demonstrated to improve hypertension, bone and ance regimens have included either a complete steroid-free
joint complications, and cataracts, this strategy has not approach or early steroid withdrawal before 7 days post-
been shown to effectively address the suppression of lin- transplant. There is a large variation in the use of steroid-
ear growth in children.216 Alternate-day administration of avoidance protocols among pediatric transplant centers
equivalent cumulative prednisone dose has been suggested in the US.224 In general, steroid avoidance is less likely to
as a strategy to allow catch-up growth without compro- be used for children who are highly sensitized, receiving a
mising graft survival.217,218 This regimen, however, may deceased-donor transplant, have underlying glomerular
be more cumbersome for the patient and family and more disease, or are of African American ethnicity. However,
prone to nonadherence. steroid avoidance after Thymoglobulin induction has been
More recently, studies of immunosuppressant protocols successfully used in a small series of high immunologic risk
to minimize glucocorticoid exposure in pediatric kidney pediatric recipients with equivalent rates of rejection com-
transplant recipients have focused on either steroid with- pared with low immunologic risk recipients receiving ste-
drawal (early or late) or total steroid avoidance. The follow- roid-free maintenance after daclizumab induction.225
ing paragraphs will summarize the major studies reporting Initial protocols used long-acting IL-2 receptor antagonist,
these approaches in children. daclizumab, for induction. However, with the withdrawal of
daclizumab from the market in 2009, most steroid-avoidance
Steroid Withdrawal (Late). Initial reports of late steroid regimens now utilize induction therapy with depletional
withdrawal were concerning for increased risk of rejection agents, such as ATG226 or alemtuzumab.207,208
and allograft loss.219 More recently, however, a multicenter Several studies of early steroid withdrawal (within 5–7 days
randomized open-label study in children with low immuno- posttransplant) have reported similar rates of acute rejection
logic risk compared late steroid withdrawal (greater than and graft survival compared with historic or nonrandom-
1 year posttransplant) to continuous steroid treatment ized controls receiving long-term steroids. The TWIST study
and maintenance therapy with cyclosporine microemul- was a multicenter randomized open-label trial of 196 pedi-
sion and mycophenolate, with similar rates of acute rejec- atric patients to evaluate the effect of daclizumab induction
tion observed between groups and stable graft function (two doses) with tacrolimus, MMF, and rapid steroid with-
at 2 years after steroid withdrawal.220 The patients who drawal (by day 5) compared with triple therapy with tacroli-
underwent late steroid withdrawal benefited from superior mus, MMF, and standard-dose prednisone (no induction) on
longitudinal growth, less frequent hypertension, reduc- growth and steroid-related metabolic complications in pediat-
tion in the number of antihypertensive medications, and ric kidney transplant recipients.227 At 2 years follow-up (106
improved carbohydrate and lipid metabolism compared patients), they reported similar rates of graft survival and esti-
with the patients remaining on steroids (5 mg/m2 predni- mated GFR between steroid-based and steroid-withdrawal
sone daily). This study suggests that withdrawal of steroids groups. However, linear growth was superior in the steroid-
in select stable pediatric transplant recipients receiving CNI withdrawal group, especially for prepubertal children.228
therapy and MMF is safe and may mitigate the metabolic Long-term results (3-year follow-up) of a multicenter,
and cardiovascular side effects of prolonged steroid use. Of randomized trial comparing steroid-free (SF) and steroid-
note, the effect on growth observed in this study was more based (SB) immunosuppression with concomitant tacroli-
pronounced and occurred earlier in prepubertal patients, mus, MMF, and standard dose daclizumab (SB) or extended
but even the pubertal patients had significant improvement daclizumab induction (SF) in unsensitized pediatric kidney
in mean standardized height after 24 months of withdrawal transplant recipients found no difference in biopsy-proven
of steroids. acute rejection or graft survival between the two groups.229
Valid concerns have arisen regarding excessive immu- Furthermore, subclinical rejection was no different between
nosuppression with newer agents in protocols designed patients receiving SF immunosuppression and those receiv-
to minimize exposure to steroids.221 This was poignantly ing a SB regimen in a randomized controlled trial on steroid
exemplified by the early termination of the only randomized avoidance.230 Finally, an analysis of UNOS data found that
double-blinded controlled study of late steroid withdrawal children receiving steroid-avoidance maintenance immu-
(commencing at 6 months) in pediatric kidney transplant nosuppression had similar rates of rejection and graft sur-
recipients due to an unacceptably high rate of PTLD.222 vival compared with children on SB regimens.231
Patients in this study received intensified early immunosup- Multiple studies have suggested improved cardiovascu-
pression with basiliximab induction and maintenance with lar profile, including decreased hypertension, lower choles-
prednisone, cyclosporine or tacrolimus, and sirolimus fol- terol, and decreased rates of left ventricular hypertrophy in
lowed by randomization to either undergo complete steroid children receiving steroid-avoidance immunosuppressive
withdrawal (gradually over 6 months if they had no clini- regimen.226,232,233 Linear growth and lean body mass are
cal or histologic evidence of rejection on a 6-month proto- optimized for prepubertal patients maintained on steroid-
col biopsy) or to continue low-dose steroids. There were no avoidance regimens in multiple studies.226,232–234
654 Kidney Transplantation: Principles and Practice

Finally, Tan et al. reported their 4-year experience with There also remains significant interpatient variability in
induction therapy utilizing a single dose of alemtuzumab tacrolimus metabolism. Advances in pharmacogenomics
and two perioperative doses of methylprednisolone (day are beginning to improve our understanding of tacrolimus
−1 as premedication for alemtuzumab infusion and intra- metabolism in pediatric kidney transplant patients and
operatively before graft reperfusion) followed by tacrolimus may, one day, allow individualized drug dosing to optimize
monotherapy in 42 low immunologic risk pediatric living- efficacy while minimizing toxicity.243–245
donor kidney transplant recipients.235 A subset of these Given barriers to adherence with more frequent medica-
patients was also able to tolerate weaning of tacrolimus tion dosing, there has been growing interest in the devel-
to every-other-day dosing. They reported excellent graft opment of tacrolimus formulations to achieve once daily
survival (97.6% at 1 year and 85.4% at 2–4 years) and dosing. Several small studies have demonstrated the feasi-
low cumulative risk of acute cellular rejection (ACR; 0% bility of converting stable pediatric patients from twice daily
at 1 year and 4.8% at 4 years) using this approach. Fur- to once daily tacrolimus dosing using the prolonged release
thermore, they reported no episodes of antibody-mediated formulation (Advagraf).246,247 Conversion from twice a
rejection (AMR), CMV infection, BK nephropathy, or PTLD. day to once daily dosing using a 1:1 conversion of the total
Finally, this group previously reported increased eGFR and daily dose appears to result in decreased AUC.246–248 There-
a greater increase in height z-score at 1 year for 15 preado- fore pharmacokinetic monitoring may be warranted after
lescent children receiving tacrolimus monotherapy.236 A conversion. A limited sampling AUC model has been devel-
low incidence (7.7%) of posttransplant diabetes was also oped for the prolonged release formulation and validated in
reported.237 pediatric and adolescent patients.239 Due to the risk of sei-
zures, intravenous administration of tacrolimus is usually
Calcineurin Inhibitors avoided.
As mentioned earlier, more than 90% of pediatric kidney Tacrolimus is primarily metabolized by the liver via the
transplants reported by SRTR receive CNIs at the time CYP3A system. Drug interactions related to inducers or
of hospital discharge. Tacrolimus has for the most part inhibitors of CYP3A can be expected to affect drug levels.
replaced cyclosporine (CsA) in the US. Diarrhea, a common problem in children, can dramatically
Historically, dosing of CsA in children was complicated increase tacrolimus serum levels. Therefore tacrolimus
by variable gastrointestinal absorption of oil-based formu- drug levels should be monitored closely during diarrheal
lations and a higher rate of metabolism in young children, illness (and dosage adjusted accordingly) to avoid toxicity
necessitating higher mg/kg and more frequent dosing (up and rechecked as diarrhea improves to ensure continued
to thrice daily). The introduction of the microemulsion for- therapeutic levels.
mulations enhanced bioavailability and reduced variability The most common side effects seen in children treated
in serum drug levels. Age not only affects pharmacokinetics with long-term tacrolimus therapy include impaired glu-
of CsA, but there also seems to be an effect on pharmacody- cose tolerance, posttransplant diabetes, tremor, alopecia,
namics, with young children having increased sensitivity to and mild sleep disturbances. In contrast to CsA, tacrolimus
the immunosuppressive effects of CsA independently of drug rarely causes the cosmetic side effects mentioned earlier.
levels.238 The side effect profile of CsA in children is similar There are a few studies of protocols to minimize exposure to
to adults. The undesirable cosmetic effects of hypertricho- CNIs in pediatric kidney transplant recipients; most report
sis and gingival hypertrophy are particularly distressful for using alemtuzumab induction. As mentioned earlier, the
adolescent recipients, possibly contributing to dangerous Pittsburgh group reported excellent graft survival and func-
nonadherence. Hypercholesterolemia and hypertriglyceri- tion in 42 consecutive pediatric kidney transplant recipients
demia are also common. during a 4-year period with alemtuzumab induction (or
Due to the undesirable side effect profile and inferior effi- Thymoglobulin) and low-dose tacrolimus monotherapy.235
cacy of CsA to prevent rejection, tacrolimus has become Results of an NIH-sponsored multicenter single-arm pro-
the favored form of CNI in the US since the late 1990s. spective phase II trial of alemtuzumab induction followed
Initial tacrolimus dosing is 0.1 to 0.15 mg/kg divided into by tacrolimus withdrawal in pediatric recipients of living-
twice daily doses and further adjusted based on trough lev- donor kidney transplants are also anticipated.209
els that have demonstrated good correlation with overall
drug exposure. However, abbreviated sampling for area mTOR Inhibitors
under the concentration-time curve (AUC) can be helpful Inhibitors of the mammalian target of rapamycin (mTOR)
in cases where trough levels are low despite large daily dose have a side effect profile unique from CNIs, including a
or if concerns for toxicity arise in the setting of appropriate lower risk for neoplasm and nephrotoxicity, and therefore
trough levels.239,240 Early therapeutic tacrolimus trough offer an attractive alternative for protocols that seek to min-
thresholds vary between transplant centers. Although imize CNI exposure. As demonstrated in Fig. 37.4, mTOR
there is no consensus concerning optimal maintenance inhibitors are not commonly administered to pediatric kid-
tacrolimus levels, most centers wean target troughs ney transplant recipients in the US.
to reach nadir around 5 to 7 ng/mL over the first 3 to 4 Pharmacokinetics of sirolimus are different in prepuber-
months posttransplant. tal children, resulting in shorter drug half-life; therefore,
With the advent of generic/off-brand formulations, con- twice daily dosing is often required to achieve therapeutic
cerns have been raised about decreased bioavailability levels. Furthermore, the optimal target trough level has yet
compared with brand-name Prograf.241,242 Therefore pro- to be fully defined in children. The most common side effects
viders and families alike should be cognizant of the need reported for sirolimus in pediatric patients include recurrent
for close drug monitoring after any change in formulation. aphthous ulcers, impaired wound healing, hyperlipidemia,
37 • Kidney Transplantation in Children 655

and proteinuria. Adverse effects, especially aphthous a reduction in MMF dose. Therapeutic drug monitoring of
ulcers, are reportedly more significant at higher trough lev- MMF in children (via mycophenolic acid, or MPA levels) has
els (>9 ng/mL).249 There are concerns for mild testosterone been criticized due to high inter- and intraindividual vari-
deficiency during mTOR therapy; however, pubertal devel- ability.258 The metabolism of MMF may be enhanced by
opment does not appear to be affected in adolescent kidney corticosteroids, and the bioavailability can be decreased by
transplant patients.250 Furthermore, the antiproliferative concomitant CsA or antacids containing magnesium or alu-
and antiangiogenic actions of sirolimus have the potential minum hydroxides. Pharmacokinetics are reported to vary
to alter growth plate function and thereby impair linear based on age, with young children requiring higher doses to
growth in children. Inhibition of mTOR has also been dem- achieve comparable MPA AUC.259 Genetic variants in the
onstrated to disrupt intracellular signaling of the IGF-1 axis glucuronosyltransferase and multidrug resistance-associ-
in chondrocytes, which might cause interference with the ated protein 2 may also be predictive of interindividual vari-
effects of endogenous or exogenous growth hormone. How- ability in MPA exposure.260 Generic formulations of MMF
ever, retrospective investigations of the effect of sirolimus appear to have similar bioavailability to brand-name coun-
therapy on linear growth in pediatric kidney transplant terparts.261 There is no consensus about the therapeutic
recipients have resulted in conflicting conclusions.251,252 drug-monitoring approach for MMF in pediatric transplant
Initial use of mTOR inhibitors in pediatric transplant was recipients, however lower MPA exposure has been associated
in protocols to avoid long-term CNI exposure. We reported with the formation of de novo donor-specific antibodies.262
our single-center experience with conversion from tacroli- For patients receiving tacrolimus, MMF is typically
mus to sirolimus at 3 to 6 months posttransplant in stable, started at 300 to 450 mg/m2/dose twice daily, but some
low-risk pediatric kidney transplant recipients.253 Patients have advocated for higher doses (500 mg/m2 dosed twice
with stable serum creatinine levels and surveillance biopsy daily) for children under the age of 2 years.259 When used
without evidence of acute rejection, including borderline alone or in steroid-free regimens, MMF is often dosed at 600
histology, were eligible for conversion to sirolimus. Addi- mg/m2/dose.
tional exclusion criteria included a history of glomerular
disease as the etiology of ESRD, second renal transplant, and
biopsy-proven BK nephropathy. Actuarial 5-year graft sur-
Acute Rejection in Pediatric
vival was 91% for those patients that remained on sirolimus Transplantation
(78% of the patients undergoing conversion). Acute rejec-
tion occurred in 13% in the 12 months after conversion. With modern immunosuppressive therapy, the rates of
Given the side effect profile with high-dose mTOR inhibi- acute rejection episodes within the first 12 months for pedi-
tors, more recent protocols have used low-dose everolimus in atric kidney transplant in the US are reported at 11.2%
combination with low-dose CsA to minimize the side effects (2011–2014 era).8 Chronic allograft rejection is a leading
of both immunosuppressants and allow withdrawal of cor- cause of graft loss in children. As with adults, late episodes
ticosteroids.254 The use of everolimus with low-dose CsA in of acute rejection and multiple episodes of acute rejection
a steroid-free protocol appears to result in similar growth are associated with worse long-term allograft survival.
benefits compared with CNI-based steroid-free regimens.255 Therefore early identification and treatment of acute rejec-
Early results are encouraging in terms of preventing acute tion are paramount to improve long-term outcomes.
rejection with low-dose everolimus in combination with
low-dose CsA.254,256 However, hyperlipidemia persisted DIAGNOSIS OF ACUTE REJECTION
in some studies despite lower mTOR exposure.254 Rates of
infectious complications, notably BK viremia254 and CMV The diagnosis of acute rejection is not always straightfor-
disease257 appear lower compared with CNI-based regimens. ward in pediatric kidney transplant recipients. Small chil-
dren who have adult-sized allografts have a large renal
Antimetabolites reserve relative to their muscle mass. Therefore significant
MMF has largely replaced azathioprine in the US for pedi- renal injury may occur without a change in the serum cre-
atric kidney transplant adjunctive maintenance therapy. atinine, and a small increase in serum creatinine can rep-
More than 90% of pediatric kidney transplant patients in resent substantial renal dysfunction. Children with acute
the US are discharged on a regimen that includes MMF. rejection can present with low-grade fever and mild hyper-
As mentioned earlier, the incorporation of MMF into stan- tension, but they can also be asymptomatic.
dard immunosuppression regimens has facilitated the use The differential diagnosis for elevated serum creatinine
of lower corticosteroid and CNI doses after transplantation. in pediatric kidney transplant recipients is similar to that
MMF has also proven useful in steroid-free protocols, where in adults and includes hypovolemia, CNI toxicity, UTI, uri-
it is combined with tacrolimus or sirolimus. Furthermore, it nary obstruction, BK nephropathy, and acute rejection.
is used in combination with sirolimus and corticosteroids in Renal biopsy is the gold standard to diagnose rejection and
CNI-sparing protocols. is well tolerated by children under sedation. Diagnosis of
The most common, and troublesome, side effects dur- rejection based on biopsy has become the standard of care
ing MMF use in children include leukopenia and diarrhea. in children (so-called biopsy-proven rejection). Urinalysis
MMF dosage reduction is often first tried for these undesir- and urine culture, viral PCR (for polyoma BK, CMV, and
able effects. Enteric-coated tablets can also be tried for gas- EBV), and renal ultrasound studies should be performed in
trointestinal disturbance in older children and adolescents. a timely manner before biopsy. Intravenous crystalloid may
However, in small children, conversion to azathioprine is be required to eliminate prerenal azotemia as a possible fac-
often performed if diarrhea and/or leukopenia persist despite tor in raising the serum creatinine level.
656 Kidney Transplantation: Principles and Practice

The role of surveillance (i.e., protocol) biopsy in pediat- AMR and subsequent graft loss.269–271 A prospective study
ric kidney transplant is not well established.263 Innova- of donor-specific anti-HLA antibody (DSA) surveillance in
tive protocols of steroid or CNI minimization often rely on 215 pediatric kidney transplant patients found that 35% of
surveillance biopsies to survey for early pathology (i.e., patients developed DSA at median of 3 months posttransplant
subclinical rejection) that might benefit from intensified with a high prevalence (70%) class II DSA (45% directed
immunosuppression or for evidence of drug toxicity that against HLA-DQ alleles).271 Interestingly, DSA was more
might benefit from dose reduction. However, not all pediat- likely to resolve in patients with lower mean fluorescence
ric centers perform regularly scheduled surveillance biop- intensity (MFI) and earlier detection of DSA. These studies
sies, and their value in enhancing long-term outcomes in underscore the need to better predict which DSA detection
children receiving standard immunosuppressive regimens (e.g., MFI or ability to fix complement) warrants aggressive
remains unproven. treatment to prevent AMR and improve outcomes.
AMR in pediatrics can be treated with intravenous immu-
TREATMENT OF ACUTE REJECTION noglobulin (IVIG), plasmapheresis, or rituximab,272–274 but
the long-term efficacy of these approaches is unclear.275
Acute Cellular Rejection Successful use of the proteasome inhibitor, bortezomib
There are few controlled trials of children to guide the treat- (Velcade, Millennium Pharmaceuticals), for treatment of
ment of acute allograft rejection. As with adults, high-dose AMR has been reported for pediatric kidney transplant
(“pulse”) corticosteroids are the first-line treatment in chil- patients.276–279 Although these small, retrospective case
dren with suspected or biopsy-proven ACR. Intravenous series demonstrate that bortezomib therapy is relatively well
methylprednisolone succinate is typically administered at 10 tolerated and resulted in decreased DSA and stabilization of
to 15 mg/kg/dose for three consecutive doses, occasionally graft function in the short term, long-term outcomes remain
extended to five doses. A taper of oral corticosteroids back unclear. A case series of 10 consecutive kidney transplant
down to maintenance doses over 3 to 5 days is used by some, patients with humoral rejection (including several adoles-
but not all, pediatric transplant centers. It is not uncommon cent patients) who were administered bortezomib in com-
for the serum creatinine to slightly increase during the first bination with plasmapheresis and IVIG reported a trend
few days of steroid pulse therapy. In addition, the serum toward improved graft survival (6 of 10 patients with func-
creatinine may not return to baseline levels for several days tioning grafts at 18 months) compared with historical con-
after completion of the pulse, but typically a trend of decreas- trols who received rituximab with plasmapheresis and IVIG
ing creatinine is suggestive of a response to therapy. (1 of 10 with functioning graft at 18 months).280 Finally,
Severe rejection (i.e., Banff grade IA or higher) or steroid- use of antic5 convertase inhibition with eculizumab has
resistant rejection has historically been treated with lympho- been reported to be effective in small pediatric cases series
cyte depleting antibodies (e.g., Thymoglobulin). However, with AMR resistant to conventional therapy.281,282
there are limited data available on optimal dosing or outcomes Another emerging challenge in pediatric kidney trans-
after treatment for ACR. Alemtuzumab has also been reported plant is the identification of non-HLA AMR.283 Examples
to be effective in some cases of refractory cellular rejection.264 include antibodies directed against angiotensin II type 1
Research in the past decade has identified an additional receptor (AT1)284,285 and endothelial cell antigens, and
role for B cells during ACR, even in the absence of identi- major histocompatibility complex (MHC) class I-related
fiable AMR. B cells are postulated to contribute to cellular chain A (MICA).
rejection via antigen presentation, activation of dendritic
cells, and provision of costimulatory help to T cells. B cell
transcript signatures265 and dense B cell infiltrates in biop-
Long-Term Management
sies of pediatric kidney transplant recipients have been Posttransplant
associated with poor graft survival266 and are predictive
of graft loss.267 A prospective, randomized, open-label trial The success of kidney transplantation in children with
compared standard pulse steroids and Thymoglobulin to ESRD now results in a 10-year patient survival rate of 90%
anti-CD20 monoclonal antibody (rituximab) for treatment to 95%. Therefore the long-term management of these
of biopsy-proven ACR with B cell infiltrate in pediatric kid- patients is focused on maintaining quality of life and mini-
ney transplant patients.268 Patients receiving rituximab mizing significant long-term side effects of immunosuppres-
therapy had significantly increased recovery of graft func- sion. Optimal management of pediatric kidney transplant
tion and improvement in rejection scores on biopsy at 6 and recipients includes identifying and reducing the cardiovas-
12 months after treatment compared with those receiving cular and metabolic effects of long-term immunosuppres-
standard therapy, despite higher rejection grade and greater sive therapy, preventing infection and chronic rejection,
number of patients with humoral rejection randomized to ensuring good long-term quality of life, and managing a
the rituximab group. These findings suggest a role for char- smooth transition into adulthood.
acterizing infiltrating cells in certain cases of biopsy-proven
rejection to individualize therapy. HYPERTENSION AND CARDIOVASCULAR
Antibody-Mediated Rejection DISEASE
Surveillance monitoring of de novo donor-specific antibodies Children with ESRD have increased cardiovascular mor-
is not standardized among pediatric transplant centers. There tality relative to their peers, and cardiovascular dis-
are several retrospective studies suggesting that the develop- ease accounts for nearly 40% of deaths among pediatric
ment of de novo donor-specific antibodies is associated with ESRD patients.286,287 A high prevalence of traditional
37 • Kidney Transplantation in Children 657

cardiovascular risk factors, including hypertension (50%– changes and increased physical activity, is typically pre-
75% of uremic children) and hyperlipidemia (70%–90% ferred for young children with hyperlipidemia. Although
of children on dialysis), accompany ESRD in childhood. the use of 3-hydroxy-3-methylgultaryl-coenzyme A (HMG-
Although kidney transplantation leads to a dramatic CoA) reductase inhibitors (statins) is generally considered
improvement in renal function and elimination of many tra- safe and effective in children older than the age of 8 years,
ditional risk factors, cardiovascular disease still accounts for few data are available on long-term safety.301 Omega-3
more than one-third of deaths among patients who receive fatty acids have been reported to be effective at reducing
transplants before 21 years of age (Foster, 2011 #707).288 LDL levels in young kidney transplant recipients in small
Hypertension remains common in children posttrans- case series.302
plant and is likely due to a combination of rapid weight
gain, medication side effects (e.g., CNI and steroids), and INFECTIONS AFTER TRANSPLANTATION
renal injury. A multicenter retrospective study in the US
found that 68% of children with healthy weight and 80% Although modern immunosuppression regimens (CNI,
of overweight/obese children had uncontrolled blood pres- MMF, and steroids) have reduced the rate of acute rejec-
sure 1 year after kidney transplant.289 Approximately 63% tion, infectious complications have become more fre-
of pediatric kidney transplant patients in the Netherlands quent.303–305 In particular, pediatric transplant recipients
have been reported to have hypertension after 1 year, with are at increased risk for developing virus-related complica-
nearly half of the patients with uncontrolled hypertension tions compared with adults due to immunologic naiveté at
despite treatment.290 At 5 years, 55% of pediatric trans- the time of transplantation.
plant recipients remained on antihypertensive therapy in a
single-center study in Denmark.291 Viral Infections
In addition to the concerns for long-term cardiovascular Cytomegalovirus. CMV remains an important viral
risk associated with uncontrolled hypertension, retrospective pathogen after kidney transplant in children. According
analyses of pediatric kidney transplant recipients in the US to the 2015 SRTR report, 50% of US pediatric recipients
and the Netherlands suggest that systolic hypertension inde- of deceased-donor and 56% of living-donor transplants
pendently predicts poor long-term allograft survival.292 For between 2011 and 2015 were serologically naïve to CMV
example, hypertension at 5-years posttransplant was associ- at the time of transplant. The combination of a donor who
ated with inferior graft survival at 15 years (53% vs. 68%) was positive for CMV (D+) and a pediatric recipient who was
and 20 years (33% vs. 53%) compared with children with- serologically naïve to CMV (D−), which represents the high-
out hypertension.291 Furthermore, masked hypertension est risk for infection and disease posttransplant, occurred in
and nocturnal hypertension are common in pediatric kidney 29.4% of deceased-donor and 25.1% of living-donor trans-
transplant recipients, suggesting that the use of ambulatory plants.8 The development of improved diagnostic methods
blood pressure monitoring may be a beneficial tool to identify and effective prophylaxis has decreased the mortality previ-
patients with hidden cardiovascular risk factors.293–295 ously associated with posttransplant CMV disease, but mor-
There are limited data to provide guidance on optimal bidity remains high.
blood pressure targets or classes of antihypertensive medi- There are two approaches to the prevention of CMV dis-
cations to improve graft and patient survival in pediatrics. ease in pediatric kidney transplant patients. Because the
However, in a retrospective study of 298 pediatric kidney rate of high-risk donor/recipient CMV mismatch (D+/R−)
transplant recipients in the US, the use of angiotensin-con- is much higher in children compared with adult recipients,
verting enzyme inhibitors (ACE inhibitors) among patients many centers choose to prescribe prophylactic antiviral
receiving antihypertensive monotherapy at 5 years post- medication during the early posttransplant period. Another
transplant was associated with improved long-term graft approach is to use frequent monitoring for viral DNA
survival at 15 years (100%) compared with those receiving in blood with prompt preemptive treatment while CMV
non-ACE inhibitor therapy (44%).291 infection is subclinical in nature. There have been no ran-
Metabolic effects of corticosteroids and tacrolimus lead- domized controlled clinical trials in pediatric kidney trans-
ing to obesity and posttransplant diabetes are additional plantation to determine whether either of these approaches
risk factors for cardiovascular morbidity and mortality. is superior to the other. However, there is suggestion in ret-
Hypercholesterolemia and hypertriglyceridemia remain rospective analyses that prophylaxis results in lower viral
common in children with a kidney transplant and are load during CMV infection and improved graft function in
likely due to a combination of drug side effects296 (e.g., the long-term.306
CsA, mTOR inhibitors, glucocorticoids) and obesity.297,298 A typical approach to prophylaxis considers the risk of
Weight gain should be monitored closely after transplant CMV disease posttransplant based on donor and recipient
to identify children early who are at risk for obesity, as CMV-specific serologic testing. CMV-negative recipients
weight gain and increases in BMI in the first 6 months after of CMV-negative donor kidneys (D−/R−) have the lowest
transplant are likely to be persistent.299 Collaboration with risk (<5%) of developing CMV disease in the immediate
pediatric dieticians is important to educate patients and postoperative period and often do not require prophy-
their families about heart-healthy dietary habits, especially laxis. CMV positive recipients (R+) are at intermediate
because many of the severe dietary restrictions placed on risk and are recommended to receive prophylaxis. Finally,
children during ESRD are suddenly lifted after transplant. the highest risk group is that of CMV negative recipients
In addition, biannual screening for hypercholesterol- of CMV positive grafts (D+/R−). Prophylaxis is generally
emia, hypertriglyceridemia, and impaired glucose tolerance recommended for this highest risk group for at least 100
is suggested.300 Lifestyle modification, including dietary days, but some centers extend the period of prophylaxis to
658 Kidney Transplantation: Principles and Practice

6 months.307 It is important to note that CMV infection or Children, particularly those who were seronegative
disease can occur once prophylaxis is discontinued, and for EBV at the time of transplant, should be monitored
therefore monitoring for viral DNA is recommended for at with peripheral blood EBV viral PCR to detect increasing
least 6 months after discontinuation of prophylaxis. viral load that may lead to the development of PTLD. The
Valganciclovir (Valcyte, Genentech), a prodrug of gan- majority of patients will respond to a reduction in immu-
ciclovir with improved bioavailability, has largely replaced nosuppression, but sometimes anti-B cell antibodies (e.g.,
ganciclovir as the drug of choice for CMV prophylaxis and rituximab) and/or chemotherapy are required to control
treatment in pediatric kidney transplant recipients. There disease.310,311
are no randomized clinical trials in pediatric transplant
recipients to determine the appropriate dose needed to pre- Polyomavirus. BK virus infection (e.g., viremia) can occur
vent CMV. As such, most studies have compared AUC with early (within the first month) or late (several years) after
ganciclovir exposure rates shown to be effective at prevent- kidney transplant in children. Definitive diagnosis of BK
ing CMV disease in adult transplant recipients. Daily dosing nephropathy is obtained only by renal biopsy. The inci-
for prophylaxis is typically reported using either weight- dence of BK virus nephropathy (BKN) in pediatric kidney
based formulae (14–17 mg/kg, maximum 900 mg/dose) transplant recipients is estimated to be 4% to 5%.312,313
or using the manufacturer’s instructions which normal- Children with BKN are at greater risk for graft loss.
ize the adult dosing based on body surface area (BSA) and Prospective monitoring for BK viremia using PCR assays
estimated GFR (dose [mg] = 7 × BSA × eGFR).308 These to detect viral DNA with preemptive lowering of immuno-
two approaches result in large differences in dosing for the suppression has been suggested as an effective approach.314
smallest (BSA <0.7 m2) and youngest (<3 years of age) pedi- BK virus specific cellular immunity has been associated with
atric recipients. Therefore some centers recommend thera- viral clearance and underscores the approach of reducing T
peutic drug monitoring for young and small children. There cell suppression.315 Other options include treatment with
remains the need for validated AUC algorithms that utilize IVIG, ciprofloxacin, cidofovir, or leflunomide, although the
fewer blood samples in these young patients.309 One small efficacy of these are not proven in pediatrics.316,317
study suggested that the weight-based approach, despite
resulting in lower prescribed doses and lower ganciclovir Bacterial Infections
exposure (AUC0–24), was adequate in preventing CMV UTI is the most common infectious complication after kid-
viremia in pediatric solid-organ transplant recipients.309 ney transplant in children, occurring in 15% to 33% of
Valganciclovir dosing should also be adjusted for GFR under patients.303,318 According to the NAPRTCS registry, at
60 mL/min/1.73 m2. Duration of prophylactic therapy var- 1 year posttransplant, antibiotics for UTI prophylaxis are
ies from center to center with as little as 12 weeks to upwards prescribed for 55% of patients with renal dysplasia, 65% of
of 200 days have been reported in single-center studies and patients diagnosed with reflux nephropathy, and 68% of
retrospective analyses. Finally, there have been no random- those with obstructive uropathy. Recurrent UTI has been
ized controlled clinical trials in children to determine supe- associated with faster deterioration in graft function in
riority or inferiority of valganciclovir with other antiviral children.318,319
medications (acyclovir, valacyclovir, ganciclovir) for the Risk factors for febrile UTI include anatomic abnormali-
prevention of CMV infection or disease in pediatric kidney ties, dysfunctional bladder, presence of foreign material
transplant patients. Leukopenia and neutropenia are the (e.g., urinary catheter or stents), and baseline immunosup-
most common medication adverse effects of valganciclovir. pression. Parenteral antibiotics are indicated for febrile UTI
in pediatric transplant patients.
Epstein-Barr Virus. The cumulative incidence of PTLD by
5 years in pediatric transplant recipients in the US is 1.76% LOWER URINARY TRACT SYMPTOMS
overall. PTLD in children is often associated with EBV infec-
tion, and children who are immunologically naïve to EBV Even children with nonurologic causes of ESRD have
at the time of transplant are at highest risk of developing been reported to have lower urinary tract symptoms after
PTLD. The cumulative incidence at 5 years posttransplant transplant that may include urinary urgency, bladder
was 3.2% for EBV-negative compared with 0.7% for EBV- pain, incomplete bladder emptying, nighttime or daytime
positive recipients transplanted in the US between 2003 to incontinence, and UTI.320 Timed voiding may be helpful
2013. Of the pediatric transplants reported in the SRTR from for management of incontinence and dysfunctional void-
2011 to 2015, 42% of deceased-donor recipients and 56% of ing. For those with bladder obstruction, intermittent cath-
living-donor recipients were seronegative for EBV at the time eterization should be attempted to avoid graft damage and
of transplant. Furthermore, 36.5% of deceased-donor recipi- dysfunction.321 Anticholinergic therapy and intermittent
ents and 44.7% of living-donor recipients were EBV-negative catheterization should be continued as medical manage-
and received grafts from EBV-positive donors (D+/R−).8 ment of neurogenic bladder after transplantation.
PTLD, or its precursor, EBV-related lymphoid hyperpla-
sia, may present as adenotonsillar hypertrophy in chil- GROWTH AND BONE HEALTH
dren, or develop in the gastrointestinal tract (presenting as
chronic diarrhea), peripheral lymph nodes (presenting with Impaired linear growth in children with CKD is multifac-
lymphadenopathy or frank lymphoma), or within the renal torial and is mainly due to disturbances in the axis involv-
allograft leading to graft failure. Central nervous system ing growth hormone (GH), insulin-like growth factor (IGF)
involvement is also described and can be fatal. Therefore and IGF-binding protein. Despite satisfactory renal func-
prevention and early recognition of PTLD are essential. tion after transplant, spontaneous catch-up growth is often
37 • Kidney Transplantation in Children 659

insufficient. Abnormalities in mineral metabolism includ- psychological conflicts present during adolescence, trans-
ing disturbances in serum calcium, phosphorus, PTH, and plant recipients are exposed to additional psychosocial
vitamin D stores, are common in children after kidney stress related to their chronic illness. Developmental delay,
transplant and increase the risk for growth delay and osteo- issues with body image from drug side effects, difficulty
penia or rickets.322–324 interacting with peers, fastidious schedules required for
Determinants of growth after transplant include age immunosuppressant medication regimens, symptoms of
at the time of transplant, exposure to glucocorticoids, posttraumatic stress, and family disruption due to financial
allograft function, and administration of recombinant burden or role strain may all exacerbate psychosocial dif-
human growth hormone (rhGH). Children under the age of ficulties in the adolescent transplant recipient. Collabora-
6 years have increased growth rates after transplant com- tion with psychologists, psychiatrists, and social workers is
pared with older children325 Glucocorticoids induce hypo- important for the early identification and intensified treat-
responsiveness to the action of GH and prepubertal children ment of high-risk individuals.
on steroid-free immunosuppression protocols have been
demonstrated to have improved linear growth compared Puberty
with children receiving prednisone-based immunosuppres- Onset of puberty is an important milestone during adoles-
sion.326 At 30 days eGFR posttransplant is predictive of cence. There are few reports describing puberty and sexual
long-term height z-score, with those recipients who have maturity in children after kidney transplant, with most
an eGFR <60 mL/min/1.73 m2 having more negative focusing on pubertal growth velocity. The majority of chil-
height z-scores.327 The degree of growth stunting before dren achieve normal puberty after transplant, but many
transplant is also predictive of final adult height.328 Finally, may have a delayed onset and shortened duration of puber-
rhGH is effective in improving the growth velocity and final tal growth spurt.334 In general, onset of puberty is often
adult height of children after kidney transplant.329,330 delayed in children who have delayed bone age compared
with their chronologic age.335
NONADHERENCE IN PEDIATRIC Sexuality and Body Image
TRANSPLANTATION
Sexuality-related issues including prevention of sexually
Nonadherence (NA) is a major problem in pediatric kid- transmitted diseases and family planning also need to be
ney transplant. A systematic review estimated that NA addressed with teenage patients. Adolescent female trans-
accounts for 44% of graft losses and 23% of late acute rejec- plant recipients have successfully become pregnant while
tion episodes.187 Overall prevalence of NA across 16 stud- receiving CsA or tacrolimus. The effect of contraception on
ies involving pediatric kidney transplant patients was 31% the metabolism of immunosuppressant medications needs
(weighted mean). Adolescent patients were at higher risk to be considered when counseling adolescent girls about
with weighted mean prevalence of NA of 43% in the same pregnancy prevention. In addition, the teratogenicity of
review. certain immunosuppressant medications (notably MMF
The use of self-reporting of missed medication doses likely and CNIs) and antihypertensive medications (e.g., ACE
underrepresents the true incidence as patients and families inhibitors) should be explained to the adolescent female and
are often reluctant to report NA. Missed clinic appointments her family.
and scheduled tests are also common forms of NA reported Teenagers also need to balance their developing sexu-
for pediatric solid-organ transplant patients.331 Factors ality with changes in body image after transplant. It may
associated with NA include lack of supervision in taking be difficult for teenagers to accept the cosmetic side effects
medications, family conflicts, miscommunication between of immunosuppressant medications, including weight
parents and the medical team, numbers of medications, and gain, Cushingoid features, acne, and gum hypertrophy.
fastidious schedules of medication dosing.332,333 The accompanying psychological stress and effect on self-
image for teenagers can provide a dangerous disincentive
to adhere to medication regimens.
ADOLESCENCE AND EMERGING ADULTHOOD
Adolescence is an important transition period between Transitional Care
childhood and adulthood characterized by a quest for The term “emerging adulthood” is now commonly used
independence and autonomy. This rapidly changing and to define the interval of 18 to 25 years of age when young
volatile developmental period places adolescent transplant adults appear physically mature, yet brain maturation is
recipients at increased risk for medication NA, acute rejec- not yet completed. This age group is at increased risk of graft
tion episodes, and graft loss. Medical management of this failure likely due to a combination of age-related risk behav-
population can be challenging and benefits from special- ior and the processes involved in transitioning from pediat-
ized, multidisciplinary approaches to improve outcomes. ric-based to adult-based healthcare delivery systems.336
Several reports have demonstrated a high risk of graft
Psychosocial Development failure particularly at the time of transfer from pediatric
The adolescent striving to establish an identity indepen- to adult care.337,338 It is increasingly recognized for other
dent from parents and other adult authority figures can chronic conditions (e.g., survivors of pediatric cancer) that
result in oppositional and defiant behaviors. The need for adolescents and young adults (aged 14–29 years) are more
independence coupled with a sense of invulnerability and vulnerable to adverse outcomes and probably deserve a
evolving capacity for abstract thought can result in illogi- unique approach to health care delivery than that used
cal thinking and risk-taking behaviors. Beyond the normal for younger children or older adults.339 The identification
660 Kidney Transplantation: Principles and Practice

of strategies to facilitate safe transition to adult health care 5. MCDONALD SP. Australia and New Zealand Dialysis and Transplant
delivery systems and improve outcomes for adolescent Registry. Kidney Int Suppl 2015;5(2011):39–44.
6. Garcia C, Pestana JM, Martins S, Nogueira P, Barros V, Rohde R,
transplant recipients has become a priority for the pediatric Camargo M, Feltran L, Esmeraldo R, Carvalho R, Schvartsman B,
transplant community.336,340,341 Vaisbich M, Watanabe A, Cunha M, Meneses R, Prates L, Belangero
Transition is a process that includes a purposeful, V, Palma L, Carvalho D, Matuk T, Benini V, Laranjo S, Abbud-Filho
planned effort to prepare the patient to move from health M, Charpiot IM, Ramalho HJ, Lima E, Penido J, Andrade C, Gesteira
M, Tavares M, Penido M, De Souza V, Wagner M. Collaborative Bra-
care that is caregiver-directed to care that assumes disease zilian Pediatric Renal Transplant Registry (CoBrazPed-RTx): a report
self-management. Although the timing should be individu- from 2004 to 2013. Transplant Proc 2015;47:950–3.
alized, there is a growing consensus that preparation for 7. Harambat J, Van Stralen KJ, Kim JJ, Tizard EJ. Epidemiology of chronic
transition should begin early (between 10 and 14 years of kidney disease in children. Pediatr Nephrol 2012;27:363–73.
age). There is significant variability in the attainment of the 8. Hart A, Smith JM, Skeans MA, Gustafson SK, Stewart DE, Cherikh
WS, Wainright JL, Kucheryavaya A, Woodbury M, Snyder JJ,
skills needed to accomplish successful transition and there- Kasiske BL, Israni AK. Optn/Srtr 2015 Annual data report: kidney.
fore transfer to an adult unit may not be optimally decided Am J Transplant 2017;17(Suppl. 1):21–116.
based solely on a patient’s age. 9. Amaral S, Patzer R. Disparities, race/ethnicity and access to
Critical milestones for patients to achieve a successful trans- pediatric kidney transplantation. Curr Opin Nephrol Hypertens
2013;22:336–43.
fer to adult care include (1) the understanding and ability to 10. Agarwal S, Oak N, Siddique J, Harland RC, Abbo ED. Changes
describe the underlying cause of ESRD and need for trans- in pediatric renal transplantation after implementation of the
plant, (2) awareness of the long- and short-term implications revised deceased donor kidney allocation policy. Am J Transplant
of transplant on their overall health, (3) comprehension of 2009;9:1237–42.
the effect of their condition on their reproductive health, (4) 11. Harambat J, Van Stralen KJ, Schaefer F, Grenda R, Jankauskiene
A, Kostic M, Macher MA, Maxwell H, Puretic Z, Raes A, Rubik J,
demonstration of a sense of responsibility for their own health Sorensen SS, Toots U, Topaloglu R, Tonshoff B, Verrina E, Jager KJ.
care, (5) capacity to provide most care independently, and (6) Disparities in policies, practices and rates of pediatric kidney trans-
an expressed readiness to move into adulthood.342,343 plantation in Europe. Am J Transplant 2013;13:2066–74.
The application of transitional care has been described in 12. Keith DS, Vranic G, Barcia J, Norwood V, Nishio-Lucar A. Longitudi-
nal analysis of living donor kidney transplant rates in pediatric can-
a variety of forms ranging from having adult providers meet didates in the United States. Pediatr Transplant 2017;21.
the patient transfer, having a pediatric team member accom- 13. Parker WF, Thistlethwaite Jr JR, Ross LF. Kidney donor profile index
pany the patient to their first adult clinic visit, overlapping/ does not accurately predict the graft survival of pediatric deceased
alternating visits between pediatric and adult teams, to fully donor kidneys. Transplantation 2016;100:2471–8.
shared adolescent/young adult transplant clinics. Mecha- 14. Vitola SP, Gnatta D, Garcia VD, Garcia CD, Bittencourt VB, Keitel E,
Pires FS, D’avila AR, Silva JG, Amaral RL, Santos LN, Kruel CD. Kid-
nisms to monitor and assess the readiness for transfer have ney transplantation in children weighing less than 15 kg: extraperi-
been described but are not standardized. Furthermore, there toneal surgical access-experience with 62 cases. Pediatr Transplant
are still significant systems issues (insurance coverage, 2013;17:445–53.
health care policies, resource allocations) to overcome. 15. Herthelius M, Celsi G, Edstrom Halling S, Krmar RT, Sandberg J,
Tyden G, Asling-Monemi K, Berg UB. Renal transplantation in
There is a need for longitudinal analyses of the outcomes infants and small children. Pediatr Nephrol 2012;27:145–50.
of children who are transitioned to adult care. A great 16. Sarwal MM, Cecka JM, Millan MT, Salvatierra JR O,. Adult-size kid-
opportunity exists to form collaborative efforts between neys without acute tubular necrosis provide exceedingly superior
pediatric and adult transplant communities to improve out- long-term graft outcomes for infants and small children: a single
comes for this high-risk population. center and UNOS analysis. United Network for Organ Sharing.
Transplantation 2000;70:1728–36.
17. Vats AN, Donaldson L, Fine RN, Chavers BM. Pretransplant dialy-
References sis status and outcome of renal transplantation in North American
1. 2017. United States Renal Data System. USRDS Annual data report: children: a NAPRTCS Study. North American Pediatric Renal Trans-
epidemiology of kidney disease in the United States. Bethesda, MD: plant Cooperative Study. Transplantation 2000;69:1414–9.
National Institutes of Health, National Institure of Diabetes and 18. Samuel SM, Tonelli MA, Foster BJ, Alexander RT, Nettel-Aguirre A,
Digestive Kidney Diseases; 2017. Soo A, Hemmelgarn BR. Survival in pediatric dialysis and transplant
2. Smith JM, Martz K, Blydt-Hansen TD. Pediatric kidney transplant patients. Clin J Am Soc Nephrol 2011c;6:1094–9.
practice patterns and outcome benchmarks, 1987-2010: a report of 19. Lofaro D, Jager KJ, Abu-Hanna A, Groothoff JW, Arikoski P, Hoecker
the North American Pediatric Renal Trials and Collaborative Stud- B, Roussey-Kesler G, Spasojevic B, Verrina E, Schaefer F, Van Stralen
ies. Pediatr Transplant 2013;17:149–57. KJ, Registry EE-E. Identification of subgroups by risk of graft failure
3. Samuel SM, Tonelli MA, Foster BJ, Nettel-Aguirre A, Na Y, Williams after paediatric renal transplantation: application of survival tree
R, Soo A, Hemmelgarn BR. Overview of the Canadian pediatric end- models on the ESPN/ERA-EDTA Registry. Nephrol Dial Transplant
stage renal disease database. BMC Nephrol 2010;11:21. 2016;31:317–24.
4. Pippias M, Kramer A, Noordzij M, Afentakis N, Alonso De La Torre R, 20. Butani L, Perez RV. Effect of pretransplant dialysis modality and
Ambuhl PM, Aparicio Madre MI, Arribas Monzon F, Asberg A, Bon- duration on long-term outcomes of children receiving renal trans-
thuis M, Bouzas Caamano E, Bubic I, Caskey FJ, Castro De La Nuez plants. Transplantation 2011;91:447–51.
P, Cernevskis H, De Los Angeles Garcia Bazaga M, Des Grottes JM, 21. Kramer A, Stel VS, Geskus RB, Tizard EJ, Verrina E, Schaefer F, Heaf
Fernandez Gonzalez R, Ferrer-Alamar M, Finne P, Garneata L, Golan JG, Kramar R, Krischock L, Leivestad T, Palsson R, Ravani P, Jager
E, Heaf JG, Hemmelder MH, Idrizi A, Ioannou K, Jarraya F, Kantaria KJ. The effect of timing of the first kidney transplantation on sur-
N, Kolesnyk M, Kramar R, Lassalle M, Lezaic VV, Lopot F, Macario vival in children initiating renal replacement therapy. Nephrol Dial
F, Magaz A, Martin De Francisco AL, Martin Escobar E, Martinez Transplant 2012;27:1256–64.
Castelao A, Metcalfe W, Moreno Alia I, Nordio M, Ots-Rosenberg M, 22. Gillen DL, Stehman-Breen CO, Smith JM, Mcdonald RA, Warady BA,
Palsson R, Ratkovic M, Resic H, Rutkowski B, Santiuste De Pablos C, Brandt JR, Wong CS. Survival advantage of pediatric recipients of a
Seyahi N, Fernanda Slon Roblero M, Spustova V, Stas KJF, Stendahl first kidney transplant among children awaiting kidney transplanta-
ME, Stojceva-Taneva O, Vazelov E, Ziginskiene E, Massy Z, Jager KJ, tion. Am J Transplant 2008;8:2600–6.
Stel VS. The European Renal Association - European Dialysis and 23. Hwang AH, Cho YW, Cicciarelli J, Mentser M, Iwaki Y, Hardy BE.
Transplant Association Registry Annual Report 2014: a summary. Risk factors for short- and long-term survival of primary cadaveric
Clin Kidney J 2017;10:154–69. renal allografts in pediatric recipients: a UNOS analysis. Transplan-
tation 2005;80:466–70.
37 • Kidney Transplantation in Children 661

24. Pitt SC, Vachharajani N, Doyle MB, Lowell JA, Chapman WC, Ander- 45. Choi JY, Kwon OJ, Kang CM. The Effect of Donor-Recipient relation-
son CD, Shenoy S, Wellen JR. Organ allocation in pediatric renal trans- ship on long-term outcomes of living related donor renal transplan-
plants: is there an optimal donor? Clin Transplant 2013;27:938–44. tation. Transplant Proc 2012;44:257–60.
25. Van Arendonk KJ, Orandi BJ, James NT, Segev DL, Colombani PM. 46. Ghoneim MA, Refaie AF. Is matching for human leukocyte antigen-
Living unrelated renal transplantation: a good match for the pediat- DR beneficial in pediatric kidney transplantation? Nat Clin Pract
ric candidate? J Pediatr Surg 2013b;48:1277–82. Nephrol 2009;5:70–1.
26. Papachristou F, Stabouli S, Printza N, Mitsioni A, Stefanidis C, Miser- 47. Gritsch HA, Veale JL, Leichtman AB, Guidinger MK, Magee JC,
lis G, Dotis J, Kapogiannis A, Georgaki-Angelaki H, Gkogka C, Kollios Mcdonald RA, Harmon WE, Delmonico FL, Ettenger RB, Cecka JM.
K, Papanikolaou V. Long-term outcome of pediatric kidney trans- Should pediatric patients wait for HLA-DR-matched renal trans-
plantation: a single-center experience from Greece. Pediatr Trans- plants? Am J Transplant 2008;8:2056–61.
plant 2016;20:500–6. 48. Opelz G, Dohler B. Impact of HLA mismatching on incidence of post-
27. Foster BJ, Dahhou M, Zhang X, Platt RW, Hanley JA. Relative impor- transplant non-hodgkin lymphoma after kidney transplantation.
tance of HLA mismatch and donor age to graft survival in young kid- Transplantation 2010a;89:567–72.
ney transplant recipients. Transplantation 2013;96:469–75. 49. Foster BJ, Dahhou M, Zhang X, Platt RW, Smith JM, Hanley JA. Impact
28. Van Arendonk KJ, James NT, Locke JE, Montgomery RA, Colombani of HLA mismatch at first kidney transplant on lifetime with graft func-
PM, Segev DL. Late graft loss among pediatric recipients of DCD kid- tion in young recipients. Am J Transplant 2014;14:876–85.
neys. Clin J Am Soc Nephrol 2011;6:2705–11. 50. Gralla J, Tong S, Wiseman AC. The impact of human leukocyte
29. Moudgil A, Dharnidharka VR, Lamb KE, Meier-Kriesche HU. Best antigen mismatching on sensitization rates and subsequent retrans-
allograft survival from share-35 kidney donors occurs in middle- plantation after first graft failure in pediatric renal transplant recipi-
aged adults and young children-an analysis of OPTN data. Trans- ents. Transplantation 2013;95:1218–24.
plantation 2013;95:319–25. 51. Keller AK, Jorgensen TM, Jespersen B. Identification of risk factors
30. Bobanga ID, Vogt BA, Woodside KJ, Cote DR, Dell KM, Cunning- for vascular thrombosis may reduce early renal graft loss: a review of
ham 3rd RJ, Noon KA, Barksdale EM, Humphreville VR, Sanchez recent literature. J Transplant 2012;2012:793461.
EQ, Schulak JA. Outcome differences between young children and 52. Mendley SR, Zelko FA. Improvement in specific aspects of neurocog-
adolescents undergoing kidney transplantation. J Pediatr Surg nitive performance in children after renal transplantation. Kidney
2015;50:996–9. Int 1999;56:318–23.
31. Van Arendonk KJ, James NT, Boyarsky BJ, Garonzik-Wang JM, 53. Fuentes GM, Meseguer CG, Carrion AP, Hijosa MM, Garcia-Pose
Orandi BJ, Magee JC, Smith JM, Colombani PM, Segev DL. Age at A, Melgar AA, Torres MN. Long-term outcome of focal segmental
graft loss after pediatric kidney transplantation: exploring the high- glomerulosclerosis after pediatric renal transplantation. Pediatr
risk age window. Clin J Am Soc Nephrol 2013a;8:1019–26. Nephrol 2010;25:529–34.
32. Van Arendonk KJ, King EA, Orandi BJ, James NT, Smith JM, Colom- 54. Hickson LJ, Gera M, Amer H, Iqbal CW, Moore TB, Milliner DS, Cosio
bani PM, Magee JC, Segev DL. Loss of pediatric kidney grafts during FG, Larson TS, Stegall MD, Ishitani MB, Gloor JM, Griffin MD. Kid-
the “high-risk age window”: insights from pediatric liver and simul- ney transplantation for primary focal segmental glomerulosclerosis:
taneous liver-kidney recipients. Am J Transplant 2015;15:445–52. outcomes and response to therapy for recurrence. Transplantation
33. Heidotting NA, Ahlenstiel T, Kreuzer M, Franke D, Pape L. The 2009;87:1232–9.
influence of low donor age, living related donation and pre-emptive 55. Lee SE, Min SI, Kim YS, Ha J, Ha IS, Cheong HI, Kim SJ, Choi Y, Kang
transplantation on end-organ damage based on arterial hyperten- HG. Recurrence of idiopathic focal segmental glomerulosclerosis
sion after paediatric kidney transplantation. Nephrol Dial Trans- after kidney transplantation: experience of a Korean tertiary center.
plant 2012;27:1672–6. Pediatr Transplant 2014;18:369–76.
34. Opelz G, Dohler B. Pediatric kidney transplantation: analysis of donor 56. Nehus EJ, Goebel JW, Succop PS, Abraham EC. Focal segmental
age, HLA match, and posttransplant non-Hodgkin lymphoma: a col- glomerulosclerosis in children: multivariate analysis indicates
laborative transplant study report. Transplantation 2010b;90:292–7. that donor type does not alter recurrence risk. Transplantation
35. Dubourg L, Cochat P, Hadj-Aissa A, Tyden G, Berg UB. Better long- 2013;96:550–4.
term functional adaptation to the child’s size with pediatric com- 57. Ding WY, Koziell A, Mccarthy HJ, Bierzynska A, Bhagavatula MK,
pared to adult kidney donors. Kidney Int 2002;62:1454–60. Dudley JA, Inward CD, Coward RJ, Tizard J, Reid C, Antignac C,
36. Melk A, Schmidt BM, Braun H, Vongwiwatana A, Urmson J, Zhu LF, Boyer O, Saleem MA. Initial steroid sensitivity in children with ste-
Rayner D, Halloran PF. Effects of donor age and cell senescence on roid-resistant nephrotic syndrome predicts post-transplant recur-
kidney allograft survival. Am J Transplant 2009;9:114–23. rence. J Am Soc Nephrol 2014;25:1342–8.
37. Moudgil A, Martz K, Stablein DM, Puliyanda DP. Good outcome of 58. Abbott KC, Sawyers ES, Oliver Iii JD, Ko CW, Kirk AD, Welch PG,
kidney transplants in recipients of young donors: a NAPRTCS data Peters TG, Agodoa LY. Graft loss due to recurrent focal segmental
analysis. Pediatr Transplant 2011;15:167–71. glomerulosclerosis in renal transplant recipients in the united states.
38. Sui M, Zhao W, Chen Y, Zhu F, Zhu Y, Zeng L, Zhang L. Optimiz- Am J Kidney Dis 2001;37:366–73.
ing the utilization of kidneys from small pediatric deceased donors 59. Baum MA, Stablein DM, Panzarino VM, Tejani A, Harmon WE,
under 15 kg by choosing pediatric recipients. Pediatr Transplant Alexander SR. Loss of living donor renal allograft survival advan-
2016;20:39–43. tage in children with focal segmental glomerulosclerosis. Kidney Int
39. Guan I, Singer P, Frank R, Chorny N, Infante L, Sethna CB. Role of 2001;59:328–33.
race in kidney transplant outcomes in children with focal segmental 60. Baum MA. Outcomes after renal transplantation for FSGS in chil-
glomerulosclerosis. Pediatr Transplant 2016;20:790–7. dren. Pediatr Transplant 2004;8:329–33.
40. Patzer RE, Mohan S, Kutner N, Mcclellan WM, Amaral S. Racial and 61. Gonzalez E, Ettenger R, Rianthavorn P, Tsai E, Malekzadeh M. Pre-
ethnic disparities in pediatric renal allograft survival in the United emptive plasmapheresis and recurrence of focal segmental glomeru-
States. Kidney Int 2015;87:584–92. losclerosis in pediatric renal transplantation. Pediatr Transplant
41. Patzer RE, Sayed BA, Kutner N, Mcclellan WM, Amaral S. Racial and 2011b;15:495–501.
ethnic differences in pediatric access to preemptive kidney transplan- 62. Canaud G, Dion D, Zuber J, Gubler MC, Sberro R, Thervet E, Sna-
tation in the United States. Am J Transplant 2013;13:1769–81. noudj R, Charbit M, Salomon R, Martinez F, Legendre C, Noel LH,
42. Omoloja A, Mitsnefes M, Talley L, Benfield M, Neu A. Racial differ- Niaudet P. Recurrence of nephrotic syndrome after transplantation
ences in graft survival: a report from the North American Pediatric in a mixed population of children and adults: course of glomerular
Renal Trials and Collaborative Studies (NAPRTCS). Clin J Am Soc lesions and value of the Columbia classification of histological vari-
Nephrol 2007;2:524–8. ants of focal and segmental glomerulosclerosis (FSGS). Nephrol Dial
43. Foster BJ, Dahhou M, Zhang X, Platt RW, Hanley JA. Relative impor- Transplant 2010;25:1321–8.
tance of HLA mismatch and donor age to graft survival in young kid- 63. Ponticelli C. Recurrence of focal segmental glomerular sclero-
ney transplant recipients. Transplantation 2013;96:469–75. sis (FSGS) after renal transplantation. Nephrol Dial Transplant
44. Tejani A, Sullivan EK. Do six-antigen-matched cadaver donor kid- 2010;25:25–31.
neys provide better graft survival to children compared with one- 64. Weber S, Gribouval O, Esquivel EL, Moriniere V, Tete MJ, Legendre
haploidentical living-related donor transplants? A report of the C, Niaudet P, Antignac C. NPHS2 mutation analysis shows genetic
North American Pediatric Renal Transplant Cooperative Study. heterogeneity of steroid-resistant nephrotic syndrome and low post-
Pediatr Transplant 2000;4:140–5. transplant recurrence. Kidney Int 2004;66:571–9.
662 Kidney Transplantation: Principles and Practice

65. Jungraithmayr TC, Hofer K, Cochat P, Chernin G, Cortina G, Fargue 81. Wang CS, Liverman RS, Garro R, George RP, Glumova A, Karp A,
S, Grimm P, Knueppel T, Kowarsch A, Neuhaus T, Pagel P, Pfeiffer Jernigan S, Warshaw B. Ofatumumab for the treatment of childhood
KP, Schafer F, Schonermarck U, Seeman T, Toenshoff B, Weber S, nephrotic syndrome. Pediatr Nephrol 2017;32:835–41.
Winn MP, Zschocke J, Zimmerhackl LB. Screening for NPHS2 muta- 82. Benoit G, Machuca E, Antignac C. Hereditary nephrotic syndrome:
tions may help predict FSGS recurrence after transplantation. J Am a systematic approach for genetic testing and a review of associated
Soc Nephrol 2011;22:579–85. podocyte gene mutations. Pediatr Nephrol 2010;25:1621–32.
66. Becker-Cohen R, Bruschi M, Rinat C, Feinstein S, Zennaro C, Ghig- 83. Patrakka J, Ruotsalainen V, Reponen P, Qvist E, Laine J, Holmberg
geri GM, Frishberg Y. Recurrent nephrotic syndrome in homozygous C, Tryggvason K, Jalanko H. Recurrence of nephrotic syndrome in
truncating NPHS2 mutation is not due to anti-podocin antibodies. kidney grafts of patients with congenital nephrotic syndrome of the
Am J Transplant 2007;7:256–60. Finnish type: role of nephrin. Transplantation 2002;73:394–403.
67. Cattran D, Neogi T, Sharma R, Mccarthy ET, Savin VJ, Group 84. Kim MS, Stablein D, Harmon WE. Renal transplantation in children
FTNANS. Serial estimates of serum permeability activity and clinical with congenital nephrotic syndrome: a report of the North American
correlates in patients with native kidney focal segmental glomerulo- Pediatric Renal Transplant Cooperative Study (NAPRTCS). Pediatr
sclerosis. J Am Soc Nephrol 2003;14:448–53. Transplant 1998;2:305–8.
68. Wei C, El Hindi S, Li J, Fornoni A, Goes N, Sageshima J, Maiguel D, 85. Ito S, Takata A, Hataya H, Ikeda M, Kikuchi H, Hata J, Honda M. Iso-
Karumanchi SA, Yap HK, Saleem M, Zhang Q, Nikolic B, Chaudhuri lated diffuse mesangial sclerosis and Wilms tumor suppressor gene. J
A, Daftarian P, Salido E, Torres A, Salifu M, Sarwal MM, Schaefer Pediatr 2001;138:425–7.
F, Morath C, Schwenger V, Zeier M, Gupta V, Roth D, Rastaldi MP, 86. Neuhaus TJ, Arnold W, Gaspert A, Hopfer H, Fischer A. Recurrence
Burke G, Ruiz P, Reiser J. Circulating urokinase receptor as a cause of membranoproliferative glomerulonephritis after renal transplan-
of focal segmental glomerulosclerosis. Nat Med 2011;17:952–60. tation in Denys-Drash. Pediatr Nephrol 2011;26:317–22.
69. Staeck O, Slowinski T, Lieker I, Wu K, Rudolph B, Schmidt D, 87. Nso Roca AP, Pena Carrion A, Benito Gutierrez M, Garcia Meseguer
Brakemeier S, Neumayer HH, Wei C, Reiser J, Budde K, Halleck F, C, Garcia Pose A, Navarro M. Evolutive study of children with diffuse
Khadzhynov D. Recurrent primary focal segmental glomeruloscle- mesangial sclerosis. Pediatr Nephrol 2009;24:1013–9.
rosis managed with intensified plasma exchange and concomitant 88. Gumber MR, Kute VB, Goplani KR, Vanikar AV, Shah PR, Patel HV,
monitoring of soluble urokinase-type plasminogen activator recep- Trivedi HL. Outcome of renal transplantation in Alport’s syndrome:
tor-mediated podocyte beta3-integrin activation. Transplantation a single-center experience. Transplant Proc 2012;44:261–3.
2015;99:2593–7. 89. Kruegel J, Rubel D, Gross O. Alport syndrome-insights from basic
70. Straatmann C, Kallash M, Killackey M, Iorember F, Aviles D, and clinical research. Nat Rev Nephrol 2012.
Bamgbola O, Carson T, Florman S, Vehaskari MV. Success with 90. Gross O, Weber M, Fries JW, Muller GA. Living donor kidney trans-
plasmapheresis treatment for recurrent focal segmental glomerulo- plantation from relatives with mild urinary abnormalities in Alport
sclerosis in pediatric renal transplant recipients. Pediatr Transplant syndrome: long-term risk, benefit and outcome. Nephrol Dial Trans-
2014;18:29–34. plant 2009;24:1626–30.
71. Gohh RY, Yango AF, Morrissey PE, Monaco AP, Gautam A, Sharma 91. Rheault MN. Women and Alport syndrome. Pediatr Nephrol
M, Mccarthy ET, Savin VJ. Preemptive plasmapheresis and recur- 2012;27:41–6.
rence of FSGS in high-risk renal transplant recipients. Am J Trans- 92. Byrne MC, Budisavljevic MN, Fan Z, Self SE, Ploth DW. Renal
plant 2005;5:2907–12. transplant in patients with Alport’s syndrome. Am J Kidney Dis
72. Shishido S, Satou H, Muramatsu M, Hamasaki Y, Ishikura K, 2002;39:769–75.
Hataya H, Honda M, Asanuma H, Aikawa A. Combination of pulse 93. De Sandes-Freitas TV, Holanda-Cavalcanti A, Mastroianni-Kirsz-
methylprednisolone infusions with cyclosporine-based immuno- tajn G, Franco MF, Medina-Pestana JO. Late presentation of Alport
suppression is safe and effective to treat recurrent focal segmen- posttransplantation anti-glomerular basement membrane disease.
tal glomerulosclerosis after pediatric kidney transplantation. Clin Transplant Proc 2011;43:4000–1.
Transplant 2013;27:E143–50. 94. Charytan D, Torre A, Khurana M, Nicastri A, Stillman IE, Kalluri R.
73. Raafat RH, Kalia A, Travis LB, Diven SC. High-dose oral cyclosporin Allograft rejection and glomerular basement membrane antibodies
therapy for recurrent focal segmental glomerulosclerosis in children. in Alport’s syndrome. J Nephrol 2004;17:431–5.
Am J Kidney Dis 2004;44:50–6. 95. Wang XP, Fogo AB, Colon S, Giannico G, Abul-Ezz SR, Miner JH,
74. Salomon R, Gagnadoux MF, Niaudet P. Intravenous cyclosporine Borza DB. Distinct epitopes for anti-glomerular basement membrane
therapy in recurrent nephrotic syndrome after renal transplantation alport alloantibodies and goodpasture autoantibodies within the
in children. Transplantation 2003;75:810–4. noncollagenous domain of alpha3(IV) collagen: a janus-faced anti-
75. Cheong HI, Han HW, Park HW, Ha IS, Han KS, Lee HS, Kim SJ, Choi gen. J Am Soc Nephrol 2005;16:3563–71.
Y. Early recurrent nephrotic syndrome after renal transplantation 96. Browne G, Brown PA, Tomson CR, Fleming S, Allen A, Herriot R,
in children with focal segmental glomerulosclerosis. Nephrol Dial Pusey CD, Rees AJ, Turner AN. Retransplantation in Alport post-
Transplant 2000;15:78–81. transplant anti-GBM disease. Kidney Int 2004;65:675–81.
76. Hubsch H, Montane B, Abitbol C, Chandar J, Shariatmadar S, Cian- 97. Angelo JR, Bell CS, Braun MC. Allograft failure in kidney transplant
cio G, Burke G, Miller J, Strauss J, Zilleruelo G. Recurrent focal glo- recipients with membranoproliferative glomerulonephritis. Am J
merulosclerosis in pediatric renal allografts: the Miami experience. Kidney Dis 2011;57:291–9.
Pediatr Nephrol 2005;20:210–6. 98. Lorenz EC, Sethi S, Leung N, Dispenzieri A, Fervenza FC, Cosio FG.
77. Chaudhuri A, Kambham N, Sutherland S, Grimm P, Alexander Recurrent membranoproliferative glomerulonephritis after kidney
S, Concepcion W, Sarwal M, Wong C. Rituximab treatment for transplantation. Kidney Int 2010;77:721–8.
recurrence of nephrotic syndrome in a pediatric patient after renal 99. Moroni G, Casati C, Quaglini S, Gallelli B, Banfi G, Montagnino
transplantation for congenital nephrotic syndrome of Finnish type. G, Messa P. Membranoproliferative glomerulonephritis type I in
Pediatr Transplant 2012;16:E183–7. renal transplantation patients: a single-center study of a cohort
78. Chikamoto H, Hattori M, Kuroda N, Kajiho Y, Matsumura H, Fujii H, of 68 renal transplants followed up for 11 years. Transplantation
Ishizuka K, Hisano M, Akioka Y, Nozu K, Kaito H, Shimizu M. Pre- 2011;91:1233–9.
transplantation combined therapy with plasmapheresis and ritux- 100. Braun MC, Stablein DM, Hamiwka LA, Bell L, Bartosh SM, Strife CF.
imab in a second living-related kidney transplant pediatric recipient Recurrence of membranoproliferative glomerulonephritis type II in
with a very high risk for focal segmental glomerulosclerosis recur- renal allografts: The North American Pediatric Renal Transplant
rence. Pediatr Transplant 2012;16:E286–90. Cooperative Study experience. J Am Soc Nephrol 2005;16:2225–
79. Kumar J, Shatat IF, Skversky AL, Woroniecki RP, Del Rio M, Perel- 33.
stein EM, Johnson VL, Mahesh S. Rituximab in post-transplant pedi- 101. Muller T, Sikora P, Offner G, Hoyer PF, Brodehl J. Recurrence of renal
atric recurrent focal segmental glomerulosclerosis. Pediatr Nephrol disease after kidney transplantation in children: 24 years of experi-
2013;28:333–8. ence in a single center. Clin Nephrol 1998;49:82–90.
80. Pescovitz MD, Book BK, Sidner RA. Resolution of recurrent focal seg- 102. Zand L, Lorenz EC, Cosio FG, Fervenza FC, Nasr SH, Gandhi MJ, Smith
mental glomerulosclerosis proteinuria after rituximab treatment. N RJ, Sethi S. Clinical findings, pathology, and outcomes of C3GN after
Engl J Med 2006;354:1961–3. kidney transplantation. J Am Soc Nephrol 2014;25:1110–7.
37 • Kidney Transplantation in Children 663

103. Haffner K, Michelfelder S, Pohl M. Successful therapy of C3Nef-pos- 122. Loirat C, Fakhouri F, Ariceta G, Besbas N, Bitzan M, Bjerre A, Coppo R,
itive C3 glomerulopathy with plasma therapy and immunosuppres- Emma F, Johnson S, Karpman D, Landau D, Langman CB, Lapeyraque
sion. Pediatr Nephrol 2015;30:1951–9. AL, Licht C, Nester C, Pecoraro C, Riedl M, Van De Kar NC, Van De
104. Perez-Saez MJ, Toledo K, Navarro MD, Lopez-Andreu M, Redondo Walle J, Vivarelli M, Fremeaux-Bacchi V, International HUS. An inter-
MD, Ortega R, Perez-Seoane C, Aguera ML, Rodriguez-Benot A, national consensus approach to the management of atypical hemo-
Aljama P. Recurrent membranoproliferative glomerulonephritis lytic uremic syndrome in children. Pediatr Nephrol 2016;31:15–39.
after second renal graft treated with plasmapheresis and rituximab. 123. Licht C, Ardissino G, Ariceta G, Cohen D, Cole JA, Gasteyger C,
Transplant Proc 2011;43:4005–9. Greenbaum LA, Johnson S, Ogawa M, Schaefer F, Vande Walle J,
105. Sanchez-Moreno A, De La Cerda F, Cabrera R, Fijo J, Lopez-Trascasa Fremeaux-Bacchi V. The global aHUS registry: methodology and ini-
M, Bedoya R, Rodriguez De Cordoba S, Ybot-Gonzalez P. Eculizumab tial patient characteristics. BMC Nephrol 2015a;16:207.
in dense-deposit disease after renal transplantation. Pediatr Nephrol 124. Bresin E, Daina E, Noris M, Castelletti F, Stefanov R, Hill P, Goodship
2014;29:2055–9. TH, Remuzzi G. Outcome of renal transplantation in patients with non-
106. Gurkan S, Fyfe B, Weiss L, Xiao X, Zhang Y, Smith RJ. Eculizumab and Shiga toxin-associated hemolytic uremic syndrome: prognostic signif-
recurrent C3 glomerulonephritis. Pediatr Nephrol 2013;28:1975– icance of genetic background. Clin J Am Soc Nephrol 2006;1:88–99.
81. 125. Loirat C, Noris M, Fremeaux-Bacchi V. Complement and the
107. Ortiz F, Gelpi R, Koskinen P, Manonelles A, Raisanen-Sokolowski A, atypical hemolytic uremic syndrome in children. Pediatr Nephrol
Carrera M, Honkanen E, Grinyo JM, Cruzado JM. IgA nephropathy 2008;23:1957–72.
recurs early in the graft when assessed by protocol biopsy. Nephrol 126. Noris M, Caprioli J, Bresin E, Mossali C, Pianetti G, Gamba S, Daina
Dial Transplant 2012;27:2553–8. E, Fenili C, Castelletti F, Sorosina A, Piras R, Donadelli R, Maranta
108. Han SS, Sun HK, Lee JP, Ha JW, Kim SJ, Kim YS. Outcome of renal R, Van Der Meer I, Conway EM, Zipfel PF, Goodship TH, Remuzzi G.
allograft in patients with Henoch-Schonlein nephritis: single-center Relative role of genetic complement abnormalities in sporadic and
experience and systematic review. Transplantation 2010;89:721– familial aHUS and their impact on clinical phenotype. Clin J Am Soc
6. Nephrol 2010;5:1844–59.
109. Ponticelli C, Glassock RJ. Posttransplant recurrence of primary glo- 127. Fremeaux-Bacchi V, Arzouk N, Ferlicot S, Charpentier B, Snanoudj
merulonephritis. Clin J Am Soc Nephrol 2010;5:2363–72. R, Durrbach A. Recurrence of HUS due to CD46/MCP mutation after
110. Thervet E, Aouizerate J, Noel LH, Brocheriou I, Martinez F, Mamzer renal transplantation: a role for endothelial microchimerism. Am J
MF, Legendre C. Histologic recurrence of Henoch-Schonlein Pur- Transplant 2007;7:2047–51.
pura nephropathy after renal transplantation on routine allograft 128. Kim JJ, Goodship TH, Tizard J, Inward C. Plasma therapy for atypical
biopsy. Transplantation 2011;92:907–12. haemolytic uraemic syndrome associated with heterozygous factor
111. Ponticelli C, Moroni G, Glassock RJ. Recurrence of secondary glo- H mutations. Pediatr Nephrol 2011;26:2073–6.
merular disease after renal transplantation. Clin J Am Soc Nephrol 129. Haller W, Milford DV, Goodship TH, Sharif K, Mirza DF, Mckiernan
2011;6:1214–21. PJ. Successful isolated liver transplantation in a child with atypical
112. Kanaan N, Mourad G, Thervet E, Peeters P, Hourmant M, Vanren- hemolytic uremic syndrome and a mutation in complement factor
terghem Y, De Meyer M, Mourad M, Marechal C, Goffin E, Pirson Y. H. Am J Transplant 2010;10:2142–7.
Recurrence and graft loss after kidney transplantation for henoch- 130. Saland JM, Emre SH, Shneider BL, Benchimol C, Ames S, Bromberg
schonlein purpura nephritis: a multicenter analysis. Clin J Am Soc JS, Remuzzi G, Strain L, Goodship TH. Favorable long-term out-
Nephrol 2011;6:1768–72. come after liver-kidney transplant for recurrent hemolytic uremic
113. Samuel JP, Bell CS, Molony DA, Braun MC. Long-term outcome of syndrome associated with a factor H mutation. Am J Transplant
renal transplantation patients with Henoch-Schonlein purpura. Clin 2006;6:1948–52.
J Am Soc Nephrol 2011a;6:2034–40. 131. Saland JM, Shneider BL, Bromberg JS, Shi PA, Ward SC, Magid MS,
114. Hickey CA, Beattie TJ, Cowieson J, Miyashita Y, Strife CF, Frem JC, Benchimol C, Seikaly MG, Emre SH, Bresin E, Remuzzi G. Successful
Peterson JM, Butani L, Jones DP, Havens PL, Patel HP, Wong CS, split liver-kidney transplant for factor H associated hemolytic uremic
Andreoli SP, Rothbaum RJ, Beck AM, Tarr PI. Early volume expan- syndrome. Clin J Am Soc Nephrol 2009;4:201–6.
sion during diarrhea and relative nephroprotection during sub- 132. Hofer J, Giner T, Cortina G, Jungraithmayr T, Masalskiene J, Dobil-
sequent hemolytic uremic syndrome. Arch Pediatr Adolesc Med iene D, Mitkiene R, Pundziene B, Rudaitis S. Successful living-related
2011;165:884–9. renal transplantation in a patient with factor H antibody-asso-
115. Garg AX, Suri RS, Barrowman N, Rehman F, Matsell D, Rosas- ciated atypical hemolytic uremic syndrome. Pediatr Transplant
Arellano MP, Salvadori M, Haynes RB, Clark WF. Long-term renal 2015;19:E121–5.
prognosis of diarrhea-associated hemolytic uremic syndrome: a 133. Khandelwal P, Sinha A, Hari P, Bansal VK, Dinda AK, Bagga A. Out-
systematic review, meta-analysis, and meta-regression. JAMA comes of renal transplant in patients with anti-complement factor
2003;290:1360–70. H antibody-associated hemolytic uremic syndrome. Pediatr Trans-
116. Sharma AP, Filler G, Dwight P, Clark WF. Chronic renal disease is plant 2014;18:E134–9.
more prevalent in patients with hemolytic uremic syndrome who 134. Al-Akash SI, Almond PS, Savell Jr VH, Gharaybeh SI, Hogue C.
had a positive history of diarrhea. Kidney Int 2010;78:598–604. Eculizumab induces long-term remission in recurrent post-trans-
117. Ferraris JR, Ramirez JA, Ruiz S, Caletti MG, Vallejo G, Piantanida JJ, plant HUS associated with C3 gene mutation. Pediatr Nephrol
Araujo JL, Sojo ET. Shiga toxin-associated hemolytic uremic syn- 2011;26:613–9.
drome: absence of recurrence after renal transplantation. Pediatr 135. Weitz M, Amon O, Bassler D, Koenigsrainer A, Nadalin S. Prophylac-
Nephrol 2002;17:809–14. tic eculizumab prior to kidney transplantation for atypical hemolytic
118. Loirat C, Niaudet P. The risk of recurrence of hemolytic uremic uremic syndrome. Pediatr Nephrol 2011;26:1325–9.
syndrome after renal transplantation in children. Pediatr Nephrol 136. Zuber J, Quintrec ML, Krid S, Bertoye C, Gueutin V, Lahoche A,
2003;18:1095–101. Heyne N, Ardissino G, Chatelet V, Noel LH, Hourmant M, Niaudet
119. Banerjee R, Hersh AL, Newland J, Beekmann SE, Polgreen PM, P, V FB, Rondeau E, Legendre C, Loirat C. Eculizumab for Atypical
Bender J, Shaw J, Copelovitch L, Kaplan BS, Shah SS. Streptococcus Hemolytic Uremic Syndrome Recurrence in Renal Transplantation.
pneumoniae-associated hemolytic uremic syndrome among chil- Am J Transplant 2012.
dren in North America. Pediatr Infect Dis J 2011;30:736–9. 137. Licht C, Greenbaum LA, Muus P, Babu S, Bedrosian CL, Cohen DJ,
120. Zuber J, Le Quintrec M, Sberro-Soussan R, Loirat C, Fremeaux-Bac- Delmas Y, Douglas K, Furman RR, Gaber OA, Goodship T, Herthe-
chi V, Legendre C. New insights into postrenal transplant hemolytic lius M, Hourmant M, Legendre CM, Remuzzi G, Sheerin N, Trivelli
uremic syndrome. Nat Rev Nephrol 2011;7:23–35. A, Loirat C. Efficacy and safety of eculizumab in atypical hemolytic
121. Moore I, Strain L, Pappworth I, Kavanagh D, Barlow PN, Herbert uremic syndrome from 2-year extensions of phase 2 studies. Kidney
AP, Schmidt CQ, Staniforth SJ, Holmes LV, Ward R, Morgan L, Good- Int 2015b;87:1061–73.
ship THJ, Marchbank KJ. Association of factor H autoantibodies with 138. Bartosh SM, Fine RN, Sullivan EK. Outcome after transplantation of
deletions of CFHR1, CFHR3, CFHR4, and with mutations in CFH, young patients with systemic lupus erythematosus: a report of the
CFI, CD46, and C3 in patients with atypical hemolytic uremic syn- North American pediatric renal transplant cooperative study. Trans-
drome. Blood 2010;115:379–87. plantation 2001;72:973–8.
664 Kidney Transplantation: Principles and Practice

139. Fargue S, Harambat J, Gagnadoux MF, Tsimaratos M, Janssen F, Lla- 161. Rocca S, Santilli V, Cotugno N, Concato C, Manno EC, Nocentini G,
nas B, Bertheleme JP, Boudailliez B, Champion G, Guyot C, Macher Macchiarulo G, Cancrini C, Finocchi A, Guzzo I, Dello Strologo L,
MA, Nivet H, Ranchin B, Salomon R, Taque S, Rolland MO, Cochat Palma P. Waning of vaccine-induced immunity to measles in kidney
P. Effect of conservative treatment on the renal outcome of children transplanted children. Medicine (Baltimore) 2016;95:e4738.
with primary hyperoxaluria type 1. Kidney Int 2009;76:767–73. 162. Ojo AO, Hanson JA, Wolfe RA, Agodoa LY, Leavey SF, Leichtman A,
140. Khorsandi SE, Samyn M, Hassan A, Vilca-Melendez H, Waller S, Young EW, Port FK. Dialysis modality and the risk of allograft thrombo-
Shroff R, Koffman G, Van’t Hoff W, Baker A, Dhawan A, Heaton sis in adult renal transplant recipients. Kidney Int 1999;55:1952–60.
N. An institutional experience of pre-emptive liver transplanta- 163. Irish A. Hypercoagulability in renal transplant recipients. Identify-
tion for pediatric primary hyperoxaluria type 1. Pediatr Transplant ing patients at risk of renal allograft thrombosis and evaluating strat-
2016;20:523–9. egies for prevention. Am J Cardiovasc Drugs 2004;4:139–49.
141. Squires J, Nguyen C. Complexity of pre-emptive liver transplantation 164. Van Lieburg AF, De Jong MC, Hoitsma AJ, Buskens FG, Schroder
in children with primary hyperoxaluria type 1. Pediatr Transplant CH, Monnens LA. Renal transplant thrombosis in children. J Pediatr
2016;20:604–6. Surg 1995;30:615–9.
142. Eytan Mor MD, Weismann I. Current treatment for primary hyper- 165. Kist-Van Holthe JE, Ho PL, Stablein D, Harmon WE, Baum MA. Out-
oxaluria type 1: when should liver/kidney transplantation be con- come of renal transplantation for Wilms’ tumor and Denys-Drash
sidered. Pediatr Transplant 2009;13:805–7. syndrome: a report of the North American Pediatric Renal Trans-
143. Bergstralh EJ, Monico CG, Lieske JC, Herges RM, Langman CB, Hoppe plant Cooperative Study. Pediatr Transplant 2005;9:305–10.
B, Milliner DS. Transplantation outcomes in primary hyperoxal- 166. European best practice guidelines for renal transplantation. Section IV:
uria. Am J Transplant 2010;10:2493–501. Long-term management of the transplant recipient. IV.11 Paediatrics
144. Langlois V, Geary D, Murray L, Champoux S, Hebert D, Goodyer (specific problems). Nephrol Dial Transplant 2002;17(Suppl. 4):55–8.
P. Polyuria and proteinuria in cystinosis have no impact on renal 167. Grigoriev Y, Lange J, Peterson SM, Takashima JR, Ritchey ML, Ko D,
transplantation. A report of the North American Pediatric Renal Feusner JH, Shamberger RC, Green DM, Breslow NE. Treatments and
Transplant Cooperative Study. Pediatr Nephrol 2000;15:7–10. outcomes for end-stage renal disease following Wilms tumor. Pediatr
145. Van Stralen KJ, Emma F, Jager KJ, Verrina E, Schaefer F, Laube GF, Nephrol 2012;27:1325–33.
Lewis MA, Levtchenko EN. Improvement in the renal prognosis in 168. Bagga HS, Lin S, Williams A, Schold J, Chertack N, Goldfarb D, Wood
nephropathic cystinosis. Clin J Am Soc Nephrol 2011;6:2485–91. H. Trends in renal transplantation rates in patients with congenital
146. Malatesta-Muncher R, Wansapura J, Taylor M, Lindquist D, Hor urinary tract disorders. J Urol 2016;195:1257–62.
K, Mitsnefes M. Early cardiac dysfunction in pediatric patients on 169. Sierralta MC, Gonzalez G, Nome C, Pinilla C, Correa R, Mansilla J, Rodri-
maintenance dialysis and post kidney transplant. Pediatr Nephrol. guez J, Delucchi A, Ossandon F. Kidney transplant in pediatric patients
2012;27:1157–64. with severe bladder pathology. Pediatr Transplant 2015;19:675–83.
147. Chinali M, Matteucci MC, Franceschini A, Doyon A, Pongiglione G, 170. Aki FT, Aydin AM, Dogan HS, Donmez MI, Erkan I, Duzova A,
Rinelli G, Schaefer F. Advanced Parameters of cardiac mechanics in Topaloglu R, Tekgul S. Does lower urinary tract status affect
children with CKD: the 4C study. Clin J Am Soc Nephrol. 2015;10: renal transplantation outcomes in children? Transplant Proc
1357–63. 2015;47:1114–6.
148. Shamszad P, Slesnick TC, Smith EO, Taylor MD, Feig DI. Association 171. Pereira PL, Urrutia MJ, Lobato R, Jaureguizar E. Renal transplanta-
between left ventricular mass index and cardiac function in pediatric tion in augmented bladders. Curr Urol Rep 2014;15:431.
dialysis patients. Pediatr Nephrol. 2012;27:835–41. 172. Alexopoulos S, Lightner A, Concepcion W, Rose M, Salcedo-Concep-
149. Fraser N, Lyon PC, Williams AR, Christian MT, Shenoy MU. Native cion K, Salvatierra O. Pediatric kidney recipients with small capacity,
nephrectomy in pediatric transplantation—less is more!. J Pediatr defunctionalized urinary bladders receiving adult-sized kidney with-
Urol 2013;9:84–9. out prior bladder augmentation. Transplantation 2011;91:452–6.
150. Kim MS, Primack W, Harmon WE. Congenital nephrotic syndrome: 173. Vyas S, Roberti I. Anuria since birth: does it impact outcome of kid-
preemptive bilateral nephrectomy and dialysis before renal trans- ney transplant in infants? Pediatr Transplant 2016;20:1032–7.
plantation. J Am Soc Nephrol 1992;3:260–3. 174. Riley P, Marks SD, Desai DY, Mushtaq I, Koffman G, Mamode N. Chal-
151. Mattoo TK, Al-Sowailem AM, Al-Harbi MS, Mahmood MA, Katawee lenges facing renal transplantation in pediatric patients with lower
Y, Hassab MH. Nephrotic syndrome in 1st year of life and the role of urinary tract dysfunction. Transplantation 2010;89:1299–307.
unilateral nephrectomy. Pediatr Nephrol 1992;6:16–8. 175. Chaudhuri A, Salvatierra Jr O, Alexander SR, Sarwal MM. Option
152. Kovacevic L, Reid CJ, Rigden SP. Management of congenital of pre-emptive nephrectomy and renal transplantation for Bartter’s
nephrotic syndrome. Pediatr Nephrol 2003;18:426–30. syndrome. Pediatr Transplant 2006;10:266–70.
153. Licht C, Eifinger F, Gharib M, Offner G, Michalk DV, Querfeld U. A 176. Ghane Sharbaf F, Bitzan M, Szymanski KM, Bell LE, Gupta I, Tcher-
stepwise approach to the treatment of early onset nephrotic syn- venkov J, Capolicchio JP. Native nephrectomy prior to pediatric kid-
drome. Pediatr Nephrol 2000;14:1077–82. ney transplantation: biological and clinical aspects. Pediatr Nephrol
154. Vincenti F, Larsen CP, Alberu J, Bresnahan B, Garcia VD, Kothari J, 2012;27:1179–88.
Lang P, Urrea EM, Massari P, Mondragon-Ramirez G, Reyes-Acev- 177. Auber F, Jeanpierre C, Denamur E, Jaubert F, Schleiermacher G,
edo R, Rice K, Rostaing L, Steinberg S, Xing J, Agarwal M, Harler MB, Patte C, Cabrol S, Leverger G, Nihoul-Fékété C, Sarnacki S. Man-
Charpentier B. Three-year outcomes from BENEFIT, a randomized, agement of Wilms tumors in Drash and Frasier syndromes. Pediatr
active-controlled, parallel-group study in adult kidney transplant Blood Cancer 2009;52:55–9.
recipients. Am J Transplant 2012;12:210–7. 178. Baez-Trinidad LG, Lendvay TS, Broecker BH, Smith EA, Warshaw
155. Jonas MM, Zilleruelo GE, Larue SI, Abitbol C, Strauss J, Lu Y. BL, Hymes L, Kirsch AJ. Efficacy of nephrectomy for the treat-
Hepatitis C infection in a pediatric dialysis population. Pediatrics ment of nephrogenic hypertension in a pediatric population. J Urol
1992;89:707–9. 2003;170:1655–7. 2.
156. Molle ZL, Baqi N, Gretch D, Hidalgo G, Tejani A, Rabinowitz SS. Hep- 179. Schlomer BJ, Smith PJ, Barber TD, Baker LA. Nephrectomy for
atitis C infection in children and adolescents with end-stage renal hypertension in pediatric patients with a unilateral poorly function-
disease. Pediatr Nephrol 2002;17:444–9. ing kidney: a contemporary cohort. J Pediatr Urol 2011;7:373–7.
157. Abuali MM, Arnon R, Posada R. An update on immunizations before 180. Cavallini M, Di Zazzo G, Giordano U, Pongiglione G, Dello Strologo L,
and after transplantation in the pediatric solid organ transplant Capozza N, Emma F, Matteucci MC. Long-term cardiovascular effects
recipient. Pediatr Transplant 2011;15:770–7. of pre-transplant native kidney nephrectomy in children. Pediatr
158. Esposito S, Mastrolia MV, Prada E, Pietrasanta C, Principi N. Vac- Nephrol 2010;25:2523–9.
cine administration in children with chronic kidney disease. Vaccine 181. Goslin BJ, Bunchman T, Robertson D, Decou J. Benefits of transperi-
2014;32:6601–6. toneal approach to bilateral pretransplant laparoscopic nephrec-
159. Prelog M, Pohl M, Ermisch B, Fuchshuber A, Huzly D, Jungraithmayr tomies in pediatric patients. J Laparoendosc Adv Surg Tech A
T, Forster J, Zimmerhackl LB. Demand for evaluation of vaccination 2013;23:402–4.
antibody titers in children considered for renal transplantation. Pedi- 182. Capozza N, Collura G, Falappa P, Caione P. Renal embolization as
atr Transplant 2007;11:73–6. an alternative to surgical nephrectomy in children. Transplant Proc
160. Genc G, Ozkaya O, Aygun C, Yakupoglu YK, Nalcacioglu H. Vac- 2007;39:1782–4.
cination status of children considered for renal transplants: missed 183. Slickers J, Duquette P, Hooper S, Gipson D. Clinical predictors of neu-
opportunities for vaccine preventable diseases. Exp Clin Transplant rocognitive deficits in children with chronic kidney disease. Pediatr
2012;10:314–8. Nephrol 2007;22:565–72.
37 • Kidney Transplantation in Children 665

184. Qvist E, Pihko H, Fagerudd P, Valanne L, Lamminranta S, Karikoski of depletional induction and posttransplant lymphoproliferative
J, Sainio K, Ronnholm K, Jalanko H, Holmberg C. Neurodevelopmen- disease in kidney recipients treated with alemtuzumab. Am J Trans-
tal outcome in high-risk patients after renal transplantation in early plant 2007;7:2619–25.
childhood. Pediatr Transplant 2002;6:53–62. 205. Ortiz J, Palma-Vargas J, Wright F, Bingaman A, Agha I, Rosenblatt
185. 2010. NAPRTCS Annual Transplant Report. North American Pedi- S, Foster P. Campath induction for kidney transplantation: report of
atric Renal Trials and Collaborative Studies. 297 cases. Transplantation 2008;85:1550–6.
186. Hamiwka LD, Midgley JP, Hamiwka LA. Seizures in children after 206. Bartosh SM, Knechtle SJ, Sollinger HW. Campath-1H use in pediatric
kidney transplantation: has the risk changed and can we predict renal transplantation. Am J Transplant 2005;5:1569–73.
who is at greatest risk? Pediatr Transplant 2008;12:527–30. 207. Sung J, Barry JM, Jenkins R, Rozansky D, Iragorri S, Conlin M, Al-
187. Dobbels F, Ruppar T, De Geest S, Decorte A, Van Damme-Lombaerts Uzri A. Alemtuzumab induction with tacrolimus monotherapy
R, Fine RN. Adherence to the immunosuppressive regimen in pediat- in 25 pediatric renal transplant recipients. Pediatr Transplant
ric kidney transplant recipients: a systematic review. Pediatr Trans- 2013;17:718–25.
plant 2010;14:603–13. 208. Supe-Markovina K, Melquist JJ, Connolly D, Dicarlo HN, Waltzer
188. Salvatierra Jr O, Singh T, Shifrin R, Conley S, Alexander S, Tan- WC, Fine RN, Darras FS. Alemtuzumab with corticosteroid minimi-
ney D, Lemley K, Sarwal M, Mackie F, Alfrey E, Orlandi P, Zarins C, zation for pediatric deceased donor renal transplantation: a seven-yr
Herfkens R. Successful transplantation of adult-sized kidneys into experience. Pediatr Transplant 2014;18:363–8.
infants requires maintenance of high aortic blood flow. Transplanta- 209. De Serres SA, Mfarrej BG, Magee CN, Benitez F, Ashoor I, Sayegh
tion 1998;66:819–23. MH, Harmon WE, Najafian N. Immune profile of pediatric renal
189. Drake K, Nehus E, Goebel J. Hyponatremia, hypo-osmolality, and transplant recipients following alemtuzumab induction. J Am Soc
seizures in children early post-kidney transplant. Pediatr Transplant Nephrol 2012;23:174–82.
2015;19:698–703. 210. Abraham EC, Wilson AC, Goebel J. Current kidney allocation rules
190. Penna FJ, Harvey E, John P, Armstrong D, Luginbuehl I, Odeh RI, and their impact on a pediatric transplant center. Am J Transplant
Alyami F, Koyle MA, Lorenzo AJ. Intra-arterial nitroglycerin for 2009;9:404–8.
intra-operative arterial vasospasm during pediatric renal transplan- 211. Kim IK, Choi J, Vo AA, Kang A, Patel M, Toyoda M, Mirocha J, Kamil
tation. Pediatr Transplant 2016;20:463–6. ES, Cohen JL, Louie S, Galera O, Jordan SC, Puliyanda DP. Safety and
191. Fontana I, Bertocchi M, Centanaro M, Varotti G, Santori G, Mondello Efficacy of Alemtuzumab Induction in Highly Sensitized Pediatric
R, Tagliamacco A, Cupo P, Barabani C, Palombo D. Abdominal com- Renal Transplant Recipients. Transplantation 2017;101:883–9.
partment syndrome: an underrated complication in pediatric kidney 212. Sarwal MM, Ettenger RB, Dharnidharka V, Benfield M, Mathias R,
transplantation. Transplant Proc 2014;46:2251–3. Portale A, Mcdonald R, Harmon W, Kershaw D, Vehaskari VM,
192. Giuliani S, Gamba P, Kiblawi R, Midrio P, Ghirardo G, Zanon GF. Kamil E, Baluarte HJ, Warady B, Tang L, Liu J, Li L, Naesens M,
Lymphocele after pediatric kidney transplantation: incidence and Sigdel T, Waskerwitz J, Salvatierra O. Complete steroid avoidance
risk factors. Pediatr Transplant 2014;18:720–5. is effective and safe in children with renal transplants: a multi-
193. Damasio MB, Ording Muller LS, Piaggio G, Marks SD, Riccabona center randomized trial with three-year follow-up. Am J Transplant
M. Imaging in pediatric renal transplantation. Pediatr Transplant 2012b;12:2719–29.
2017;21. 213. Swiatecka-Urban A. Anti-interleukin-2 receptor antibodies for the
194. Lerret SM, Weiss ME, Stendahl GL, Chapman S, Menendez J, Williams prevention of rejection in pediatric renal transplant patients: current
L, Nadler ML, Neighbors K, Amsden K, Cao Y, Nugent M, Alonso EM, status. Paediatr Drugs 2003;5:699–716.
Simpson P. Pediatric solid organ transplant recipients: transition to 214. Dolan N, Waldron M, O’connell M, Eustace N, Carson K, Awan A.
home and chronic illness care. Pediatr Transplant 2015;19:118–29. Basiliximab induced non-cardiogenic pulmonary edema in two pedi-
195. Sampaio MS, Poommipanit N, Kuo HT, Reddy PN, Cho YW, Shah T, atric renal transplant recipients. Pediatr Nephrol 2009;24:2261–5.
Bunnapradist S. Induction therapy in pediatric kidney transplant 215. Offner G, Toenshoff B, Hocker B, Krauss M, Bulla M, Cochat P, Feh-
recipients discharged with a triple drug immunosuppressive regi- renbach H, Fischer W, Foulard M, Hoppe B, Hoyer PF, Jungraith-
men. Pediatr Transplant 2010;14:770–8. mayr TC, Klaus G, Latta K, Leichter H, Mihatsch MJ, Misselwitz J,
196. Crowson CN, Reed RD, Shelton BA, Maclennan PA, Locke JE. Lym- Montoya C, Muller-Wiefel DE, Neuhaus TJ, Pape L, Querfeld U, Plank
phocyte-depleting induction therapy lowers the risk of acute rejec- C, Schwarke D, Wygoda S, Zimmerhackl LB. Efficacy and safety of
tion in African American pediatric kidney transplant recipients. basiliximab in pediatric renal transplant patients receiving cyclo-
Pediatr Transplant 2017;21. sporine, mycophenolate mofetil, and steroids. Transplantation
197. Shang W, Feng G, Gao S, Wang Z, Pang X, Li J, Liu L, Feng Y, Xie H, 2008;86:1241–8.
Zhang S, Qiao B. Reduced ATG-F dosage for induction in pediatric 216. Fryer JP, Benedetti E, Gillingham K, Najarian JS, Matas AJ. Steroid-
renal transplantation: a single-center experience. Pediatr Trans- related complications in pediatric kidney transplant recipients in the
plant 2014;18:240–5. cyclosporine era. Transplant Proc 1994;26:91–2.
198. Peddi VR, Bryant M, Roy-Chaudhury P, Woodle ES, First MR. Safety, 217. Jabs K, Sullivan EK, Avner ED, Harmon WE. Alternate-day steroid
efficacy, and cost analysis of thymoglobulin induction therapy dosing improves growth without adversely affecting graft survival
with intermittent dosing based on CD3+ lymphocyte counts in kid- or long-term graft function. A report of the North American Pediat-
ney and kidney-pancreas transplant recipients. Transplantation ric Renal Transplant Cooperative Study. Transplantation 1996;61:
2002;73:1514–8. 31–6.
199. Olaitan OK, Zimmermann JA, Shields WP, Rodriguez-Navas G, 218. Broyer M, Guest G, Gagnadoux MF. Growth rate in children receiv-
Awan A, Mohan P, Little DM, Hickey DP. Long-term outcome of ing alternate-day corticosteroid treatment after kidney transplanta-
intensive initial immunosuppression protocol in pediatric deceased tion. J Pediatr 1992;120:721–5.
donor renal transplantation. Pediatr Transplant 2010;14:87–92. 219. Kasiske BL, Chakkera HA, Louis TA, Ma JZ. A meta-analysis of
200. Colleen Hastings M, Wyatt RJ, Lau KK, Jones DP, Powell SL, Hays immunosuppression withdrawal trials in renal transplantation. J
DW, Gaber LW, Osama Gaber A, Ault BH. Five years’ experience Am Soc Nephrol 2000;11:1910–7.
with thymoglobulin induction in a pediatric renal transplant popu- 220. Hocker B, Weber LT, Feneberg R, Drube J, John U, Fehrenbach
lation. Pediatr Transplant 2006;10:805–10. H, Pohl M, Zimmering M, Frund S, Klaus G, Wuhl E, Tonshoff B.
201. Brophy PD, Thomas SE, Mcbryde KD, Bunchman TE. Comparison of Improved growth and cardiovascular risk after late steroid with-
polyclonal induction agents in pediatric renal transplantation. Pedi- drawal: 2-year results of a prospective, randomised trial in paediatric
atr Transplant 2001;5:174–8. renal transplantation. Nephrol Dial Transplant 2010;25:617–24.
202. Fernberg P, Edgren G, Adami J, Ingvar A, Bellocco R, Tufveson G, 221. Marks SD, Trompeter RS. Steroid preservation: the rationale for con-
Hoglund P, Kinch A, Simard JF, Baecklund E, Lindelof B, Pawitan Y, tinued prescribing. Pediatr Nephrol 2006;21:305–7.
Smedby KE. Time trends in risk and risk determinants of non-Hodg- 222. Mcdonald RA, Smith JM, Ho M, Lindblad R, Ikle D, Grimm P, Wyatt
kin lymphoma in solid organ transplant recipients. Am J Transplant R, Arar M, Liereman D, Bridges N, Harmon W. Incidence of PTLD in
2011;11:2472–82. pediatric renal transplant recipients receiving basiliximab, calcineurin
203. Faull RJ, Hollett P, Mcdonald SP. Lymphoproliferative disease after inhibitor, sirolimus and steroids. Am J Transplant 2008;8:984–9.
renal transplantation in Australia and New Zealand. Transplanta- 223. Benfield MR, Bartosh S, Ikle D, Warshaw B, Bridges N, Morrison Y,
tion 2005;80:193–7. Harmon W. A randomized double-blind, placebo controlled trial of
204. Kirk AD, Cherikh WS, Ring M, Burke G, Kaufman D, Knechtle SJ, steroid withdrawal after pediatric renal transplantation. Am J Trans-
Potdar S, Shapiro R, Dharnidharka VR, Kauffman HM. Dissociation plant 2010;10:81–8.
666 Kidney Transplantation: Principles and Practice

224. Nehus E, Liu C, Hooper DK, Macaluso M, Kim MO. Clinical Practice of 241. Duong SQ, Lal AK, Joshi R, Feingold B, Venkataramanan R. Transi-
Steroid Avoidance in Pediatric Kidney Transplantation. Am J Trans- tion from brand to generic tacrolimus is associated with a decrease in
plant 2015;15:2203–10. trough blood concentration in pediatric heart transplant recipients.
225. Li L, Chaudhuri A, Chen A, Zhao X, Bezchinsky M, Concepcion W, Pediatr Transplant 2015;19:911–7.
Salvatierra Jr O, Sarwal MM. Efficacy and safety of thymoglobulin 242. Jacobo-Cabral CO, Garcia-Roca P, Reyes H, Lozada-Rojas L, Cruz-
induction as an alternative approach for steroid-free maintenance Antonio L, Medeiros M, Castaneda-Hernandez G. Limustin(R), a
immunosuppression in pediatric renal transplantation. Transplan- non-innovator tacrolimus formulation, yields reduced drug expo-
tation 2010a;90:1516–20. sure in pediatric renal transplant recipients. Pediatr Transplant
226. Warejko JK, Hmiel SP. Single-center experience in pediatric renal 2014;18:706–13.
transplantation using thymoglobulin induction and steroid minimi- 243. Gijsen VM, Van Schaik RH, Soldin OP, Soldin SJ, Nulman I, Koren
zation. Pediatr Transplant 2014;18:816–21. G, De Wildt SN. P450 oxidoreductase *28 (POR*28) and tacrolimus
227. Grenda R, Watson A, Trompeter R, Tonshoff B, Jaray J, Fitzpatrick M, disposition in pediatric kidney transplant recipients—a pilot study.
Murer L, Vondrak K, Maxwell H, Van Damme-Lombaerts R, Loirat Ther Drug Monit 2014;36:152–8.
C, Mor E, Cochat P, Milford DV, Brown M, Webb NJ. A randomized 244. Lalan S, Abdel-Rahman S, Gaedigk A, Leeder JS, Warady BA, Dai H,
trial to assess the impact of early steroid withdrawal on growth in Blowey D. Effect of CYP3A5 genotype, steroids, and azoles on tacro-
pediatric renal transplantation: the TWIST study. Am J Transplant limus in a pediatric renal transplant population. Pediatr Nephrol
2010;10:828–36. 2014;29:2039–49.
228. Webb NJ, Douglas SE, Rajai A, Roberts SA, Grenda R, Marks SD, Wat- 245. Sy SK, Heuberger J, Shilbayeh S, Conrado DJ, Derendorf H. A Markov
son AR, Fitzpatrick M, Vondrak K, Maxwell H, Jaray J, Van Damme- chain model to evaluate the effect of CYP3A5 and ABCB1 polymor-
Lombaerts R, Milford DV, Godefroid N, Cochat P, Ognjanovic M, phisms on adverse events associated with tacrolimus in pediatric
Murer L, Mcculloch M, Tonshoff B. Corticosteroid-free Kidney Trans- renal transplantation. AAPS J 2013;15:1189–99.
plantation Improves Growth: 2-Year Follow-up of the TWIST Ran- 246. Min SI, Ha J, Kang HG, Ahn S, Park T, Park DD, Kim SM, Hong
domized Controlled Trial. Transplantation 2015;99:1178–85. HJ, Min SK, Ha IS, Kim SJ. Conversion of twice-daily tacrolimus to
229. Sarwal MM, Ettenger RB, Dharnidharka V, Benfield M, Mathias R, once-daily tacrolimus formulation in stable pediatric kidney trans-
Portale A, Mcdonald R, Harmon W, Kershaw D, Vehaskari VM, plant recipients: pharmacokinetics and efficacy. Am J Transplant
Kamil E, Baluarte HJ, Warady B, Tang L, Liu J, Li L, Naesens M, 2013;13:2191–7.
Sigdel T, Waskerwitz J, Salvatierra O. Complete steroid avoidance 247. Lapeyraque AL, Kassir N, Theoret Y, Krajinovic M, Clermont MJ, Lit-
is effective and safe in children with renal transplants: a multi- alien C, Phan V. Conversion from twice- to once-daily tacrolimus in
center randomized trial with three-year follow-up. Am J Transplant pediatric kidney recipients: a pharmacokinetic and bioequivalence
2012a;12:2719–29. study. Pediatr Nephrol 2014;29:1081–8.
230. Naesens M, Salvatierra O, Benfield M, Ettenger R, Dharnidharka V, 248. Zhao W, Fakhoury M, Baudouin V, Storme T, Maisin A, Deschenes
Harmon W, Mathias R, Sarwal MM. Subclinical inflammation and G, Jacqz-Aigrain E. Population pharmacokinetics and pharmaco-
chronic renal allograft injury in a randomized trial on steroid avoid- genetics of once daily prolonged-release formulation of tacrolimus
ance in pediatric kidney transplantation. Am J Transplant 2012. in pediatric and adolescent kidney transplant recipients. Eur J Clin
231. Nehus E, Goebel J, Abraham E. Outcomes of steroid-avoidance proto- Pharmacol 2013a;69:189–95.
cols in pediatric kidney transplant recipients. Am J Transplant 2012. 249. Monteverde ML, Ibanez J, Balbarrey Z, Chaparro A, Diaz M, Turconi
232. Mericq V, Salas P, Pinto V, Cano F, Reyes L, Brown K, Gonzalez M, A. Conversion to sirolimus in pediatric renal transplant patients: a
Michea L, Delgado I, Delucchi A. Steroid withdrawal in pediatric kid- single-center experience. Pediatr Transplant 2012;16:582–8.
ney transplant allows better growth, lipids and body composition: a 250. Forster J, Ahlenstiel-Grunow T, Zapf A, Mynarek M, Pape L. Puber-
randomized controlled trial. Horm Res Paediatr 2013;79:88–96. tal Development in Pediatric Kidney Transplant Patients Receiving
233. Weaver Jr DJ, Selewski D, Janjua H, Iorember F. Improved cardiovas- Mammalian Target of Rapamycin Inhibitors or Conventional Immu-
cular risk factors in pediatric renal transplant recipients on steroid nosuppression. Transplantation 2016;100:2461–70.
avoidance immunosuppression: a study of the Midwest Pediatric 251. Hymes LC, Warshaw BL. Linear growth in pediatric renal transplant
Nephrology Consortium. Pediatr Transplant 2016;20:59–67. recipients receiving sirolimus. Pediatr Transplant 2011b;15:570–2.
234. Wittenhagen P, Thiesson HC, Baudier F, Pedersen EB, Neland M. 252. Gonzalez D, Garcia CD, Azocar M, Waller S, Alonso A, Ariceta G,
Long-term experience of steroid-free pediatric renal transplantation: Mejia N, Santos F. Growth of kidney-transplanted pediatric patients
effects on graft function, body mass index, and longitudinal growth. treated with sirolimus. Pediatr Nephrol 2011a;26:961–6.
Pediatr Transplant 2014;18:35–41. 253. Hymes LC, Warshaw BL. Five-year experience using sirolimus-
235. Tan HP, Donaldson J, Ellis D, Moritz ML, Basu A, Morgan C, Vats AN, based, calcineurin inhibitor-free immunosuppression in pediatric
Erkan E, Shapiro R. Pediatric living donor kidney transplantation renal transplantation. Pediatr Transplant 2011a;15:437–41.
under alemtuzumab pretreatment and tacrolimus monotherapy: 254. Brunkhorst LC, Fichtner A, Hocker B, Burmeister G, Ahlenstiel-
4-year experience. Transplantation 2008;86:1725–31. Grunow T, Krupka K, Bald M, Zapf A, Tonshoff B, Pape L. Efficacy
236. Ellis D, Shapiro R, Moritz M, Vats A, Basu A, Tan H, Kayler L, Janosky and Safety of an Everolimus- vs. a Mycophenolate Mofetil-Based
J, Starzl TE. Renal transplantation in children managed with lym- Regimen in Pediatric Renal Transplant Recipients. PLoS One
phocyte depleting agents and low-dose maintenance tacrolimus 2015;10e0135439.
monotherapy. Transplantation 2007;83:1563–70. 255. Billing H, Burmeister G, Plotnicki L, Ahlenstiel T, Fichtner A, Sander
237. Shapiro R, Ellis D, Tan HP, Moritz ML, Basu A, Vats AN, Kayler LK, A, Hocker B, Tonshoff B, Pape L. Longitudinal growth on an everoli-
Erkan E, Mcfeaters CG, James G, Grosso MJ, Zeevi A, Gray EA, Mar- mus- versus an MMF-based steroid-free immunosuppressive regimen
cos A, Starzl TE. Alemtuzumab pre-conditioning with tacrolimus in paediatric renal transplant recipients. Transpl Int 2013;26:903–9.
monotherapy in pediatric renal transplantation. Am J Transplant 256. Grushkin C, Mahan JD, Mange KC, Hexham JM, Ettenger R. De novo
2007;7:2736–8. therapy with everolimus and reduced-exposure cyclosporine fol-
238. Billing H, Sommerer C, Giese T, Zeier M, Meuer S, Czock D, Ton- lowing pediatric kidney transplantation: a prospective, multicenter,
shoff B. Increased cyclosporin a sensitivity in vivo in pediatric 12-month study. Pediatr Transplant 2013;17:237–43.
renal transplant recipients compared with adults. Ther Drug Monit 257. Hocker B, Zencke S, Pape L, Krupka K, Koster L, Fichtner A, Dello
2012b;34:554–60. Strologo L, Guzzo I, Topaloglu R, Kranz B, Konig J, Bald M, Webb NJ,
239. Zhao W, Maisin A, Baudouin V, Fakhoury M, Storme T, Deschenes Noyan A, Dursun H, Marks S, Ozcakar ZB, Thiel F, Billing H, Pohl
G, Jacqz-Aigrain E. Limited sampling strategy using Bayesian estima- M, Fehrenbach H, Schnitzler P, Bruckner T, Ahlenstiel-Grunow T,
tion for estimating individual exposure of the once-daily prolonged- Tonshoff B. Impact of everolimus and low-dose cyclosporin on cyto-
release formulation of tacrolimus in kidney transplant children. Eur megalovirus replication and disease in pediatric renal transplanta-
J Clin Pharmacol 2013b;69:1181–5. tion. Am J Transplant 2016b;16:921–9.
240. Almeida-Paulo GN, Lubomirov R, Alonso-Sanchez NL, Espinosa- 258. Todorova EK, Huang SH, Kobrzynski MC, Filler G. What is the
Roman L, Fernandez Camblor C, Diaz C, Munoz Bartola G, Carcas- intrapatient variability of mycophenolic acid trough levels? Pediatr
Sansuan AJ. Limited sampling strategies for tacrolimus exposure Transplant 2015;19:669–74.
(AUC0-24) prediction after Prograf((R)) and Advagraf((R)) adminis- 259. Filler G, Foster J, Berard R, Mai I, Lepage N. Age-dependency of myco-
tration in children and adolescents with liver or kidney transplants. phenolate mofetil dosing in combination with tacrolimus after pediat-
Transpl Int 2014;27:939–48. ric renal transplantation. Transplant Proc 2004;36:1327–31.
37 • Kidney Transplantation in Children 667

260. Fukuda T, Goebel J, Cox S, Maseck D, Zhang K, Sherbotie JR, Ellis 279. Pearl MH, Nayak AB, Ettenger RB, Puliyanda D, Palma Diaz MF,
EN, James LP, Ward RM, Vinks AA. UGT1A9, UGT2B7, and MRP2 Zhang Q, Reed EF, Tsai EW. Bortezomib may stabilize pediatric renal
genotypes can predict mycophenolic acid pharmacokinetic vari- transplant recipients with antibody-mediated rejection. Pediatr
ability in pediatric kidney transplant recipients. Ther Drug Monit Nephrol 2016;31:1341–8.
2012;34:671–9. 280. Waiser J, Budde K, Schutz M, Liefeldt L, Rudolph B, Schonemann
261. Gonzalez-Ramirez R, Gonzalez-Banuelos J, Villa Mde L, Jimenez B, C, Neumayer HH, Lachmann N. Comparison between bortezomib
Garcia-Roca P, Cruz-Antonio L, Castaneda-Hernandez G, Medeiros and rituximab in the treatment of antibody-mediated renal allograft
M. Bioavailability of a generic of the immunosuppressive agent rejection. Nephrol Dial Transplant 2012;27:1246–51.
mycophenolate mofetil in pediatric patients. Pediatr Transplant 281. Ghirardo G, Benetti E, Poli F, Vidal E, Della Vella M, Cozzi E, MureR
2014;18:568–74. L. Plasmapheresis-resistant acute humoral rejection successfully
262. Filler G, Todorova EK, Bax K, Alvarez-Elias AC, Huang SH, Kobrzyn- treated with anti-C5 antibody. Pediatr Transplant 2014;18:E1–5.
ski MC. Minimum mycophenolic acid levels are associated with 282. Chehade H, Rotman S, Matter M, Girardin E, Aubert V, Pascual M.
donor-specific antibody formation. Pediatr Transplant 2016;20: Eculizumab to treat antibody-mediated rejection in a 7-year-old kid-
34–8. ney transplant recipient. Pediatrics 2015;135:e551–5.
263. Rose EM, Kennedy SE, Mackie FE. Surveillance biopsies after pae- 283. Li L, Sigdel T, Vitalone M, Lee SH, Sarwal M. Differential immuno-
diatric kidney transplantation: A review. Pediatr Transplant genicity and clinical relevance of kidney compartment specific anti-
2016;20:748–55. gens after renal transplantation. J Proteome Res 2010b;9:6715–21.
264. Upadhyay K, Midgley L, Moudgil A. Safety and efficacy of alemtu- 284. Bjerre A, Tangeraas T, Heidecke H, Dragun D, Dechend R, Staff AC.
zumab in the treatment of late acute renal allograft rejection. Pediatr Angiotensin II type 1 receptor antibodies in childhood kidney trans-
Transplant 2012;16:286–93. plantation. Pediatr Transplant 2016;20:627–32.
265. Sarwal M, Chua MS, Kambham N, Hsieh SC, Satterwhite T, Masek 285. Pearl MH, Leuchter RK, Reed EF, Zhang Q, Ettenger RB, Tsai EW.
M, Salvatierra JR O,. Molecular heterogeneity in acute renal allograft Accelerated rejection, thrombosis, and graft failure with angiotensin
rejection identified by DNA microarray profiling. N Engl J Med II type 1 receptor antibodies. Pediatr Nephrol 2015;30:1371–4.
2003;349:125–38. 286. Mcdonald SP, Craig JC, Australian & New Zealand Paediatric
266. Tsai EW, Rianthavorn P, Gjertson DW, Wallace WD, Reed EF, Nephrology, A. Long-term survival of children with end-stage renal
Ettenger RB. CD20+ lymphocytes in renal allografts are associ- disease. N Engl J Med 2004;350:2654–62.
ated with poor graft survival in pediatric patients. Transplantation 287. Parekh RS, Carroll CE, Wolfe RA, Port FK. Cardiovascular mortality
2006;82:1769–73. in children and young adults with end-stage kidney disease. J Pediatr
267. Muorah MR, Brogan PA, Sebire NJ, Trompeter RS, Marks SD. Dense 2002;141:191–7.
B cell infiltrates in paediatric renal transplant biopsies are predictive 288. Foster, 2011 #707
of allograft loss. Pediatr Transplant 2009;13:217–22. 289. Hooper DK, Williams JC, Carle AC, Amaral S, Chand DH, Ferris ME,
268. Zarkhin V, Kambham N, Li L, Kwok S, Hsieh SC, Salvatierra O, Sar- Patel HP, Licht C, Barletta GM, Zitterman V, Mitsnefes M, Patel UD.
wal MM. Characterization of intra-graft B cells during renal allograft The quality of cardiovascular disease care for adolescents with kid-
rejection. Kidney Int 2008a;74:664–73. ney disease: a Midwest Pediatric Nephrology Consortium study.
269. Ginevri F, Nocera A, Comoli P, Innocente A, Cioni M, Parodi A, Fon- Pediatr Nephrol 2013b;28:939–49.
tana I, Magnasco A, Nocco A, Tagliamacco A, Sementa A, Ceriolo P, 290. Dobrowolski LC, Van Huis M, Van Der Lee JH, Peters Sengers H,
Ghio L, Zecca M, Cardillo M, Garibotto G, Ghiggeri GM, Poli F. Post- Lilien MR, Cransberg K, Cornelissen M, Bouts AH, De Fijter JW,
transplant de novo donor-specific hla antibodies identify pediatric Berger SP, Van Zuilen A, Nurmohamed SA, Betjes MH, Hilbrands L,
kidney recipients at risk for late antibody-mediated rejection. Am J Hoitsma AJ, Bemelman FJ, Krediet CTP, Groothoff JW. Epidemiology
Transplant 2012;12:3355–62. and management of hypertension in paediatric and young adult kid-
270. Cioni M, Nocera A, Innocente A, Tagliamacco A, Trivelli A, Basso S, ney transplant recipients in The Netherlands. Nephrol Dial Trans-
Quartuccio G, Fontana I, Magnasco A, Drago F, Gurrado A, Guido I, plant 2016;31:1947–56.
Compagno F, Garibotto G, Klersy C, Verrina E, Ghiggeri GM, Cardillo 291. Suszynski TM, Rizzari MD, Gillingham KJ, Rheault MN, Kraszkiewicz
M, Comoli P, Ginevri F. De novo donor-specific hla antibodies devel- W, Matas AJ, Chavers BM. Antihypertensive pharmacotherapy and
oping early or late after transplant are associated with the same risk long-term outcomes in pediatric kidney transplantation. Clin Trans-
of graft damage and loss in nonsensitized kidney recipients. J Immu- plant 2013;27:472–80.
nol Res 2017;2017:1747030. 292. Mitsnefes MM, Khoury PR, Mcenery PT. Early posttransplantation
271. Kim JJ, Balasubramanian R, Michaelides G, Wittenhagen P, Sebire hypertension and poor long-term renal allograft survival in pediatric
NJ, Mamode N, Shaw O, Vaughan R, Marks SD. The clinical spec- patients. J Pediatr 2003;143:98–103.
trum of de novo donor-specific antibodies in pediatric renal trans- 293. Hamdani G, Nehus EJ, Hanevold CD, Sebestyen Van Sickle J,
plant recipients. Am J Transplant 2014;14:2350–8. Woroniecki R, Wenderfer SE, Hooper DK, Blowey D, Wilson A,
272. Gulleroglu K, Baskin E, Bayrakci US, Turan M, Ozdemir BH, Moray Warady BA, Mitsnefes MM. Ambulatory blood pressure, left ven-
G, Karakayali H, Haberal M. Antibody-mediated rejection and treat- tricular hypertrophy, and allograft function in children and
ment in pediatric patients: one center’s experience. Exp Clin Trans- young adults after kidney transplantation. Transplantation
plant 2013;11:404–7. 2017;101:150–6.
273. Billing H, Rieger S, Susal C, Waldherr R, Opelz G, Wuhl E, Tonshoff 294. Hamdani G, Nehus EJ, Hooper DK, Mitsnefes MM. Masked hyper-
B. IVIG and rituximab for treatment of chronic antibody-mediated tension and allograft function in pediatric and young adults kidney
rejection: a prospective study in paediatric renal transplantation transplant recipients. Pediatr Transplant 2016;20:1026–31.
with a 2-year follow-up. Transpl Int 2012a;25:1165–73. 295. Tainio J, Qvist E, Miettinen J, Holtta T, Pakarinen M, Jahnukainen
274. Pape L, Becker JU, Immenschuh S, Ahlenstiel T. Acute and chronic T, Jalanko H. Blood pressure profiles 5 to 10 years after transplant
antibody-mediated rejection in pediatric kidney transplantation. in pediatric solid organ recipients. J Clin Hypertens (Greenwich)
Pediatr Nephrol 2015;30:417–24. 2015;17:154–61.
275. Zarkhin V, Li L, Kambham N, Sigdel T, Salvatierra O, Sarwal MM. 296. Habbig S, Volland R, Krupka K, Querfeld U, Dello Strologo L, Noyan
A randomized, prospective trial of rituximab for acute rejection A, Yalcinkaya F, Topaloglu R, Webb NJ, Kemper MJ, Pape L, Bald M,
in pediatric renal transplantation. Am J Transplant 2008b;8: Kranz B, Taylan C, Hocker B, Tonshoff B, Weber LT. Dyslipidemia
2607–17. after pediatric renal transplantation-The impact of immunosuppres-
276. Twombley K, Thach L, Ribeiro A, Joseph C, Seikaly M. Acute anti- sive regimens. Pediatr Transplant 2017;21.
body-mediated rejection in pediatric kidney transplants: a single 297. Wilson AC, Greenbaum LA, Barletta GM, Chand D, Lin JJ, Patel HP,
center experience. Pediatr Transplant 2013;17:E149–55. Mitsnefes M. High prevalence of the metabolic syndrome and associ-
277. Roberti I, Vyas S. Successful treatment of severe acute antibody- ated left ventricular hypertrophy in pediatric renal transplant recipi-
mediated rejection of renal allografts with bortezomib—a report of ents. Pediatr Transplant 2010;14:52–60.
two pediatric cases. Pediatr Transplant 2015;19:E189–92. 298. He S, Le N, Frediani J, Winterberg P, Jin R, Liverman R, Hernandez
278. Nguyen S, Gallay B, Butani L. Efficacy of bortezomib for reducing A, Cleeton R, Vos M. Cardiometabolic risks vary by weight status in
donor-specific antibodies in children and adolescents on a steroid pediatric kidney and liver transplant recipients: a cross-sectional,
minimization regimen. Pediatr Transplant 2014;18:463–8. single-center study in the U.S. 2017.
668 Kidney Transplantation: Principles and Practice

299. Foster BJ, Martz K, Gowrishankar M, Stablein D, Al-Uzri A. Weight 317. Jung YH, Moon KC, Ha JW, Kim SJ, Ha IS, Cheong HI, Kang HG.
and height changes and factors associated with greater weight and Leflunomide therapy for BK virus allograft nephropathy after pedi-
height gains after pediatric renal transplantation: a NAPRTCS study. atric kidney transplantation. Pediatr Transplant 2013;17:E50–4.
Transplantation 2010;89:1103–12. 318. Weigel F, Lemke A, Tonshoff B, Pape L, Fehrenbach H, Henn M,
300. Hooper DK, Kirby CL, Margolis PA, Goebel J. Reliable individualized Hoppe B, Jungraithmayr T, Konrad M, Laube G, Pohl M, Seeman T,
monitoring improves cholesterol control in kidney transplant recipi- Staude H, Kemper MJ, John U. Febrile urinary tract infection after
ents. Pediatrics 2013a;131:e1271–9. pediatric kidney transplantation: a multicenter, prospective observa-
301. Braamskamp MJ, Wijburg FA, Wiegman A. Drug therapy of hyper- tional study. Pediatr Nephrol 2016;31:1021–8.
cholesterolaemia in children and adolescents. Drugs 2012;72:759– 319. Herthelius M, Oborn H. Urinary tract infections and bladder dysfunc-
72. tion after renal transplantation in children. J Urol 2007;177:1883–6.
302. Filler G, Weiglein G, Gharib MT, Casier S. Omega3 fatty acids may 320. Van Der Weide MJ, Cornelissen EA, Van Achterberg T, De Gier RP,
reduce hyperlipidemia in pediatric renal transplant recipients. Pedi- Feitz WF. Lower urinary tract symptoms after renal transplantation
atr Transplant 2012;16:835–9. in children. J Urol 2006;175:297–302; discussion 302.
303. Hogan J, Pietrement C, Sellier-Leclerc AL, Louillet F, Salomon R, 321. Luke PP, Herz DB, Bellinger MF, Chakrabarti P, Vivas CA, Scantle-
Macher MA, Berard E, Couchoud C. Infection-related hospitaliza- bury VP, Hakala TR, Jevnikar AM, Jain A, Shapiro R, Jordan ML.
tions after kidney transplantation in children: incidence, risk factors, Long-term results of pediatric renal transplantation into a dysfunc-
and cost. Pediatr Nephrol 2017;32:2331–41. tional lower urinary tract. Transplantation 2003;76:1578–82.
304. Kizilbash SJ, Rheault MN, Bangdiwala A, Matas A, Chinnakotla 322. Shroff R, Knott C, Gullett A, Wells D, Marks SD, Rees L. Vitamin D
S, Chavers BM. Infection rates in tacrolimus versus cyclosporine- deficiency is associated with short stature and may influence blood
treated pediatric kidney transplant recipients on a rapid discontinu- pressure control in paediatric renal transplant recipients. Pediatr
ation of prednisone protocol: 1-year analysis. Pediatr Transplant Nephrol 2011;26:2227–33.
2017;21. 323. Bonthuis M, Busutti M, Van Stralen KJ, Jager KJ, Baiko S, Bakkaloglu
305. Ettenger R, Chin H, Kesler K, Bridges N, Grimm P, Reed EF, Sarwal S, Battelino N, Gaydarova M, Gianoglio B, Parvex P, Gomes C, Heaf
M, Sibley R, Tsai E, Warshaw B, Kirk AD. Relationship among vire- JG, Podracka L, Kuzmanovska D, Molchanova MS, Pankratenko
mia/viral infection, alloimmunity, and nutritional parameters in TE, Papachristou F, Reusz G, Sanahuja MJ, Shroff R, Groothoff
the first year after pediatric kidney transplantation. Am j transplant JW, Schaefer F, Verrina E. Mineral metabolism in European chil-
2017;17:1549–62. dren living with a renal transplant: a European society for paedi-
306. Hocker B, Zencke S, Krupka K, Fichtner A, Pape L, Dello Strologo L, atric nephrology/european renal association-European dialysis
Guzzo I, Topaloglu R, Kranz B, Konig J, Bald M, Webb NJ, Noyan A, and transplant association registry study. Clin J Am Soc Nephrol
Dursun H, Marks S, Yalcinkaya F, Thiel F, Billing H, Pohl M, Fehren- 2015;10:767–75.
bach H, Bruckner T, Tonshoff B. Cytomegalovirus Infection in Pedi- 324. Ebbert K, Chow J, Krempien J, Matsuda-Abedini M, Dionne J. Vita-
atric Renal Transplantation and the Impact of Chemoprophylaxis min D insufficiency and deficiency in pediatric renal transplant
With (Val-)Ganciclovir. Transplantation 2016a;100:862–70. recipients. Pediatr Transplant 2015;19:492–8.
307. Varela-Fascinetto G, Benchimol C, Reyes-Acevedo R, Genevray M, 325. Franke D, Thomas L, Steffens R, Pavicic L, Gellermann J, Froede K,
Bradley D, Ives J, Silva Jr HT. Tolerability of up to 200 days of pro- Querfeld U, Haffner D, Zivicnjak M. Patterns of growth after kidney
phylaxis with valganciclovir oral solution and/or film-coated tablets transplantation among children with ESRD. Clin J Am Soc Nephrol
in pediatric kidney transplant recipients at risk of cytomegalovirus 2015;10:127–34.
disease. Pediatr Transplant 2017;21. 326. Tsampalieros A, Knoll GA, Molnar AO, Fergusson N, Fergusson
308. Vaudry W, Ettenger R, Jara P, Varela-Fascinetto G, Bouw MR, Ives J, DA. Corticosteroid use and growth after pediatric solid organ trans-
Walker R, Valcyte WVSG. Valganciclovir dosing according to body plantation: a systematic review and meta-analysis. Transplantation
surface area and renal function in pediatric solid organ transplant 2017;101:694–703.
recipients. Am J Transplant 2009;9:636–43. 327. Fine RN, Martz K, Stablein D. What have 20 years of data from the
309. Villeneuve D, Brothers A, Harvey E, Kemna M, Law Y, Nemeth T, North American Pediatric Renal Transplant Cooperative Study
Gantt S. Valganciclovir dosing using area under the curve calcula- taught us about growth following renal transplantation in infants,
tions in pediatric solid organ transplant recipients. Pediatr Trans- children, and adolescents with end-stage renal disease? Pediatr
plant 2013;17:80–5. Nephrol 2010;25:739–46.
310. Dharnidharka VR, Gupta S. PTLD treatment: reducing the chemo- 328. Englund MS, Tyden G, Wikstad I, Berg UB. Growth impairment at
therapy burden through addition of rituximab. Pediatr Transplant renal transplantation—a determinant of growth and final height.
2010;14:10–1. Pediatr Transplant 2003;7:192–9.
311. Gallego S, Llort A, Gros L, Sanchez De Toledo Jr J, Bueno J, Moreno 329. Gil S, Vaiani E, Guercio G, Ciaccio M, Turconi A, Delgado N, Riva-
A, Nieto J, Sanchez De Toledo J. Post-transplant lymphoproliferative rola MA, Belgorosky A. Effectiveness of rhGH treatment on final
disorders in children: the role of chemotherapy in the era of ritux- height of renal-transplant recipients in childhood. Pediatr Nephrol
imab. Pediatr Transplant 2010;14:61–6. 2012;27:1005–9.
312. Smith JM, Dharnidharka VR, Talley L, Martz K, Mcdonald RA. BK 330. Berard E, Andre JL, Guest G, Berthier F, Afanetti M, Cochat P, Broyer
virus nephropathy in pediatric renal transplant recipients: an analy- M. Long-term results of rhGH treatment in children with renal fail-
sis of the North American Pediatric Renal Trials and Collaborative ure: experience of the French Society of Pediatric Nephrology. Pedi-
Studies (NAPRTCS) registry. Clin J Am Soc Nephrol 2007;2:1037– atr Nephrol 2008;23:2031–8.
42. 331. Dew MA, Dabbs AD, Myaskovsky L, Shyu S, Shellmer DA, Dimartini
313. Momynaliev KT, Gorbatenko EV, Shevtsov AB, Gribanov OG, AF, Steel J, Unruh M, Switzer GE, Shapiro R, Greenhouse JB. Meta-
Babenko NN, Kaabak MM. Prevalence and subtypes of BK virus in analysis of medical regimen adherence outcomes in pediatric solid
pediatric renal transplant recipients in Russia. Pediatr Transplant organ transplantation. Transplantation 2009;88:736–46.
2012;16:151–9. 332. Delucchi A, Gutierrez H, Arrellano P, Slater C, Meneses M, Lopez I.
314. Smith JM, Dharnidharka VR. Viral surveillance and subclinical Factors that influence nonadherence in immunosuppressant treat-
viral infection in pediatric kidney transplantation. Pediatr Nephrol ment in pediatric transplant recipients: a proposal for an educational
2015;30:741–8. strategy. Transplant Proc 2008;40:3241–3.
315. Trydzenskaya H, Sattler A, Muller K, Schachtner T, Dang-Heine C, 333. Claes A, Decorte A, Levtchenko E, Knops N, Dobbels F. Facilitators
Friedrich P, Nickel P, Hoerstrup J, Schindler R, Thiel A, Melzig MF, and barriers of medication adherence in pediatric liver and kidney
Reinke P, Babel N. Novel approach for improved assessment of phe- transplant recipients: a mixed-methods study. Prog Transplant
notypic and functional characteristics of BKV-specific T-cell immu- 2014;24:311–21.
nity. Transplantation 2011;92:1269–77. 334. Tainio J, Qvist E, Vehmas R, Jahnukainen K, Holtta T, Valta H, Jah-
316. Zaman RA, Ettenger RB, Cheam H, Malekzadeh MH, Tsai EW. nukainen T, Jalanko H. Pubertal development is normal in adoles-
A novel treatment regimen for BK viremia. Transplantation cents after renal transplantation in childhood. Transplantation
2014;97:1166–71. 2011;92:404–9.
37 • Kidney Transplantation in Children 669

335. Ghanem ME, Emam ME, Albaghdady LA, Bakr NI, Helal AS, Sadek 341. Kreuzer M, Prufe J, Oldhafer M, Bethe D, Dierks ML, Muther S, Thum-
EE, Sobh MA. Effect of childhood kidney transplantation on puberty. fart J, Hoppe B, Buscher A, Rascher W, Hansen M, Pohl M, Kemper
Fertil Steril 2010;94:2248–52. MJ, Drube J, Rieger S, John U, Taylan C, Dittrich K, Hollenbach S,
336. Foster BJ. Heightened graft failure risk during emerging adulthood Klaus G, Fehrenbach H, Kranz B, Montoya C, Lange-Sperandio B,
and transition to adult care. Pediatr Nephrol 2015;30:567–76. Ruckenbrodt B, Billing H, Staude H, Heindl-Rusai K, Brunkhorst R,
337. Koshy SM, Hebert D, Lam K, Stukel TA, GuttmanN A. Renal allograft Pape L. Transitional Care and Adherence of Adolescents and Young
loss during transition to adult healthcare services among pediatric Adults After Kidney Transplantation in Germany and Austria: A
renal transplant patients. Transplantation 2009;87:1733–6. Binational Observatory Census Within the TRANSNephro Trial.
338. Samuel SM, Nettel-Aguirre A, Hemmelgarn BR, Tonelli MA, Soo A, Medicine (Baltimore) 2015;94:e2196.
Clark C, Alexander RT, Foster BJ. Graft failure and adaptation period 342. Bell LE, Bartosh SM, Davis CL, Dobbels F, Al-Uzri A, Lotstein D, Reiss
to adult healthcare centers in pediatric renal transplant patients. J, Dharnidharka VR. Adolescent Transition to Adult Care in Solid
Transplantation 2011b;91:1380–5. Organ Transplantation: a consensus conference report. Am J Trans-
339. Tai E, Buchanan N, Townsend J, Fairley T, Moore A, Richardson LC. plant 2008;8:2230–42.
Health status of adolescent and young adult cancer survivors. Can- 343. Watson AR, Harden P, Ferris M, Kerr PG, Mahan J, Ramzy MF. Tran-
cer 2012;118:4884–91. sition from pediatric to adult renal services: a consensus statement
340. Harden PN, Walsh G, Bandler N, Bradley S, Lonsdale D, Taylor J, by the International Society of Nephrology (ISN) and the Interna-
Marks SD. Bridging the gap: an integrated paediatric to adult clinical tional Pediatric Nephrology Association (IPNA). Pediatr Nephrol
service for young adults with kidney failure. BMJ 2012;344:e3718. 2011;26:1753–7.
38 Kidney Transplantation in
Developing Countries
ELMI MULLER

CHAPTER OUTLINE Introduction: The Role of Organ Minimum Requirements to Perform


Transplantation in the Developing World Transplantation
Perspectives on Transplantation in the Nephrology
Developing World in Relationship to the Tissue Typing and Laboratory Requirements
World Health Organization Objectives Infectious Disease Control
The Need for Kidney Transplantation in Surgical Requirements
the Developing World
Is There a Role for Donation After Circulatory
Overview of Transplantation Activities in Death in Developing Countries?
Africa
Partnerships and Training Options for
Involvement of Governments African Clinicians
Requirements for Living Versus Deceased Existing Transplantation Outreach Programs
Donor Transplantation in Africa
The High Costs of Dialysis, Transplantation, Training Options for Clinicians
and Ongoing Immunosuppression
Going Forward: Needs and Strategies
Spatial Distribution of Dialysis and
Transplant Centers and Issues of Access
Summary

Introduction: The Role of Despite very limited transplant activity in Africa, suc-
cessful transplant programs have been developed else-
Organ Transplantation in where in the developing world. In Pakistan a large living
the Developing World donor program has been established successfully with
minimal cost to the patient.1 India’s living donor kidney
Africa is the most underdeveloped continent when it comes program expanded rapidly over the past 10 years and now
to transplantation options for patients with end-stage includes deceased donor and liver transplant from liv-
organ disease. Renal failure is a common condition, and for ing donors.2–4 It is clear that there is a large spectrum of
this reason exploring options for renal transplantation on transplant activity in the developing world, ranging from
the continent makes sense. However, starting a transplant places where there is almost no activity at all and where
program is a daunting task and despite previous meetings both living and deceased donation is still very limited, to
driven by the World Health Organization (WHO) in Africa other areas where transplant activity is rapidly expanding
(July 28–30, 2009, Abuja, Nigeria; December 5–6, 2008, and growing.
Bamako, Mali) to discuss these issues, little development Many groups are working on the improvement of trans-
and progress has been made in Africa in this regard over plantation access in the developing world: The Global Alli-
the past few years. The issues in Africa are similar to many ance for Transplantation (GAT) is a partnership between
other parts of the developing world. The Transplantation Society (TTS) and the WHO for the
It is possible to look back on these meetings and try and worldwide promotion of organ donation and transplanta-
explore why progress in the field of transplantation in the tion activities consistent with the principles outlined in
developing world is slow. One reason for this could be that the Declaration of Istanbul, the World Health Assembly’s
these meetings did not provide enough practical tools to the Resolution on Human Organ and Tissue Transplantation,
clinicians who need to drive the process. Another reason and the Madrid Resolution on government accountability
could be that driving a process like transplantation requires to achieve self-sufficiency in organ donation and trans-
a much larger group of people than those involved in these plantation.5,6 Many other members of the international
meetings. transplantation community are trying to be of assistance

670
38 • Kidney Transplantation in Developing Countries 671

to representatives of Africa and the developing world in professional societies, and experts. With respect to the
identifying needs in the pursuit of transplantation, and in development of the practice of deceased organ donation,
obtaining the commitment of governments for support in the WHO endorses a four-step process: (1) adoption of the
attaining this goal. The International Society of Nephrology Critical Pathway for organ donation from deceased per-
and the Transplantation Society both sponsor programs sons; (2) the drafting of a legal framework; (3) the develop-
in the developing world to establish ethical transplant ment of a blueprint of a national system for organ donation
practices. from deceased donors; and (4) collaboration with govern-
Previous examples from Central and Eastern Europe and ment and the private sector for regional, subregional, and
South America have demonstrated the effect of local lead- national implementation.12
ership on the development of organ donation and trans- Ultimately it is medical professionals who are at the
plantation programs. The South East Europe Initiative on crossroads between donor, patient, and recipient. The
Deceased Organ Donation (Macedonia, May 2011) and the practice of transplantation, and especially deceased
Regional Health Development Centre in Organ Donation donor organ transplantation, necessitates a level of trust
and Transplant Medicine in Croatia are two such examples in the transparency and professionalism of the health
of active and successful partnerships between clinicians, system. In addition to the responsibilities of health pro-
governments, and professional societies, which might in fessionals, there is a need for public education to gener-
turn be applied in the African region.7–9 Several profes- ate societal support for transplantation. Finally, there is
sional organizations are currently supporting clinicians an important role for governments in terms of commit-
to approach governments and to advocate for appropriate ment to allocation of resources, proper oversight, and
legislative frameworks and for the allocation of resources the creation of an appropriate normative and legisla-
to transplantation, especially in settings where dialysis tive environment in which transplantation can operate.
availability is rapidly outpacing the development of kidney Engagement with health authorities is therefore appro-
transplantation. priate from the earliest stages of program development.
Furthermore, there is no legitimate transplantation
activity that cannot be examined and monitored, and
Perspectives on Transplantation therefore registries for the surveillance of practices and
in the Developing World in outcomes are critical from the outset of the practice of
organ transplantation.13
Relationship to the World Health In the context of developing health systems it will likely be
Organization Objectives necessary to engage the private sector in the development
of transplantation services; however, such arrangements
The last World Health Organization (WHO) consulta- mandate complete transparency and specific and effective
tion on cell, tissue, and organ transplantation in Africa oversight from health authorities.14 Universal health cover-
occurred in Abuja, Nigeria in 2009. Nearly 10 years age is a current major objective of WHO, with an emphasis
on, especially given shifting economic and demographic on access, quality, and financial protection for all, based on
trends in the region, it is time to reappraise the situation financing systems designed to deliver cost effective services
with respect to organ donation and transplantation in that do not expose the user to catastrophic costs. To achieve
Africa. Total health expenditure in sub-Saharan Africa these goals with respect to the financing of organ transplanta-
is growing by 7% per annum.10 Healthcare expenditure tion, the engagement and commitment of governments will be
is a crucial component of health status improvement in essential.
sub-Saharan African countries. Increasing healthcare As the practice of tissue, cell, and organ transplanta-
expenditure is a significant step in achieving effective tion spreads around the world, there is a greater-­than-
and safe treatment options for patients in the developing ever need for global governance, upholding societal
world. In many countries policymakers are establishing values of the protection of the donor, safety of the recipi-
effective public-private partnerships in allocating health- ent, and self-­sufficiency. An example of a project where
care expenditures.11 Therefore economic growth and cor- vigilance and surveillance are actively encouraged and
responding increases in healthcare expenditures in the driven by the WHO is Project Notify.15 The WHO recog-
African region mean that we can confidently anticipate nizes that there should be crosscutting principles sur-
increased demand for organ transplantation within the rounding medical products of human origin, based on
region over the next few years. The WHO has a role to play global standards and consensus, and supported by global
in fostering these anticipated developments in accordance information standards and surveillance. Such tools will
with the Guiding Principles on Human Cell, Tissue and be of particular importance in the context of emerging
Organ Transplantation. health systems.
The WHO is interested in every region of the globe and The WHO encourages a national strategy for organ
every level of development, not just in countries with donation and transplantation that: (1) promotes the inte-
well-established transplantation programs. The goal of grated management of chronic kidney disease (CKD) from
the WHO is to achieve a common global attitude toward prevention to renal replacement therapies; (2) relies on
transplantation, via a multitude of partnerships with existing guidance and multidisciplinary collaboration with
key bodies, including health authorities, scientific and a more advanced team, through long-term agreement
672 Kidney Transplantation: Principles and Practice

40
Prevalence (pmp)

20

Cote d’lvoire
Swaziland
Congo
Guinea
Somalia
South Africa
Mauritius
Seychelles
Liberia
Togo
Zimbabwe
Gambia
Central African Rep.
Gabon
Angola
Sudan
Guinea-Bissau
Benin
Lesotho
Burkina Faso
Mauritania
Senegal
Morocco
Tanzania
D.R. Congo
Djibouti
Kenya
Chad
Cameroon
Ghana
Tunisia
Ethiopia
Mali
Eritrea

United Kingdom
United States
Niger
Mozambique
Cape Verde
Malawi
Rwanda
Namibia
Sierra Leone
Uganda
Burundi
Nigeria
Zambia
Equatorial Guinea
Madagascar
Botswana
Comoros
Raised blood pressure or on medication (age-standardized, adults 25+)
Raised fasting blood glucose or on medication (age-standardized, adults 25+)
Fig. 38.1 Estimated prevalence (per million population [pmp], age-standardized) of hypertension and diabetes in African countries, compared with the
United States and United Kingdom. (Source: WHO Global Health Observatory.)

100
Mortality rate (pmp)

50

0
Malawi
Guinea
Côte d’lvoire
Somalia
Guinea-Bissau
Ethiopia
Sudan
Namibia
Zambia
Mozambique
Cameroon
Equatorial Guinea
Seychelles
Swaziland
Central African Republic
Djibouti
Ghana
Comoros
Angola
Chad
Benin
Burundi
Sierra Leone
Lesotho
Uganda
Mauritania
Democratic Republic of
Nigeria
Gambia
Congo
United Republic of
Mali
Liberia
Botswana
Togo
Mauritius
Rwanda
Madagascar
Gabon
Senegal
Eritrea
Cape Verde
Niger
Kenya
Zimbabwe
South Africa
Tunisia

United States of America


United Kingdom
Fig. 38.2 Estimated rate (per million population [pmp]) of mortality from nephritis and nephrosis in the population younger than 60 years in African
countries, compared with the United States and United Kingdom. (Source: WHO Global Health Observatory, Global Burden of Disease Study.)

between institutions and health authorities; (3) is mindful enable quantitative estimation of the underlying burden of
of the need for transparency of activities; (4) identifies organ ESKD and its risk factors in the population. Yet although such
donation after death as a long-term objective from the out- data are largely unavailable, many have commented that the
set; (5) pioneers health system development and universal underlying burden of ESKD in Africa is likely to exceed that of
health coverage; and (6) uses donation and transplantation high-income countries. First, because the underlying preva-
as an opportunity to create dynamics in health, and acts as lence of risk factors associated with organ failure is known to
an interface between the health system and the public. be high, given increasing rates of noncommunicable diseases
in the region, in particular diabetes and hypertension, com-
bined with undiminished rates of infection-related nephropa-
The Need for Kidney thies (Figs. 38.1 and 38.2). Second, the nature of the primary
causes of ESKD and the limited capacity for secondary pre-
Transplantation in the vention result in more rapid progression to organ failure
Developing World than experienced in high income countries.17 Glomerular
nephropathies and hypertension are the leading causes of
Although we know that the availability of renal replacement treated ESKD in sub-Saharan Africa, accounting for between
therapy (RRT) is lower in low- and middle-income coun- 18% to 50%, and 25% to 75% of all cases, respectively.18,19
tries, it is realistic to estimate that there is a much greater Although diabetes currently accounts for between 3% and
need than in high-income countries as the true scale of the 24% of treated ESKD in sub-Saharan Africa (compared with
unmet need for treatment of end-stage kidney disease (ESKD) 15%–60% in high-income countries), this proportion may
is unknown.16 Ideally, population-based studies, death reg- increase given projections that the number of adults in Africa
istration data, and dialysis and transplant registries would with diabetes will double by 2030.20–24
38 • Kidney Transplantation in Developing Countries 673

facilities, capacity to achieve acceptable graft outcomes,

Hypertension

Hypertension
cultural and religious attitudes toward organ donation,

Diabetes

Diabetes
trust in the health system, and the extent to which patients
are able to meet the costs of surgery and ongoing immu-

HIV

HIV
nosuppression. Continuing demographic, epidemiologic,
400 and economic shifts will have implications for the future
408 413
incidence of organ failure in the African region and for the
Adult incidence (pmp)

300 329 326 level of demand for high-level health care including trans-
plantation; that is, the capacity of populations in Africa to
200 benefit from transplantation will evolve in coming years. It
179 is therefore timely to evaluate existing capability to deliver
100
117
organ transplantation services in the African region, and to
identify how local efforts, regional cooperation, and inter-
0 national partnerships can best address current constraints
Sub-Saharan Africa United States on the delivery of transplantation therapy.
(African American)
Fig. 38.3 Estimated incidence of end-stage kidney disease in the sub-
Saharan African adult (>25 years) population, by primary cause, based Overview of Transplantation
on cause-specific disease incidence extrapolated from the African
American population.
Activities in Africa
In Africa there are seven countries that are locally perform-
Estimating the burden of ESKD in Africa is relevant ing living-related donor transplantation: Ethiopia, Ghana,
because to effectively advocate for the allocation of Kenya, Nigeria, Sudan, South Africa, and Tunisia. Pro-
resources to organ donation and transplantation, it is nec- grams vary between those that are well established with
essary to demonstrate that there is a need within the pop- larger number of transplants (South Africa, Tunisia, Sudan)
ulation for these services. “Need” may be defined as “the to small programs with a limited number of transplants in
population’s ability to benefit from organ transplantation,” each center (Kenya, Ghana, Nigeria).13,29,30 South Africa
and has three aspects: (1) the underlying burden of organ has the highest rate of organ transplantation relative to its
failure and its risk factors, irrespective of current treatment population (250 living-related donor kidney transplants in
availability or eligibility criteria; (2) the cost and efficacy 2015), followed by Tunisia (122 living-related donor kid-
of treatment (cost will constrain the number of people able ney transplants in 2015) and Sudan (165 living-related
to benefit from transplantation, and transplantation out- donor kidney transplants in 2015). An issue for Kenya
comes must be acceptable) and; (3) comparison to the exist- and Nigeria is that there are many centers simultaneously
ing provision of services.25,26 trying to establish transplantation, with each center per-
The incidence of ESKD in Africa attributable to hyper- forming a limited number of transplants. In Nigeria there
tension and diabetes might be estimated based on the are six state and two private centers, where a total of 14
cause-specific incidence of ESKD observed for the African transplants were performed in 2015, and in Kenya there is
American population. Based on an average adult preva- one state and four private centers, which have performed
lence of hypertension in the African region of 46%, and an a total of 60 transplants in 2015. This creates a problem
incidence of ESKD with a primary diagnosis of hypertension of dispersal of expertise and facilities. None of the African
in the US African American population of 0.7 per 1000 countries except South Africa has yet commenced deceased
hypertensive adults, the incidence of ESKD due to hyper- donor transplantation. Transplant tourism and official
tension in Africa potentially equals 330 cases per million arrangements in which governments send donor-recipient
adults aged >25 years. Similarly, based on an average adult pairs abroad to undergo transplantation are relatively com-
prevalence of diabetes of 9% in Africa and an incidence of mon. When patients travel for transplantation, destination
ESKD with a primary diagnosis of diabetes in the African countries include Tunisia (mostly patients from Cameroon),
American population of 2.1 per 1000 diabetic adults, the Pakistan and India (patients from Nigeria, Rwanda, Kenya,
incidence of ESKD due to diabetes in Africa is estimated at Zambia, and Malawi).
180 cases per million adults. HIV-related CKD is also likely The majority of dialysis in African countries is conducted
to be responsible for a significant burden of ESKD in the in state-run facilities, with the exception of Ethiopia and
African region: at least 400 cases per million population per Tunisia. In Tunisia all transplantation activities are in
year.20,27,28 If we extrapolate rates of progression to ESKD state-run facilities, but the majority of dialysis takes place
as reported for the African American population, then in private facilities. Private facilities contribute significantly
based on the high prevalence of hypertension, diabetes, and to transplant activity in South Africa, Kenya, Nigeria, and
HIV in the African region, the annual incidence of ESKD Sudan, indicating a potentially significant role for public-
potentially exceeds 900 cases per million adults owing to private partnerships in the future development of trans-
these three risk factors alone (Fig. 38.3). plantation in these countries. Cameroon, Malawi, Sudan,
For the subset of this population that might be consid- Tunisia, and Zambia have a central budget dedicated to
ered medically suitable for transplantation, demand for dialysis; similarly, the costs of transplantation and post-
transplantation will be tightly constrained by the avail- transplant care are met by government or by private insur-
ability of specialist physicians and surgeons, pathology ance in Cameroon, Malawi, Rwanda, Sudan, Tunisia, and
674 Kidney Transplantation: Principles and Practice

Zambia. Elsewhere, patient costs are met by out-of-pocket Pediatric transplantation is relatively rare in the develop-
payments. Organ donation and transplantation was said to ing world. Tunisia initiated pediatric transplantation early
be a priority in the further development of the health system in the development of its living donor transplant program,
in many African countries. Legislation pertaining to organ constituting a good model for other countries in recognizing
donation and transplantation is only in existence in South the importance of pediatric transplantation in the develop-
Africa, Tunisia, Sudan, Ghana, Nigeria, and Kenya. ment of a transplantation program. Distance from pediat-
Familiarity of doctors with the Declaration of Istanbul ric kidney transplant centers may be a significant barrier
varies. Africa is a major potential destination for organ traf- in accessing care for patients and families, particularly
ficking networks. The ongoing education of clinicians and because of the lower number of pediatric kidney transplant
health authorities is essential to prevent this practice from centers compared with the number of adult centers in the
moving into Africa. In many other parts of the developing developing world.34,35
world organ trafficking is a major problem, especially in
places where there are fairly good healthcare resources and INVOLVEMENT OF GOVERNMENTS
minimal oversight.31,32
Many existing successful partnership programs are in In several countries, health ministries or individual gov-
place trying to increase access to transplantation in Africa. ernment officials have independently expressed interest in
These programs are being driven from a variety of Euro- pursuing organ transplantation. In Ethiopia, for example,
pean, North American, and South African centers. transplantation is on the government agenda as a result
Partnerships form a major part in developing transplan- of expressed interest from government ministers in trans-
tation services in Africa. Many US-based centers now reach plantation taking root locally, with the training of local
out to Africa to perform global health projects. It is impor- professionals a first priority. In Malawi, the vice president
tant that these partnerships result in education and training was the first person to receive dialysis in the country and
of local clinicians rather than simply going and performing has subsequently become a vocal advocate for transplan-
a few transplants with surgeons from the developing world. tation. Kidney transplantation is considered an aspiration
To build up local practices and infrastructure, the programs of the government of Malawi. Currently the Malawi Min-
need to include local surgeons and physicians in perform- istry of Health has committed to the upgrading of dialysis
ing these procedures and looking after their own patients.33 machines (Several donations had been received including
The importance of regional cooperation for transplan- donations from the Japanese Government and Fresenius.),
tation in developing countries cannot be overstated. For and is actively involved in programs for the screening and
example, in Tunisia 122 living-related transplants were prevention of ESKD in partnership with the International
performed in 2016. All of these were done in six centers in Society of Nephrology (ISN). The government has also
the state sector. This program certainly has the capacity to committed to publicly fund transplantation in the future. In
train other African clinicians in the future. the interim, the Malawi government is continuing to send
A similar situation exists in Sudan where 134 living patients to India at a cost of $30,000 per living donor trans-
donor transplants were performed in the state sector and 31 plant, with patients returning to Malawi with a personal
in the private sector in 2016. Current examples of regional supply of immunosuppression.
cooperation for training include government sponsorship of In 2007, the head of state in Cameroon decided to decen-
nephrology trainees from Rwanda to travel to South Africa tralize dialysis services and open dialysis centers in every
for specialist training. Cape Town has trained 20 nephrol- region of the country. Under this plan, seven centers were
ogists to date: Dr. Mweemba, the first nephrologist in the created between 2007 and 2012, and the number of public
public systems in Zambia, was trained in South Africa and centers was increased to 11 by the end of 2016. Hemodi-
dialysis commenced in Zambia upon his return in 2009. alysis is subsidized; patients pay $10 per session. With the
Training for Zambian surgeons in South Africa is also rapid expansion of dialysis in Cameroon, the government is
planned. South Africa might also provide assistance and eager to move ahead with transplantation, beginning with
training for organ procurement to other African countries a legislative framework. Currently the government of Cam-
in the future. The African Development Bank are establish- eroon pays for patients to undergo transplantation abroad,
ing an Institute of Nephrology and Urology, intended to providing financial aid of $20,000 to $36,000 unless the
train nephrologists and urologists in East Africa. patient has private insurance. Many patients relocate to
Many African patients still travel out of their country to France for transplantation. The experience of Cameroon
receive a transplant. India is currently the most popular suggests that where the government has undertaken to
destination, mostly because of cost-effective packages and fund dialysis, there may be greater incentive to pursue kid-
because renal physicians have relationships with hospitals ney transplantation if transplantation can be demonstrated
in India. It might be useful to send donor-recipient pairs to to be cost saving.
countries within Africa for transplantation, rather than There is also strong political will to support patients with
to India in the future. Currently Tunisia has cooperative ESKD in Zambia. There is currently a separate budget allo-
agreements with Senegal and Cote d’Ivoire to transplant cation for dialysis, new dialysis units are being opened, and
patients from these countries, and there are discussions the government is developing a health insurance scheme
regarding the feasibility of sending more donor-recipient that would cover the costs of dialysis and potentially trans-
pairs to South Africa for transplantation. The relative costs plantation. There is also strong political will to enact legis-
of immunosuppression in Africa versus India will need to be lation with respect to organ donation and transplantation.
addressed before current practice shifts. In addition, 10 regional hospitals had intensive care units
38 • Kidney Transplantation in Developing Countries 675

Data Systems

Post Tx care HLA lab

Deceased Organ
Waiting list donor procurement
transplantation program

Chronic On call team Drug program


Dialysis Minimum requirements ICU
Program

Funding mechanisms

Legislative and Regulatory Framework

Infrastructure/other specialist personnel


Clinical care teams (surgeons, physicians and nursing)
Legislation/regulation, financing and organization of health services delivery
Fig. 38.4 Schematic diagram of the minimum requirements to perform deceased donor organ transplantation, and the interactions between these
requirements.

(ICUs) by the end of 2014, and the University Teaching be contemplated in parallel with the development of living
Hospital has started training two to four ICU physicians and donor transplantation? To what extent is it necessary to
10 to 15 ICU nurses per year since 2015. The government consolidate experience with living donor transplantation
has requested a roadmap for the provision of cost-effective, before commencing deceased donor transplantation? What
sustainable RRT in Zambia and is willing to publicly fund are the minimum requirements in terms of ICU beds and
treatment, contingent on cost. Pubic health insurance is trained personnel?
currently in the pipeline, which will have implications for The experience of Tunisia illustrates the scale of the tran-
the funding of future transplantation programs. sition from provision of living donor transplantation to that
Elsewhere, medical professionals are in the position of of deceased donor transplantation. Despite a well-developed
lobbying governments to advance the transplantation living donor transplantation program, which was estab-
agenda. For example, by the efforts of medical profession- lished in 1986, deceased donor transplantation is not yet
als in Ghana, transplantation legislation had been finalized. available.30 Barriers to the initiation of deceased donor
In addition, in the absence of regulatory oversight and coor- transplantation in Tunisia were identified as an absence
dinating authorities, the University Hospital has established of legislation on brain death and the lack of infrastruc-
an independent ethics team. Medical professionals are also ture, personnel, and capacity for coordination required to
leading negotiations on a cost-sharing scheme between gov- support deceased donation. Whereas living donor trans-
ernments, private funding sources, and patients, to address plantation might be successfully driven by a motivated
the fact that transplant recipients in Ghana must currently individual and in a single institution, deceased donor trans-
meet all costs out-of-pocket. The Nigerian Association of plantation requires dialysis programs, tissue typing and
Nephrology has been in discussions with the government to ­cross-matching facilities, an organ procurement program,
seek coverage of dialysis under the national health scheme, an on-call surgical team, capacity to fund this infrastruc-
although currently the agreement is to cover only six ses- ture, and an appropriate legislative framework (Fig. 38.4).
sions of dialysis. A National Transplant Act had also been Moreover, a significant level of regional/national organiza-
finalized. tion is required: the historical experience of the US was that
deceased donation gained momentum only when it was
REQUIREMENTS FOR LIVING VERSUS DECEASED separated from the hospital and placed under an indepen-
dent coordinating authority. There is also the need to con-
DONOR TRANSPLANTATION
tend with the public perception of deceased donation. It will
A topic of debate in the developing world is the rela- be easier to commence deceased donation in the context of
tive resource requirements associated with living versus an established living donor program with acceptable and
deceased donor transplantation and at what stage it is consistent recipient outcomes. Therefore in advocating for
appropriate to contemplate deceased donor organ trans- deceased donor transplantation, the requirements in terms
plantation. Questions include: What is a sufficient level of of resources, infrastructure, track record, and capacity for
dialysis availability? Should deceased donor transplantation coordination need to be realistically acknowledged.
676 Kidney Transplantation: Principles and Practice

THE HIGH COSTS OF DIALYSIS, geographic area reduces disparities in access to transplan-
TRANSPLANTATION, AND ONGOING tation; on the other hand, the small number of transplant
IMMUNOSUPPRESSION patients at each of these centers makes it more difficult for
clinicians to build up experience.33,37
An issue that is commonly identified as a significant and
recurring barrier to transplantation in the developing
SUMMARY
world is the high cost of transplantation and follow-up
care, in particular the cost of maintenance immunosup- Several important and common factors are noted among
pression.36 In Africa, the costs of both dialysis and trans- transplantation programs that are succeeding in Africa. First,
plantation are often higher as a proportion of personal most of these programs experience some positive government
income per capita health expenditure than they are in involvement. This positive environment results in funding,
high-income countries. First, this has implications for dial- support, and a willingness from the Ministry of Health to work
ysis modality utilization: peritoneal dialysis (PD) is typi- on a legislative framework. Second, these places have cham-
cally more expensive and less profitable than hemodialysis pions or individuals driving dialysis and transplantation—a
(HD) in Africa given the high cost of consumables, and it is clinician who can see the need for these programs and who
therefore rare, especially where dialysis provision is driven is willing to make the effort. This is often much better than a
by the private sector (e.g., Ethiopia). Second, the high cost Minister of Health or other official who decides transplantation
of immunosuppression erodes the large cost savings asso- should be available in a given country.
ciated with transplantation versus dialysis that are typi- Issues around training and expertise remain an impor-
cally observed in high-income countries. In Tunisia for tant concern in the developing world. The wide dispersion
example, where dialysis and transplantation are both pub- of expertise, by allowing many centers in one country to
licly funded for all patients, kidney transplantation costs develop simultaneously might be a problem (Nigeria and
$16,000 in the first year and $10,000 per year in subse- Kenya). Another important issue is how to address “brain
quent years, compared with a maintenance dialysis cost drain” from the developing world. Many physicians and
of $12,000 per year. Third, the high cost associated with scientists who leave their native countries to pursue work
ongoing immunosuppression will result in catastrophic elsewhere never return.
costs to patients where the costs of transplantation are Funding for dialysis versus transplantation needs further
being met out-of-pocket. debate and exploration. Common questions raised include:
Even for countries that are not yet performing trans- Do you need a dialysis program to start a transplant pro-
plantation within their own borders, the cost of immu- gram? How do you evolve a successful dialysis program into
nosuppression is an issue. For example, in Cameroon the a successful transplantation program? How do you distrib-
government pays for patients to travel abroad to receive ute the funding for these different programs? Lessons can be
transplantation, but when patients return they must learned from the successful dialysis programs in Africa and
contend with the high cost of immunosuppression, and how these have been established. Examples of public private
this has resulted in the government currently negotiat- partnerships exist in many places; for instance, between
ing for lower cost generics from India. In response to this Fresenius Healthcare and the South African government in
issue, it will be necessary in the future for international Polokwane.
professional bodies to join with African governments For developing countries that plan to start living-related
and medical professionals to lobby for access to cheaper donation or have already commenced living donation,
immunosuppression. the transition to a deceased donation program remains a
problem. Deceased donation needs a different set of infra-
SPATIAL DISTRIBUTION OF DIALYSIS AND structure from living donation; for example, a tissue-typing
TRANSPLANT CENTERS AND ISSUES OF ACCESS laboratory with trained staff and after-hours services as
well as an appropriate legislative and regulatory environ-
Unsurprisingly, the majority of dialysis centers, and trans- ment. Deceased donation can only develop on the basis of
plant centers where these exist, tend to be located in major a well-established dialysis program and requires a signifi-
urban centers or capital cities, with implications for access cant waiting list of patients to function. Furthermore, many
to treatment. Attempts have been made to address this African countries will need a change in public perception
issue in Nigeria, where the size and diversity of the country around deceased donation for this to succeed. Examples
mean that patients cannot be expected to travel long dis- of how to address these problematic multicultural aspects
tances to receive treatment. Although 20 of 76 of dialysis and societal issues might be found in South Africa, the
centers are located in Lagos, centers have also been estab- only country performing deceased donation on the African
lished across a range of geographic areas. There was a plan continent.
to fund greater distribution of dialysis centers around the Some clinical and policy issues need further exploration
country; however this did not go ahead. A total of eight in the future, in particular:  
centers located in various regions are currently perform- □ Does it make sense to have more stringent criteria for liv-

ing living donor kidney transplantation in Nigeria, but, ing donors in the developing world as donor follow-up
individual center volumes are low. The dispersion of trans- and access to specialists are a problem?
plantation activities in Nigeria highlights potential trade □ How can developing countries address the need for cost

offs between access and volume for emerging transplant effective immunosuppression?
programs (Nigeria commenced transplantation in 2000). □  Is it possible to lobby for cheaper but effective immuno-
On the one hand, eight transplant centers across a wide suppression in the developing world?
38 • Kidney Transplantation in Developing Countries 677

Minimum Requirements to streamline organ allocation and prevent prolonged isch-


emic time: for example, calculated panel reactive antibodies
Perform Transplantation (PRA) combined with unacceptable antigens. This would
mean a virtual crossmatch, where the presence of donor-
NEPHROLOGY
specific antibodies would predict a positive crossmatch.
From a nephrologic perspective, the major problem in the Tissue-typing laboratories are still not established in
developing countries is the limited number dialysis slots Africa and other developing countries, and good pathology
and the high cost to sustain chronic dialysis program.38–40 training programs do not currently exist. However, for Afri-
In many countries dialysis is not funded by the state sector, can countries performing living-related donor transplants
and the fact that patients pay for dialysis is likely to result only, it should be an option to establish one or two central
in inadequate dialysis. In many ways transplantation will high-throughput laboratories on the continent based on
provide a good quality of life and better clinical outcome for regional collaboration, where local expertise is pooled and
the same or less money. Therefore these countries will be shared. This would also be a viable option for countries with
much better off to push transplantation programs rather small transplant numbers. Tissue typing could be tailored
than dialysis programs. However, from a nephrologic point to the local setting, but it also should be possible to build
of view those countries who start transplantation programs up expertise and to gradually build up more extensive tissue
should have the ability to dialyse transplant candidates pre- typing facilities. At the moment desensitization protocols
operatively and immediately postoperatively as a minimum are unlikely to be undertaken in Africa, so basic tissue typ-
requirement. ing might be done locally with outsourcing of some of the
To start a transplant program, there are other minimum more complicated tests to other centers. However, because
service requirements such as pathology, radiology, and sur- of frequent transfusion many people might be sensitized,
gery, and these are not always available. and it is for this reason that expertise in desensitization
Another major concern is the cost and sustainability of protocols needs to be developed in tissue typing laborato-
immunosuppression for transplanted patients. Undertaking ries in Africa. Solid-phase assays also need to be tested in
transplantation without regular and affordable availabil- the ­heterogenous African population. More experience is
ity of immunosuppression is a problem.41,42 For a patient required in this regard. The goal here might not be optimal
to import immunosuppression for himself or herself is not results, but rather cost-effective and reasonably safe trans-
a viable option in the long run, and doctors in developing plantation practices.
countries will have to have a system where these drugs are
freely available at a reasonable price to the patient before INFECTIOUS DISEASE CONTROL
transplantation programs can be started.
In terms of donation practices, much work will need to Infectious disease forms an important part of transplanta-
be undertaken to engage public opinion, particularly in the tion and the scope of infectious issues in solid-organ trans-
context of deceased donation. Living donation seems to be plant is very large. Infectious issues are present both in
a viable and easier option in many countries where there terms of screening and vaccination before transplantation
are cultural barriers toward transplantation. However, one and in prevention of disease posttransplantation. Infections
advantage of starting fresh transplant programs in develop- are the most common posttransplant complication in the
ing countries is that there is an opportunity to make peo- developing world. The rate of graft loss and hospitalization
ple aware from the outset of good ethical practices around because of infective complications in a country like Paki-
organ donation and the Declaration of Istanbul. stan is extremely high.46 This includes bacterial infections
and reactivation of diseases like tuberculosis. There are also
TISSUE TYPING AND LABORATORY issues with donor-derived infections like Chagas, Strongyloi-
des, schistosoma, malaria, and Babesia, which are important
REQUIREMENTS
in low economic settings.47–52 Standard screening includes
The laboratory requirements for living donor transplan- the facilities to test for HIV, hepatitis B and C, cytomega-
tation are significantly different from those for deceased lovirus (CMV), syphilis, varicella, Epstein-Barr virus, mea-
donor transplantation. In terms of minimum laboratory sles, mumps, rubella, toxoplasmosis, and tuberculosis in all
requirements, the question is whether people are suitable recipients. Donors should probably complete similar screen-
for transplantation if they are ABO matched and have ing. In highly endemic areas it might be also necessary to
negative donor-specific human leukocyte antigens (HLA) screen for tuberculosis.53 In terms of vaccination, hepatitis
antibodies, or whether they should be HLA matched as B, pneumovax, tetanus, and yellow fever vaccinations are
well.43–45 In living donation, ABO typing and matching are strongly recommended preoperatively.54 Postoperative
essential. In terms of HLA typing, a combination of cross- prophylactic therapy should probably include antivirals,
match and solid-phase assays are available and probably trimethoprim/sulfamethoxazole (TMP/SMX), antifungals,
ideal. One factor that will affect outcomes is the presence and tuberculosis, Strongyloides, and malaria prophylaxis.55
of donor-specific HLA antibodies. Well-matched donors and Some emerging and reemerging diseases in Africa are
recipients certainly have better outcomes than completely not controllable or treatable in the immunosuppressed
mismatched patients. In the case of deceased donation patient, and higher rates of parasitic disease might occur in
these tests are required on an on-call basis. Typing of all these patients than elsewhere. Tuberculosis is also highly
donors for A, B, Bw, C, DR, and DQ by molecular method, prevalent in Africa, therefore tuberculosis chemoprophy-
and the availability of solid-phase assays would be ideal in laxis and screening is important. Optimal duration of pro-
this setting. Furthermore, a strategy should be available to phylactic therapy varies widely and should be tailored to
678 Kidney Transplantation: Principles and Practice

each individual region. CMV is one of the most common circulatory death is more easily accepted than brain death,
posttransplant infections and although there is high sero- and DCD has fewer resource requirements compared with
positivity in Africa, it is still important to manage CMV, as donation after brain death (DBD). Family consent tends to be
the effect posttransplant is significant. Comorbidities also higher for DCD because brain-dead donors still have a beat-
increase risk of infection, and these might be more prevalent ing heart, which is harder for the family to understand.63,64
in low economic settings (e.g., uncontrolled diabetes).56–59 In high-income countries, DCD is complementary to DBD,
Infection control is essential to good transplant outcomes because there are plenty of ICU beds. In low-income coun-
and prevention of the spread of resistant infections. One tries this might be the opposite given the lower resources
solution with demonstrated efficacy might be more alcohol- requirements.
based hand sanitizers. The current experience in South Africa with DCD could
be a template for many other developing countries. In Cape
Town there is no machine perfusion and only Maastricht
SURGICAL REQUIREMENTS
type III donors are used. If consent from a potential DCD
Certain basic surgical requirements are required to per- family member is obtained, ventilation is stopped after the
form organ transplantation; these are probably equivalent team has prepared the operating theatre. The warm isch-
to those for general surgery or orthopedic surgery. At a emic time is about 20 minutes. A potential advantage for
bare minimum, a retractor is probably essential, but aside DCD in the developing world is that consent might be easier
from this device to facilitate the extraperitoneal approach, to obtain because it is more acceptable to give consent to
a routine kidney transplant could be done with a general remove organs from a body that looks dead (as in the case
laparotomy set. Laparoscopic donor nephrectomy has been of DCD) rather than a body where circulation is still intact.
recommended as a strategy to minimize the overall health In countries where there is a racial split in willingness to
and financial cost to the living donor, allowing decreased donate after brain death, perhaps DCD would be more
morbidity, length of hospitalization, and faster return to acceptable across the community. Cultural beliefs that the
work. However, the cost of staples and other laparoscopic donor will be angry if they donate the organs or fears of
consumables used in a standard technique is prohibitively alienating the ancestor might influence the family’s deci-
expensive, and therefore open donor nephrectomy is prob- sion to donate the organs. There is a generational divide
ably a better option in low-income countries. in these beliefs, so it might be a sensible response from the
An anesthesiologist who is dedicated to transplantation transplant community to respond to this complicated situa-
is essential, because this person needs to be familiar with tion in developing countries by developing all three types of
transplant-friendly muscle relaxants, vasoactive drugs, and donation (DBD, uncontrolled DCD, controlled DCD). In the
anesthetic agents. Essential monitoring equipment, airway UK, controlled DCD has been successful in increasing the
equipment, and intravenous and monitoring catheters number of donors. In addition, an uncontrolled DCD pro-
(more rigorous for pediatric transplant) should be avail- gram has been established in the UK for which the biggest
able at all centers. It is also preferable to have an ultrasound challenge is managing the family.65
available to look for collections and hydronephrosis and to
possibly perform Doppler to look at inflow of the kidney after
transplantation. Partnerships and Training
In general, the surgical aspects of kidney transplantation Options for African Clinicians
are hugely important; however, they are not the rate-limiting
step. Multiple programs in resource-limited countries have EXISTING TRANSPLANTATION OUTREACH
demonstrated excellent initial results; it is long-term follow- PROGRAMS IN AFRICA
up (and necessary resources) that is a significant problem.
Successful programs have started with living donors and Several partnerships currently exist in Africa, and individ-
only after establishment of a successful program have they ual institutions or clinicians drive some of these partner-
slowly transitioned to deceased donation. Most have started ships. One outstanding need is to document and monitor
with surgeons trained at large transplant center with contin- these partnerships to ensure actual needs are being met
ued support from a sister institution. where limited training has been provided. The ISN has
several core programs of global outreach—fellows, educa-
IS THERE A ROLE FOR DONATION AFTER tional ambassadors, sister center programs to develop local
CIRCULATORY DEATH IN DEVELOPING clinical services. TTS now partners with ISN to support or
partly fund transplant center sister programs.
COUNTRIES?
Examples of existing individual partnership programs in
A good option for African countries wishing to start the African region are as follows:   
deceased donor transplant programs is transplantation □ The Rhode Island based Brown University (US) has an
after donation after circulatory death (DCD). DCD graft affiliation with Moi Teaching and Referral Hospital in
survival in Spain is 80% at 10 years.41,60–62 Definition of Kenya that has been running for 4 years.
death may be irreversible cessation of brain function, but □ Transplant Links (Birmingham) had been working in Trin-
the most common way in which death is determined in idad, and in November will start a program in Jamaica.
the developing world is the irreversible loss of circulatory They have also given advice in Kenya and Zambia. They
function. Because this is the most well-established defini- have more-established links with Ghana and Nigeria.
tion of death, it is more likely to be acceptable and requires □ The University of Michigan has been working with St.
less of a ­cultural/­normative/legislative shift. The concept of Paul’s Millennium Medical College in Ethiopia.  
38 • Kidney Transplantation in Developing Countries 679

One example of a successful model of a partnership pro- training is necessary to learn the practical surgical meth-
gram is the Oxford-Belarus program. Belarus had limited ods. In Mauritius in the 1980s and 1990s there was an
access to dialysis and transplantation at the end of the established dialysis program, with patients traveling to
1990s and early 2000s.66 This program started with joint France and South Africa for transplants. A group from
living donor transplants, and an agreement was signed Cape Town went to Mauritius to train local surgeons. They
with the government to continue doing joint transplants did the first three transplants, assisted on the next few, and
with a government commitment to fund these procedures then the local team did the last few on their own. Now Mau-
in the future. ICU staff were brought together for a meeting ritian transplant surgeons are self-reliant.67
on brain death and donor identification; subsequently, the It is important that projects be undertaken with long-
government committed to appointing local transplant coor- term vision, focused on putting and keeping transplanta-
dinators. Publicity was also part of the partnership—getting tion on the agenda, engaging with politicians, with the
transplantation onto national television with positive sto- view to being engaged for the long term, and to allow the
ries. From eight transplants performed in Belarus in 2005, natural pace of program development. There is a need to
there were 201 transplants performed in 2011, nearly engage ministries, not just clinicians, but to do this success-
all from deceased donors. Since the initial partnership for fully you first need a local champion—a constituent within
pediatric transplantation and first pediatric transplants in the country with a link to government—and sustained gov-
2009, there have been 60 transplants. The average waiting ernment support after the partnership team has left.
time for a pediatric transplant in Belarus is now 6 months
(down from 17 years). Another positive outcome is that the TRAINING OPTIONS FOR CLINICIANS
transplant surgeons who trained in Belarus are now train-
ing surgeons in Kyrgyzstan and Kazakhstan. This domino Starting a transplant program in a developing country
effect within the region is a lesson for Africa. is more than having a local champion. It requires a local
The Transplant Links program has been working in Trin- champion who is skilled in transplantation and has been
idad and Jamaica. They have also given advice in Kenya trained very well in a highly experienced center, not just
and in Zambia, and have established links with Ghana and someone who wants to collaborate with teams from over-
Nigeria. The Transplant Links model is one where financial seas. Training for transplantation is available in many
support for skills transfer are provided, but the costs of trans- places throughout the world. Different requirements/pro-
plantation need to be met through local economic means. grams are in place in different countries with respect to
The idea is that the long-term sustainability of transplanta- observational versus hands-on training. See Table 38.1 for
tion relies on emergence of a local model of financing. a list of US training programs.
Challenges noted from existing partnerships with indi- For potential trainees considering overseas training, the
vidual centers in the developing world include: the limited numbers of transplants performed in a given center is an
availability of necessary surgical instruments for visiting important consideration in terms of the depth of experi-
surgeons (e.g., microvascular instruments), poor long-term ence they are likely to receive. In Brazil, surgeons can train
transplant outcomes because of the inability to meet the in organ procurements, and trainees would deal with two
costs of maintenance immunosuppression, a lack of moni- potential donors and see about 200 to 300 patients in the
toring and surveillance of transplant outcomes (“you can’t outpatient clinic per day. There is also a busy tissue-typing
improve what you don’t measure”), the absence of a sur- laboratory. Surgeons in Brazil do not undertake laparo-
gical champion of transplantation, and limited local histo- scopic donor nephrectomies, they do a short open incision.
logic capacity. Another issue is the difficulty in establishing It is less expensive and patients still leave hospital in 2 days.
tertiary care in settings where traveling to another country South Africa has a similar experience.
to receive high-level medical care is standard practice. On Medical training in the UK is divided into the foundation
the ground fact-finding is an important first step in estab- years, core training, specialist training, and postspecialist
lishing a sister link to determine whether linkage is likely training fellowships. Postspecialist training fellowships are
to be successful, the likely major challenges, the resources the best opportunity for doctors from developing countries
required as a priority, and the needs of the population (e.g., coming to the UK. In Australia there are six to seven larger
how many patients are on dialysis). It is also important transplant programs with almost no vacancies for training
for the success of sister programs that partnership teams posts. Hands-on training is difficult for unregistered foreign
become close and establish camaraderie. doctors to obtain; barriers to registration are significant,
One of the strengths of training in South Africa is that although observation is possible. Australia’s main respon-
hands-on training can be offered, because the Health Pro- sibility is in education and research, and potentially in pro-
fessions Council allows African and overseas trainees to viding support for aspects other than surgery (e.g., support
register on a temporary basis for clinical practice without to establish organ donation and transplantation registries,
the need for additional examinations. Initial hands-on and other infrastructure building).

TABLE 38.1 Training Options for Clinicians Traveling to the USA


Very Short Visit Short-Term Visit One-Year Stay Formal Training
Duration 1 month 3 months 1 year 2 years
Type of training Enhanced clinical experience Clinical observation Clinical research fellowship Clinical fellowship
680 Kidney Transplantation: Principles and Practice

In the future, a useful resource would be to list interna- infectious disease control). A summary of these key areas is
tional opportunities on a central website (for instance TTS/ attached in Table 38.2.
ISN) and make sure people know about opportunities to There is an unequivocal need for transplantation ser-
obtain training in the US. For example, at any one time 20% vices throughout the developing world. These observations
to 30% of training positions in the US are unfilled. require government/national health authority/ministerial
attention. Professionals must be aligned to target the gov-
ernment and advocate for support for the provision of trans-
Going Forward: Needs and plantation services within the country. Efforts to develop
Strategies transplantation require a legislative mandate: several par-
ticipating countries reported an absence of satisfactory/
The local needs that must be met for organ donation and comprehensive legislation that would place the account-
transplantation to develop in low-income countries are ability for transplantation activities with the national
significant. It is important to identify short- and long-term health authority. This issue of legislation and government
strategies through which these needs would most appropri- accountability therefore becomes a clear objective of the
ately be met. In most countries that are setting up in trans- WHO and other professional organizations. It also has
plantation it is possible to identify specific areas of concern, become evident that there must be a champion to facilitate
for which no immediate response might be evident or where engagement of the health ministry and that the champion
the scope of the issue is beyond organ donation and trans- must be a credible medical professional with a commit-
plantation (e.g., health system organization and financing, ment to organ donation and transplantation. Ministries

TABLE 38.2 African Country Reports: Needs, Strategies, and Areas of Concern With Respect to the Development of Organ
Transplantation
KENYA
NEEDS STRATEGIES
1. Human capital: surgeons, anesthetists and pediatric 1. Encourage young surgeons to be interested in transplant surgery, and better
nephrologists salaries for those working in transplantation (transplant surgery is not lucrative,
2. A closer tissue-typing laboratory (currently outsourcing especially in the public sector where the costs of transplantation are artificially kept
tissue typing to South Africa, meaning a 2-week delay down)
and no recourse if there is a problem with the results) 2. Histology training and development of telemedicine
3. Laws covering deceased donor transplantation (current 3. The Kenyan government partially subsidizes the cost of transplant for those trans-
laws in Kenya cover living donor transplantation only) planted in public hospitals (2/3); advocate for government to prioritize funding for
4. A registry or means of surveillance and monitoring of transplantation
practices and transplant outcomes (currently have a 4. Educate parents on the benefit of pediatric transplantation
template, a challenge will be to harmonize with the 5. Build up a center of excellence that performs a large volume of transplants to sup-
private sector) port emerging centers, rather than a reliance on visiting surgeons
5. To begin to consider deceased donor transplantation 6. African Development Bank is setting up a nephrology and urology center in Nai-
6. Growing need for matching and greater organization robi, explore potential for leverage
of transplant services 7. Assign specific responsibilities among Kenyan medical professionals working in
OTHER AREAS OF CONCERN transplantation to push forward the agenda
□ There is a need to reconcile public and private sector 8. Halt the practice of Indian brokers poaching patients from the transplant waiting
provision of transplantation so that the two sectors list by demonstrating that treatment in Kenya is associated with better outcomes
are no longer in competition and so patient access and 9. Develop a 5-year plan for transplantation in Kenya
outcomes are optimized 10. Explore public/private partnerships
□ Have not started transplanting other organs, but are 11. Form a Kenyan Transplant Society
looking toward pediatric liver transplants
GHANA
NEEDS STRATEGIES
1. More experience for transplant professionals 1. Transplant Links would be prepared to increase their visits to give local surgeons
2. More transplant surgeons more experience, with ancillary support from Trinidad and Tobago and Nigeria
3. Government support 2. A surgical trainee has been identified who will undertake training in Birmingham
4. Transplant coordinators 3. Support from South Africa to train a coordinator
5. Tissue typing laboratory 4. Financial support from TTS and ISN to fund the training of a surgeon and a trans-
6. A patient advocate for transplantation in Ghana plant coordinator
7. Finalize legislation (draft legislation currently before
parliament)
ETHIOPIA
NEEDS STRATEGIES
1. Training of transplant professionals, particularly surgeons 1. Legislation relating to organ transplantation will be through parliament shortly
2. Histopathology and radiology facilities 2. No one with training in pathology is currently available to read transplant biopsies,
3. Better availability of medicines and therefore will need to outsource to private or university pathology labs in
OTHER AREAS FOR CONCERN South Africa or a closer location, with a longer-term strategy to increase local
□ Measuring CNL is also a challenge, although the govern- capacity for pathology with the potential support of ISN
ment has committed to supplying resources for this 3. University of Michigan will conduct sandwich training of professionals in the short
□ Malaria prophylaxis term; in the longer term will be necessary for surgeons to seek longer fellowships,
□ Ensuring a sufficient supply of immunosuppressive medi- potentially with the support of TTS
cations for induction will require negotiations

Continued
38 • Kidney Transplantation in Developing Countries 681

TABLE 38.2 African Country Reports: Needs, Strategies, and Areas of Concern With Respect to the Development of Organ
Transplantation—cont’d
MALAWI
NEEDS STRATEGIES
1. Patient management 1. The first living donor transplant will be timed to coincide with the return of the first
2. Availability of drugs trainee who will head this service in the future
3. Therapeutic drug monitoring 2. Medical professionals consistently report back to government to keep them mind-
4. Operative, anesthetics, and recovery facilities ful of transplantation
OTHER AREAS FOR CONCERN 3. A possible strategy for cross-matching and tissue typing may be to access pathol-
□ Ethical issues ogy services through Wellcome Trust, which is active in Malawi
□ Cost: dialysis costs 27,000 GBP per year; a 1-month 4. Learn from neighbors: for example, relevant legislation was developed based on
supply of generic cyclosporine costs 1000 GBP organ donation and transplant legislation previously developed in Namibia

NIGERIA
NEEDS STRATEGIES
1. Training and exposure: need for more cooperation 1. Training: level of development, size, and diversity of the country would suggest
within Nigeria itself and between countries much greater needs than others. Training could be both in the form of reverse
2. Government support fellowships to encourage the growth of local training programs and international
3. Lower cost immunosuppression fellowships to locations where hands-on experience is possible
4. Assistance with tissue typing and drug monitoring 2. Advocacy to government for a legal framework, reduction in duty and other taxes
5. Deceased donor organ transplantation on immunosuppression, support for the various transplant programs and patients
(via subsidies for drugs and other consumables). Recommendation to identify 3–4
local public programs for direct government support
3. Lobby international drug companies to reduce the costs of drugs to developing
nations (akin to HIV and malaria)
4. Support the local societies, hold local conferences and subsidize fees to interna-
tional congresses
5. Address the fragmentation of the transplantation program across eight centers:
might suggest that government actively supports a smaller number of centers
over a wide distribution to build these up as focal centers from which to establish
expertise and to expand activities
RWANDA
NEEDS STRATEGIES
1. Funding for dialysis and clinics: currently there is a gap 1. Have applied for a sister site arrangement with Yale
in government support although the government has 2. Regarding living donation: plans to streamline international laboratory links, seek
committed to recommence funding assistance to set up own laboratory services, and train a transplant coordinator
2. Workup of donor: currently cross-matching is performed 3. Regarding pathology, will establish links to an international pathologist
in the UK 4. Surgical training options planned
3. Transplant coordinators 5. International links have been made; now need to follow through for transplanta-
4. Transplant surgeons tion practices to grow
SUDAN
NEEDS STRATEGIES
1. Need for a larger number of trained surgeons (currently 1. Need to consider deceased donor transplantation to address the large pool of
only two senior, trained in UK, and four junior surgeons, dialysis patients
trained in Pakistan and Egypt, who move from one center
to another to perform transplant operations)
2. Experiencing difficulty with retention of medical staff,
and need to establish a more cohesive, consistent team
3. Better quality control of HLA cross-matching
4. More nephrologists and pathologists
5. Need to expand on knowledge and exposure for medical
professionals
ZAMBIA
NEEDS STRATEGIES
1. Legislature with respect to both living donor and 1. Work on draft legislature with reference to templates from other African countries
deceased donor transplantation 2. In correspondence with University of California, will identify a pathologist for train-
2. National transplant coordinator for living donor tran­ ing in histology
splantation—potentially attached to South Africa 3. Support from TTS/ISN for training and assistance with linkages in, for example, tissue
3. Capacity to conduct prescreening for infections typing
4. Capacity for tissue typing and cross-matching
5. Surgical transplant expertise: urologist for open donor
nephrectomy and early postsurgical complications, and a
transplant surgeon for the recipient
6. Capacity for postoperative medical management of the
recipient, supported by cost effective protocols
7. Transplant nurses for patient follow-up
8. Histopathology/pathologist
9. Capacity for drug monitoring
682 Kidney Transplantation: Principles and Practice

ultimately addressing the epidemic of renal failure at the 18. Bamgboye EL. End-stage renal disease in sub-Saharan Africa. Ethnic-
local level might be greatly influenced if CKD were to be ity Dis 2006;16(Suppl 2):S5–9.
19. Bamgboye EL. Barriers to a functional renal transplant programme in
elevated into the realm of the noncommunicable disease developing countries. Ethnicity Dis 2009;19(Suppl 1):S56–9.
priorities of the WHO, setting aside the notion that kidney 20. Naicker S. End-stage renal disease in sub-Saharan Africa. Ethnicity
transplantation is an esoteric treatment for the rich. Dis 2009;19(Suppl 1):S13–5.
Whether transplantation is delivered in the public or pri- 21. Naicker S. Burden of end-stage renal disease in sub-Saharan Africa.
Clin Nephrol 2010;74(Suppl 1):S13–6.
vate sector, the “boat” of transplantation creates a ripple 22. Nanditha A, Ma RC, Ramachandran A, et al. Diabetes in Asia and
effect in its wake that has broader societal value and effects the Pacific: implications for the global epidemic. Diabetes Care
on the overall quality of health care, organization, and 2016;39(3):472–85.
delivery of health services. There is a natural sequence in 23. Sathish T, Oldenburg B, Tapp RJ, et al. Baseline characteristics of
which transplantation programs are established, with dial- participants in the Kerala Diabetes Prevention Programme: a cluster
randomized controlled trial of lifestyle intervention in Asian Indians.
ysis leading to living donor transplantation, to the emer- Diabet Med 2017;34(5):647–53.
gence of deceased donor transplantation once those two 24. Shaw JE, Sicree RA, Zimmet PZ. Global estimates of the prevalence of
initial modalities are sufficiently developed. The availabil- diabetes for 2010 and 2030. Diabetes Res Clin Pract 2010;87(1):4–14.
ity and cost of immunosuppressive medications is a critical 25. Bowie C, Beck S, Bevan G, Raftery J, Silverton F, Stevens A. Estimat-
ing the burden of disease in an English region. J Public Health Med
issue for the further development of transplantation in low- 1997;19(1):87–92.
income countries. 26. Stevens AD, Raftery JP. Health care needs assessment: The epidemio-
logically based needs assessment reviews. 2nd ed. Oxford: Radcliffe
References Medical Press; 1996.
1. Rizvi AH. Adibul Hasan Rizvi: Director, Sindh Institute of Urology and 27. Naicker S. End-stage renal disease in sub-Saharan and South Africa.
Transplantation (SIUT), Pakistan; Founder of the largest free kidney Kidney Int Suppl 2003;(83):S119–22.
transplant programme in Asia. Transplantation 2015;99(9):1749–50. 28. Naicker S, Aboud O, Gharbi MB. Epidemiology of acute kidney injury
2. Agarwal SK, Dash SC, Mehta SN, et al. Results of renal transplanta- in Africa. Semin Nephrol 2008;28(4):348–53.
tion on conventional immunosuppression in second decade in India: a 29. Manjomo RC, Mwagomba B, Ade S, et al. Managing and monitoring
single centre experience. J Assoc Physician India 2002;50:532–6. chronic non-communicable diseases in a primary health care clinic,
3. Malhotra S, Sibal A, Bhatia V, et al. Living related liver transplanta- Lilongwe, Malawi. Pub Health Action 2016;6(2):60–5.
tion for biliary atresia in the last 5 years: experience from the first liver 30. El Mhamdi S, Soltani MS, Haddad A, Letaief M, Ben Salem K. New
transplant programme in India. Indian J Pediatr 2015;82(10):884–9. criteria and quality of health care services in the governorate of
4. Pamecha V, Borle DP, Kumar S, et al. Deceased donor liver trans- Monastir, Tunisia. East Mediterr Health J 2010;16(1):107–12.
plant: experience from a public sector hospital in India. Indian J 31. Fatima H, Fatima Qadir T, Moin A, Bilal Pasha S. Pakistan: a trans-
Gastroenterol 2018;37(1):18–24. Available online at: https://doi. plant tourism resort? J Public Health (Oxf) 2017:1.
org/10.1007/s12664-017-0801-1. 32. Okafor UH. Transplant tourism among kidney transplant patients in
5. Chapman JR, Groth CG, Wood KJ. The needs for a global alliance for Eastern Nigeria. BMC Nephrol 2017;18(1):215.
transplantation. Clin Transpl 2005:273–9. 33. Ulasi II , Ijoma CK. Organ transplantation in Nigeria. Transplantation
6. Vajdic CM, van Leeuwen MT, Turner JJ, et al. No excess risk of fol- 2016;100(4):695–7.
licular lymphoma in kidney transplant and HIV-related immunodefi- 34. McCulloch MI, Gajjar P, Spearman CWN, et al. Overview of a pae-
ciency. Int J Cancer 2010;127(11):2732–5. diatric renal transplant programme. Samj South African Med J
7. Gopcevic A, Vucic M, Lovrencic-Huzjan A, et al. Increased donor 2006;96(9):955–9.
rate at Sestre Milosrdnice University Hospital. Acta Clin Croat 35. Cao B, Adler JT, Bababekov YJ, Markmann JF, Chang DC, Yeh H. Pedi-
2009;48(3):319–24. atric kidney transplantation and mortality: distance to transplant
8. Spasovski G, Busic M, Pipero P, et al. Current status of transplantation center matters. Pediatr Transplant 2018;22(2). Available online at:
and organ donation in the Balkans—could it be improved through the https://doi.org/10.1111/petr.13120.
South-eastern Europe Health Network (SEEHN) initiative? Nephrol 36. Walker AR, Wadee AA. Health care cost in Africa. J R Coll Physicians
Dial Transplant 2012;27(4):1319–23. Lond 2000;34(4):403–4.
9. Zivcic-Cosic S, Busic M, Zupan Z, et al. Development of the Croa- 37. Okafor UH. Kidney transplant in Nigeria: a single centre experience.
tian model of organ donation and transplantation. Croat Med J Pan Afr Med J 2016;25:112.
2013;54(1):65–70. 38. Etheredge H, Fabian J. Challenges in expanding access to dialysis
10. Novignon J, Olakojo SA, Nonvignon J. The effects of public and private in South Africa-expensive modalities, cost constraints and human
health care expenditure on health status in sub-Saharan Africa: new rights. Healthcare (Basel) 2017;5(3):pii:E38. Available online at:
evidence from panel data analysis. Health Econ Rev 2012;2(1):22. https://doi.org/10.3390/healthcare5030038.
11. Paruk F, Blackburn JM, Friedman IB, Mayosi BM. National expen- 39. Joob B, Wiwanitkit V. Cost analysis for renal dialysis: case study from
diture on health research in South Africa: what is the benchmark? Indochina. Saudi J Kidney Dis Transplant 2017;28(4):948.
South African Med J 2014;104(7):468–74. 40. Rocha MJ, Ferreira S, Martins LS, et al. Cost analysis of renal replace-
12. Dominguez-Gil B, Delmonico FL, Shaheen FAM, et al. The critical ment therapy by transplant in a system of bundled payment of dialy-
pathway for deceased donation: reportable uniformity in the approach sis. Clin Transplant 2012;26(4):529–31.
to deceased donation. Transplant Int 2011;24(4):373–8. 41. Popat R, Syed A, Puliatti C, Cacciola R. Outcome and cost analysis of
13. Muller E, White S, Delmonico F. Regional perspective: develop- induction immunosuppression with IL2Mab or ATG in DCD kidney
ing organ transplantation in Sub-Saharan Africa. Transplantation transplants. Transplantation 2014;97(11):1161–5.
2014;97(10):975–6. 42. Sudhindran S, Aboobacker S, Menon RN, Unnikrishnan G, Sudheer
14. Thresia CU. Rising private sector and falling ‘good health at low cost’: OV, Dhar P. Cost and efficacy of immunosuppression using generic
health challenges in China, Sri Lanka, and Indian state of Kerala. Int J products following living donor liver transplantation in India. Indian
Health Serv 2013;43(1):31–48. J Gastroenterol 2012;31(1):20–3.
15. Hinsenkamp M, Muylle L, Eastlund T, Fehily D, Noel L, Strong DM. 43. Ixtlapale-Carmona X, Arvizu A, De-Santiago A, et al. Graft immu-
Adverse reactions and events related to musculoskeletal allografts: nologic events in deceased donor kidney transplant recipients
reviewed by the World Health Organisation Project NOTIFY. Int with preformed HLA-donor specific antibodies. Transplant Immu-
Orthop 2012;36(3):633–41. nol 2018;46:8–13. Available online at: https://doi.org/10.1016/
16. Ashuntantang G, Osafo C, Olowu WA, et al. Outcomes in adults and j.trim.2017.09.006.
children with end-stage kidney disease requiring dialysis in sub-Saharan 44. Susal C, Fichtner A, Tonshoff B, Mehrabi A, Zeier M, Morath C. Clini-
Africa: a systematic review. Lancet Glob Health 2017;5(4):e408–17. cal relevance of HLA antibodies in kidney transplantation: recent data
17. Naicker S. Nephrology in Africa: challenges of practice in resource- from the Heidelberg Transplant Center and the Collaborative Trans-
limited environment. Clin Nephrol 2016;86:84–9. plant Study. J Immunol Res 2017:5619402.
38 • Kidney Transplantation in Developing Countries 683

45. Weber LT. Preexisting anti-HLA donor-specific antibodies in pedi- 55. Lee B, Michaels MG. Prophylactic antimicrobials in solid organ trans-
atric renal transplant recipients: a new challenge? Pediatr Trans- plant. Curr Op Crit Care 2014;20(4):420–5.
plant 2017;21(8). Available online at: https://doi.org/10.1111/ 56. Penne EL, Nurmohamed SA. Underdosing of prophylactic valganci-
petr.13085. clovir due to inaccurate estimation of glomerular filtration rate lead-
46. Nasim A, Anis S, Baqi S, Akhtar SF, Baig-Ansari N. Clinical presen- ing to severe cytomegalovirus disease in a kidney transplant recipient.
tation and outcome of dengue viral infection in live-related renal Antimicrob Agent Chemo 2014;58(2):1271–2.
transplant recipients in Karachi, Pakistan. Transplant Infect Dis 57. Clemente WT, Pierrotti LC, Abdala E, et al. Recommendations for
2013;15(5):516–25. management of endemic diseases and travel medicine in solid-organ
47. Ashoor IF, Dharnidharka VR. Sexually transmitted infection screen- transplant recipients and donors: Latin America. Transplantation
ing and reproductive health counseling in adolescent renal trans- 2018;102(2):193–208.
plant recipients: perceptions and practice patterns. A study from 58. John S, Andersson KL, Kotton CN, et al. Prophylaxis of hepatitis B
the Midwest Pediatric Nephrology Consortium. Pediatr Transplant infection in solid organ transplant recipients. Therap Adv Gastroen-
2015;19(7):704–8. terol 2013;6(4):309–19.
48. Cho J, Seville MT, Khanna S, Pardi DS, Sampathkumar P, Kashyap 59. Morris MI, Daly JS, Blumberg E, et al. Diagnosis and management of
PC. Screening for Clostridium difficile colonization on admission to a tuberculosis in transplant donors: a donor-derived infections consen-
hematopoietic stem cell transplant unit may reduce hospital-acquired sus conference report. Am J Transplant 2012;12(9):2288–300.
C difficile infection. Am J Infect Control 2018;46(4):459–61. Avail- 60. Manyalich M, Mestres CA, Balleste C, Paez G, Valero R, Gomez MP.
able online at: https://doi.org/10.1016/j.ajic.2017.10.009. Organ procurement: Spanish transplant procurement management.
49. Enache EM, Iancu LS, Hogas S, et al. Screening for latent BK virus Asian Cardiovasc Thorac Ann 2011;19(3-4):268–78.
infection in a renal transplant population for the first time in Roma- 61. Summers DM, Watson CJ, Pettigrew GJ, et al. Kidney donation after
nia: a single-center experience. Int Urol Nephrol 2012;44(2):619–23. circulatory death (DCD): state of the art. Kidney Int 2015;88(2):
50. Grover IS, Davila R, Subramony C, Daram SR. Strongyloides infec- 241–9.
tion in a cardiac transplant recipient: making a case for pretrans- 62. Cavallo MC, Sepe V, Conte F, et al. Cost-effectiveness of kidney
plantation screening and treatment. Gastroenterol Hepatol (NY) transplantation from DCD in Italy. Transplant Proc 2014;46(10):
2011;7(11):763–6. 3289–96.
51. Soriano G, Perales MA. Adenovirus viremia and infection after 63. Sharif A. Behavioral science strategies to boost organ donor consent.
reduced-intensity allogeneic hematopoietic stem cell transplant: Transplantation 2015;99(11):e172–3.
should we institute a routine screening programme? Clin Infect Dis 64. Chamberlain K, Baker MR, Kandaswamy P, et al. Donor identification
2012;55(10):1371–2. and consent for deceased organ donation: summary of NICE guidance.
52. Wu SW, Chang HR, Hsieh MC, Chiou HL, Lin CC, Lian JD. Early BMJ 2012;344:e341.
diagnosis of polyomavirus type BK infection in tailoring immuno- 65. Bendorf A, Kelly PJ, Kerridge IH, et al. An international comparison of
suppression for kidney transplant patients: screening with urine qual- the effect of policy shifts to organ donation following cardiocirculatory
itative polymerase chain reaction assay. Transplant Proc 2008;40(7): death (DCD) on donation rates after brain death (DBD) and transplan-
2389–91. tation rates. PLoS One 2013;8(5):e62010.
53. Bumbacea D, Arend SM, Eyuboglu F, et al. The risk of tuberculosis in 66. Boesken WH, Ahmed KE, Mery JP, Segaert MF, Bourgoignie JJ. Obser-
transplant candidates and recipients: a TBNET consensus statement. vations on renal replacement services in Russia, Belarus and Lithu-
Europe Resp J 2012;40(4):990–1013. ania. Nephrol Dial Transplant 1995;10(11):2013–6.
54. Kim YJ, Kim SI. Vaccination strategies in patients with solid organ 67. Gaya SB, Upadhyaya R, Kahn D. The evolution of renal transplanta-
transplant: evidences and future perspectives. Clin Exp Vaccine Res tion in Mauritius (1980–1997). Transpl Int 2001;14(2):115–6.
2016;5(2):125–31.
39 Results of Renal
Transplantation
RACHEL E. PATZER, REEM E. HAMODA, and STUART J. KNECHTLE

CHAPTER OUTLINE Introduction Frailty and Patient Fitness


Renal Failure Treatments—Dialysis Versus Graft Survival
Transplantation Graft Survival for Expanded Criteria Donor
Kidney Donation Kidneys
Donation After Cardiac Death Monozygotic Twins
Recipient Pool Family Donors
Factors Influencing Outcomes of Paired Kidney Donation and Living
Transplantation Unrelated Donor Outcomes
Donor Age Cancer Risk
Recipient Age Pregnancy After Renal Transplantation
Obesity Renal Transplantation in Human
Race/Ethnicity Immunodeficiency Virus-Positive Patients
APOL1 Prevalence of People Living With a
HLA Mismatch and Prior Sensitization Functioning Kidney Transplant
Cold Ischemia Time Re-Hospitalization
Expanded Criteria Donor Kidney Recipients Long-Term Outcomes of Renal
and Kidney Donor Profile Index Transplantation
Living Donor Kidney Recipients Quality of Life
Immunosuppression Conclusion
Adherence (Compliance) With
Immunosuppressive Treatment

Introduction data, individual center reports and multicenter trial data,


data from Europe through the Collaborative Transplant
The use of outcome data for renal transplantation in the Study (CTS), and the Australia and New Zealand Dialysis
United States (US) represents one of the best available and Transplant (ANZDATA) Registry.3–5 These data inform
examples of medical care supported by local and national decisions regarding patient access and outcomes and organ
databases to allow evidence-based decisions in the field. allocation. The data mentioned here refer to transplanta-
According to requirements directed by the United Network tion in the Western world; results from less well-developed
for Organ Sharing (UNOS), a federal government-autho- countries are discussed in Chapter 38.
rized body, all transplant centers must submit transplant
data to the Scientific Registry of Transplant Recipients
(SRTR), where the data are collated and analyzed on both
Renal Failure Treatments—
a center-specific basis and a cumulative national basis. Dialysis Versus Transplantation
Much of the data from the US summarized in this chapter
is derived from the 2015 SRTR report on kidney and pan- Renal failure is known to increase mortality from cardio-
creas transplant outcomes,1,2 available in the American vascular disease and from causes directly resulting from
Journal of Transplantation (http://onlinelibrary.wiley.com/ renal failure itself, including fluid and electrolyte imbal-
doi/10.1111/ajt.2017.17.issue-S1/issuetoc). The massive ance and uremia.6 Although dialysis addresses the immedi-
amount of data in the 2015 SRTR report has been reduced ately life-threatening complications of renal failure, it does
to that which is included in this chapter for the purposes of not provide fluid and electrolyte homeostasis comparable
greater usefulness and readability. The source of the data is to that of a well-functioning kidney. Several additional
acknowledged in figures and tables. In addition, other data metabolic functions of the kidney, such as vitamin D syn-
have been added to supplement the SRTR report, includ- thesis and erythropoietin synthesis, are also not regulated
ing United States Renal Data System (USRDS) surveillance appropriately in the absence of a well-functioning kidney.
684
39 • Results of Renal Transplantation 685

This reality is reflected by the well-documented finding that receipt.27–29 However, the new US kidney allocation sys-
patients with end-stage renal disease (ESRD) have improved tem implemented in December of 2014 suggests that racial
survival with transplantation compared with dialysis ther- and ethnic disparities in kidney transplantation among
apy.7–13 Patients who receive dialysis have an expected waitlisted patients may have at least temporarily abated.30
remaining lifetime of 6.8 years, compared with 17.8 years Patients with ESRD benefit from transplantation as early as
for transplant recipients (Table 39.1).3 In addition, kid- possible to maximize their potential for long survival after
ney transplantation is cost effective compared with dialy- transplantation.
sis and offers improved quality of life.11,14–16 Studies have
shown an increasing cardiovascular risk proportional to
the increase in serum creatinine, suggesting that renal fail- Kidney Donation
ure correlates with, if not causes, accelerated vascular and
metabolic defects that predispose patients to cardiovascular In the US the total number of deceased and live kidney
death. Dialysis patients are known to experience accelerated transplants decreased 4.4% from 2006 to 2012, but
atherosclerosis,17–19 and there are several inflammatory increased 6.8% from 2012 to 2015. Kidney donation
and atherogenic factors may account for this.20–24 Given rates from deceased donors decreased slightly since 2014,
these facts, it is not surprising that analysis of USRDS data at 72.2 eligible donors per 100 eligible deaths in 2015.
revealed that longer time on the wait list for renal transplan- The number of kidneys transplanted from living donors
tation correlates with poorer death-censored graft survival decreased 15.7% from 2009 to 2015, resulting in a total of
after renal transplantation (Fig. 39.1). Although there is a 5386 donors in 2015.1 In Europe, there is wide variation
clear advantage in receiving a kidney transplant before ever in deceased donor rates between countries. Deceased dona-
starting dialysis,25 this practice, known as preemptive renal tion has generally remained stagnant or even decreased
transplantation, is not widely adopted. In 2015, 15.9% of over recent years, with the exception of Spain, Austria,
adult US kidney transplant recipients underwent preemp- and Belgium, where donor rates are the highest in the
tive transplantation, whereas the remaining patients were world after the introduction of presumed consent (opt-out)
on dialysis at the time of transplant.1 policies.31,32
The better outcomes of patients with preemptive trans-
plants and with shorter time on dialysis underscore the DONATION AFTER CARDIAC DEATH
importance of early referral and evaluation for renal trans-
plantation. However, patients with kidney disease face a Donation after cardiac death (DCD) can yield either
number of barriers in accessing early nephrology care, expanded criteria donors (ECD) or standard criteria donors
including organ allocation policies and delayed referral (SCD). DCD has increased substantially since 2000: DCD/
to nephrology care or transplant centers.26,27 In the US, SCD represented 10.8%, and DCD/ECD represented 1.1% of
these barriers are particularly apparent for African Ameri- all organ donors in the US in 2009.9 DCD kidneys are kid-
cans versus whites: African Americans have a lower rate neys procured after cessation of cardiac activity (in Europe,
of accessing each step of the kidney transplant process, often referred to as nonheart-beating donors); this also is
including referral, evaluation, waitlisting, and transplant discussed in Chapters 6 and 9. Growth in DCD donors for

TABLE 39.1 Expected Remaining Lifetime (Years) by Age, Sex, and Treatment Modality of Period Prevalent Dialysis Patients,
Prevalent Transplant Patients, and the General US Population (2015), based on United States Renal Data System Data and the
National Vital Statistics Report (2014)
ESRD PATIENTS, 2015 GENERAL US POPULATION, 2014
Dialysis Transplant
Age Male Female Male Female Male Female
0–14 23.8 23.1 59.3 60.3 70.7 75.4
15–19 21.8 19.1 47.6 48.7 59.7 64.4
20–24 18.8 16.1 43.4 44.5 55.0 59.5
25–29 16.2 14.1 39.4 40.7 50.3 54.6
30–34 14.1 12.6 35.1 36.6 45.7 49.7
35–39 12.6 11.5 31.1 33.0 41.0 45.0
40–44 11.0 10.3 27.2 28.9 36.5 40.3
45–49 9.3 8.8 23.3 25.2 32.0 35.6
50–54 7.9 7.7 19.9 21.8 27.7 31.1
55–59 6.6 6.6 16.7 18.4 23.7 26.8
60–64 5.5 5.7 13.9 15.4 19.9 22.6
65–69 4.6 4.8 11.4 12.7 16.2 18.6
70–74 3.8 4.0 9.4 10.3 12.8 14.8
75–79 3.2 3.5 7.6a 8.6a 9.8 11.4
80–84 2.6 2.9 7.1 8.4
85+ 2.1 2.3 3.8 4.4
aCellvalues combine ages 75+.
ESRD, end-stage renal disease.
Data Source: Reference Tables H.13; special analyses, USRDS ESRDS Database; and Table 7 in National Vital Statistics Report, Deaths: Final Data for 2014.
686 Kidney Transplantation - Principles and Practice

kidney transplantation represents the largest increase in a in the modern era by Kootstra’s team at Maastricht38,39 and
type of donor kidney available for recipients in the US. The continued at the University of Wisconsin as the shortage of
ethics and methods of DCD recovery have been discussed at kidneys grew.34
length by D’Alessandro and colleagues,33 and single-center
experiences have resulted in outcomes not significantly dif- RECIPIENT POOL
ferent from SCD kidney transplantation.34 Several larger
retrospective studies have compared short- and long-term At the end of 2015, there were 97,680 patients await-
outcomes of DCD versus SCD and ECD donors and found ing renal transplantation in the US. New registrations for
equivalent patient and graft survival at 5 years.35,36 In a kidney transplantation in 2015 numbered 30,232 (Table
large UNOS registry study that examined more than 75,000 39.2). The number of kidney transplants in the US has gen-
transplant recipients, results suggest that DCD kidneys from erally remained stable since 2005, although there was a
donors younger than 50 years old have equivalent graft 1% increase in kidney transplants from 2013 to 2014. The
survival to SCD kidneys.37 The use of DCD donors, normal largest demographic increase in this population was in the
practice in the early days of transplantation, was pioneered 50- to 64-year-old age range.1 Since 2003, the age group

0.1 0.5 1 2 10

Study ID Adj HR (95% CI)


< 30 days
Transplant vs. Dialysis
Oniscu 2005* 0.91 (0.22, 3.70)
Oniscu 2004* 5.03 (1.43, 17.73)
McDonald 2002* ↑ 2.00 (1.50, 2.70)
Rabbat 2000* 2.91 (1.34, 6.32)
Ojo 1994* 3.30 P < 0.03
Port 1993* 2.43 P ≤ 0.01
> 1 year
Transplant vs. Dialysis
Oniscu 2005* 0.28 (0.20, 0.39)
Oniscu 2004* 0.27 (0.14, 0.52)
McDonald 2002* 0.19 (0.15, 0.24)
Rabbat 2000* 0.25 (0.14, 0.42)
Ojo 1994* 0.49 P < 0.03
Port 1993* 0.36 P ≤ 0.001
Full follow-up
Transplant vs. Dialysis
Bayat 2010 < 60 y 0.23 (0.13, 0.42)
Bayat 2010 ≥ 60 y 0.22 (0.10, 0.45)
Jain 2009 0.20 (0.11, 0.34)
Sorensen 2007 nDM 0.40 (0.30, 0.55)
Sorensen 2007 DM 0.21 (0.13, 0.34)
Snyder 2006 nDM'nPAD* 0.73 (0.68, 0.79)
Snyder 2006 DM'nPAD* 0.57 (0.52, 0.62)
Snyder 2006 nDM'PAD* 0.47 (0.40, 0.56)
Snyder 2006 DM'PAD* 0.36 (0.31, 0.41)
Merion 2005 ECD* 0.40 (0.37, 0.44)
Merion 2005* 0.28 (0.27, 0.30)
Abbott 2004 HCV+donor 0.76 (0.60, 0.96)
Abbott 2004 HCV-donor 0.47 (0.44, 0.50)
Brunkhorst 2003* 0.29 (0.12, 0.70)
Glanton 2003 nObese* 0.39 (0.35, 0.43)
Glanton 2003 Obese* 0.39 (0.33, 0.47)

0.1 0.5 1 2 10
*Indicate waitlisted dialysis groups Favors transplant Favors dialysis
Fig. 39.1 (A) Adjusted hazard ratios of all-cause mortality by time period for dialysis versus kidney transplantation across 38 cohorts. (B) Adjusted
hazard ratios of all-cause mortality by time period for dialysis versus kidney transplantation across 38 cohorts. (A, Reprinted from Tonelli M, Wiebe N,
Knoll G, Bello A, Browne S, Jadhav D, Klarenbach S, Gill J. Systematic review: kidney transplantation compared with dialysis in clinically relevant outcomes. Am
J Transplant 2011;11(10):2093–109. B, Reprinted from Tonelli M, Wiebe N, Knoll G, Bello A, Browne S, Jadhav D, Klarenbach S, Gill J. Systematic review: kidney
transplantation compared with dialysis in clinically relevant outcomes. Am J Transplant 2011;11(10):2093–109.)
39 • Results of Renal Transplantation 687

0.1 0.5 1 2 10

Study ID Adj HR (95% CI)


Full follow-up
Tranplant vs. Dialysis
Johnson 2000* 0.16 (0.06, 0.42)
Mazzuchi 1999 nDM 0.97 (0.61, 1.52)
Mazzuchi 1999 DM 0.35 (0.15, 0.83)
Wolfe 1999* 0.32 (0.30, 0.35)
Schnuelle 1998* 0.37 (0.14, 0.97)
Bonal 1997* 0.51 P = 0.02
Schaubel 1995 0.47 (0.33, 0.67)

You might also like