+ All Categories
Home > Documents > Transplant Infections -3E

Transplant Infections -3E

Date post: 21-Feb-2018
Category:
Upload: carlos-prado
View: 220 times
Download: 0 times
Share this document with a friend
800
Transplant Infections Transplant Infections EDITORS RALEIGH A. BOWDEN, MD Clinical Associate Professor of Pediatrics University of Washington School of Medicine Seattle, Washington PER LJUNGMAN, MD, PhD Professor of Hematology Karolinska University Hospital and Karolinska Institutet Stockholm, Sweden DAVID R. SNYDMAN, MD, FACP Professor of Medicine Tufts University School of Medicine Chief, Division of Geographic Medicine and Infectious Diseases Tufts Medical Center Boston, Massachusetts THIRD EDITION
Transcript
  • 7/24/2019 Transplant Infections -3E

    1/798

    TransplantInfectionsTransplantInfections

    E D I T O R S

    RALEIGH A. BOWDEN, MDClinical Associate Professor of Pediatrics

    University of Washington School of Medicine

    Seattle, Washington

    PER LJUNGMAN, MD, PhDProfessor of Hematology

    Karolinska University Hospital and

    Karolinska Institutet

    Stockholm, Sweden

    DAVID R. SNYDMAN, MD, FACPProfessor of Medicine

    Tufts University School of Medicine

    Chief, Division of Geographic Medicine and

    Infectious Diseases

    Tufts Medical Center

    Boston, Massachusetts

    T H I R D E D I T I O N

  • 7/24/2019 Transplant Infections -3E

    2/798

    Acquisitions Editor: Julia Seto

    Product Manager: Leanne McMillan

    Development Editor: Jenny Koleth

    Production Manager: Bridgett Dougherty

    Senior Manufacturing Manager: Benjamin Rivera

    Marketing Manager: Kimberly Schonberger

    Design Coordinator: Stephen Druding

    Production Service: MPS Limited, A Macmillan Company

    Third Edition

    Copyright 2010 by LIPPINCOTT WILLIAMS & WILKINS, a WOLTERS KLUWER business

    Two Commerce Square

    2001 Market Street

    Philadelphia, PA 19103 USA

    LWW.com

    Printed in China

    All rights reserved. This book is protected by copyright. No part of this book may be reproduced in any form by any

    means, including photocopying, or utilized by any information storage and retrieval system without written permis-

    sion from the copyright owner, except for brief quotations embodied in critical articles and reviews. Materials appear-ing in this book prepared by individuals as part of their official duties as U.S. government employees are not covered

    by the above-mentioned copyright.

    9 8 7 6 5 4 3 2 1

    Library of Congress Cataloging-in-Publication Data

    Transplant infections / editors, Raleigh A. Bowden, Per Ljungman, David R. Snydman.3rd ed.

    p. ; cm.

    Includes bibliographical references and index.

    ISBN 978-1-58255-820-2 (alk. paper)

    1. Communicable diseases. 2. Transplantation of organs, tissues, etc.Complications. 3. Nosocomial infections.

    I. Bowden, Raleigh A. II. Ljungman, Per. III. Snydman, David R.

    [DNLM: 1. Transplantsadverse effects. 2. Bacterial Infectionsetiology. 3. Mycosesetiology. 4. Virus Diseases

    etiology. WO 660 T691 2010]RC112.T73 2010

    617.9'5dc22

    2010001262

    DISCLAIMER

    Care has been taken to confirm the accuracy of the information presented and to describe generally ac-

    cepted practices. However, the authors, editors, and publisher are not responsible for errors or omissions or for any

    consequences from application of the information in this book and make no warranty, expressed or implied, with re-

    spect to the currency, completeness, or accuracy of the contents of the publication. Application of the information in a

    particular situation remains the professional responsibility of the practitioner.

    The authors, editors, and publisher have exerted every effort to ensure that drug selection and dosage set

    forth in this text are in accordance with current recommendations and practice at the time of publication. However, in

    view of ongoing research, changes in government regulations, and the constant flow of information relating to drug

    therapy and drug reactions, the reader is urged to check the package insert for each drug for any change in indicationsand dosage and for added warnings and precautions. This is particularly important when the recommended agent is a

    new or infrequently employed drug.

    Some drugs and medical devices presented in the publication have Food and Drug Administration

    (FDA) clearance for limited use in restricted research settings. It is the responsibility of the health care provider to as-

    certain the FDA status of each drug or device planned for use in their clinical practice.

    To purchase additional copies of this book, call our customer service department at (800) 638-3030 or fax orders to

    (301) 223-2320. International customers should call (301) 223-2300.

    Visit Lippincott Williams & Wilkins on the Internet: at LWW.com. Lippincott Williams & Wilkins customer service

    representatives are available from 8:30 am to 6 pm, EST.

    10 9 8 7 6 5 4 3 2 1

  • 7/24/2019 Transplant Infections -3E

    3/798

    Transplantation infectious disease has emerged as an important

    clinical subspecialty in response to a growing need for clinical ex-

    pertise in the management of patients with various forms of im-

    mune compromise. The field is evolving rapidly. In the past,

    with fairly standardized immunosuppressive regimens, clinical

    expertise in the care of immunocompromised patients required

    an understanding of the common pathogens causing infection at

    various times after transplantation and an understanding of the

    common toxicities and interactions of immunosuppressive med-

    ications and antimicrobial agents. Some of these concepts have

    now reached the level of transplant gospel. Thus, the equation

    of infectious risk after transplantation is determined by the rela-

    tionship between two factors: the individuals epidemiologic ex-

    posures and a conceptual measure of all those factors contribut-ing to an individuals infectious riskthe net state of

    immunosuppression. In the absence of assays that measure an

    individuals absolute risk for infection, allograft rejection, or

    graft-vs.-host disease, any determination of the net state of im-

    munosuppression is imprecise and is largely based on the clini-

    cians bedside skills and experience. In practice, the lack of such

    assays predicts that most patients will suffer excessive or inade-

    quate immunosuppression at some points during their posttrans-

    plant course provoking infection and/or rejection or GvHD.

    As with most good rules in medicine, exceptions to the

    rules have become common. Presentations of infection have

    been altered as transplantation has been applied to a broader

    range of clinical conditions, immunosuppressive regimenshave become more diverse, and prophylactic antimicrobial

    regimens have been deployed. How do we proceed? Some

    components of the risk equation have changed little. While

    different factors control the risk for infection in the earliest

    (weeks) periods following either transplant surgery (technical

    issues) or hematopoietic transplantation (neutropenia), the full

    impact of immunosuppression on adaptive immunity has not

    yet been achieved. Thus, in both groups, colonization by noso-

    comial flora and mechanical or technical challenges dominate

    risk including postoperative fluid collections, vascular

    catheters and surgical drains, tissue ischemia, drug side effects,

    underlying immune deficits (e.g., diabetes), organ dysfunc-tion, metabolic derangements, and antimicrobial exposures.

    Following the earliest posttransplant time period, investiga-

    tions into the pathogenesis of infection are beginning to unravel

    some of the underpinnings of host susceptibility via advances in

    microbiology, molecular biology, and immunology. While the

    equation of risk for infection balancing epidemiology and the

    net state of immune suppression remain valuable, at the basic

    science level, susceptibility to infection is now recognized to be

    a function of both the virulence of the organism and of host de-

    fenses, including both innate and adaptive immunity. The deter-

    minants of virulence of a particular organism are the genetic,

    biochemical, and structural characteristics that contribute to the

    production of disease. Susceptibility can be explained with refer-

    ence to the presence or absence of specific receptors for

    pathogens, the cells and proteins determining protective immu-

    nity, and the coordination of the hosts response to infection. The

    relationship between the host and the pathogen is dynamic.

    Thus, some of the alterations in susceptibility previously ascribed

    to indirect effects of the pathogen (e.g., for cytomegalovirus)

    can now be explained as virally mediated effects on processes in-

    cluding antigen presentation, cellular maturation and mobiliza-

    tion, and cytokine profiles. Much of the impact of these infec-

    tions appears to be at the interface of the innate immune system

    (monocytes, macrophages, dendritic cells, and NK cells) and the

    adaptive immune system (lymphocytes and antibodies). Other

    effects are the result of alterations in cell-surface (e.g., toll-like)receptors and on the milieu of other inflammatory mediators

    both locally and systemically. In an admittedly anthropomorphic

    description of these effects, the virus (and other pathogens) has

    altered the host to avoid detection and destruction and to pro-

    mote successful parasitism and persistence. As host and pathogen

    respond during the course of infection (and are modified by

    antimicrobial therapy or immunosuppression), each modifies the

    activities and functions of the other and a dynamic relationship

    develops. The outcome of such a relationship depends on the vir-

    ulence of the pathogen and the relative degree of resistance or

    susceptibility of the host, due largely to host defense mechanisms

    and to a more trivial degree, by antimicrobial therapies.

    Investigations into immune mechanisms are beginning toprovide assays that measure an individuals pathogen-specific

    immune function (T-cell subsets, HLA-restricted lymphocyte

    sorting using tetramers, antigen-specific interferon-release as-

    says) as a suggestion of pathogen-specific infectious risk. This

    approach may be of particular relevance in the future in regard

    to development of vaccines for use in immunocompromised

    hosts and in the assessment of immune reconstitution following

    chemotherapy and hematopoietic stem cell transplantation.

    The equation of risk has been further altered by a num-

    ber of additional factors. Outbreaks of epidemic infections (West

    Nile virus, H1N1 swine influenza, SARS) have disproportion-

    ately affected transplant recipients. The epidemiology of infec-tion has also been changed by the expanded population of pa-

    tients undergoing immunosuppression for transplantation,

    notably in terms of parasitic, mycobacterial, and other endemic

    infections. Thus, Chagas disease and leishmaniasis are routinely

    considered in the differential diagnosis of infection in the appro-

    priate setting. Donor-derived infections have been recognized in

    both hematopoietic and solid organ transplant recipients. Until

    recently, careful medical histories coupled with serologic and

    culture-based screening of organ donors and recipients, and rou-

    tine antimicrobial prophylaxis for surgery have successfully pre-

    vented the transmission of most infections with grafts. With the

    emergence of antimicrobial-resistant organisms in hospitals and

    ForewordForeword

  • 7/24/2019 Transplant Infections -3E

    4/798

    in the community, routine surgical prophylaxis for transplanta-

    tion surgery may fail to prevent transmission of common organ-

    isms including methicillin-resistant Staphylococcus aureus, van-

    comycin-resistantEnterococcus species, and azole-resistant yeasts.

    Highly sensitive molecular diagnostic assays have also allowed

    the identification of a series of uncommon viral infections (lym-

    phocytic choriomeningitis virus [LCMV], West Nile virus, rabies

    virus, HIV) with allografts. These infections appear to be ampli-fied in the setting of immunosuppression. Despite technological

    advances, deficiencies in the available screening assays are notable

    in that both false-positive assays (causing discarding of potentially

    usable organs) and false-negative assays (the inability to identify

    LCMV in a deceased donors transmitting LCMV to multiple re-

    cipients) have been recognized. Sensitive and specific diagnostic

    assays remain unavailable for some pathogens of interest and

    those that are available require careful validation and standardi-

    zation. Improved molecular assays and antigen detection-based

    diagnostics may help to prevent graft-derived transmissions in

    the future.

    Routine use of antimicrobial prophylaxis has also alteredthe presentation of infection following transplantation. In

    part, this manifests as a shift-to-the-right (late infections)

    due to common pathogens such as cytomegalovirus (CMV) in

    solid organ recipients. Increasingly, this is reflected in the

    emergence of antimicrobial-resistant pathogens. The impact

    of routine prophylaxis is difficult to measureit is uncertain

    that there is a clear mortality benefit of these strategies. Sicker

    patients arrive at transplantation having survived multiple in-

    fections, organ failure, or malignancies that would have been

    fatal in the past. These individuals may become Petri dishes

    for organisms for which effective therapies are lacking. The

    need for new antimicrobial agents is increasing at a time when

    the pipeline for new agents appears to be contracting.The net state of immunosuppression has also shifted. The

    duration of neutropenia following HSCT and with nonmye-

    loablative transplantation is shorter than that after traditional

    bone marrow transplantation. The duration of neutropenia

    has also been reduced with the introduction of chemothera-

    peutic agents targeting specific cellular sites (enzymes, protea-

    somes) rather than acting on rapidly dividing cancer cells.

    Among solid organ transplants, the recent introduction of ex-

    perimental protocols that use combinations of HSCT with

    renal transplantation to induce immunologic tolerance carries

    the promise of immunosuppression-free lifetimes for patients.

    A series of innovations will impact future clinical practice.The adoption of quantitative molecular and protein-based mi-

    crobiologic assays in routine clinical practice has enhanced diag-

    nosis and serves as a basis for the deployment of antiviral agents

    and modulation of exogenous immune suppression. In many

    ways, given currently available science, these assays may be the

    best measure of an individuals immune function relative to their

    own pathogens. Potent biologic agents in transplantation in-

    cluding antibody-based therapies to deplete lymphocytes (and

    other cells) have the capacity to reduce both graft rejection and

    graft-vs.-host disease in place of commonly used agents includ-

    ing corticosteroids and the calcineurin inhibitors. The short-

    term gain in terms of infectious risk and renal dysfunction from

    currently available agents must be balanced against longer term

    susceptibly to infections with organisms including mycobacteria,

    fungi, and viruses. Among the side effects of these therapies may

    be an increased risk for virally mediated malignancies (including

    PTLD) and BK (nephropathy) and JC polyomavirus-associated

    infections (i.e., progressive multifocal leukoencephalopathy,

    PML). The full impact of the biologic agents remains to be de-

    termined. High throughput sequencing and genome-wide asso-ciation studies are beginning to determine the basis of both ge-

    netic susceptibility to infection and responses to antimicrobial

    therapies (e.g., hepatitis C virus and interferon). These observa-

    tions will allow the application of specific drugs to the popula-

    tions in which they are most useful and least toxic (pharmacoge-

    nomics). The introduction of clinical xenotransplantation (i.e.,

    pig-to-human transplantation) may introduce a series of novel

    pathogens into the epidemiologic equation in the near future.

    The evolution of the immunosuppression used in organ

    and hematopoietic stem cell transplantation has reduced the in-

    cidence of acute graft rejection and graft-vs.-host disease while

    increasing the longer term risks for infection and virally medi-ated malignancies. With introduction of each new immunosup-

    pressive agent, a new series of effects on the presentation and

    epidemiology of infection have been recognized in the trans-

    plant recipient. In the absence of assays that measure infectious

    risk, transplant infectious disease remains as much a clinical art

    form as a science. In the future, improved assays for microbio-

    logic and immunologic monitoring will allow individualization

    of prophylactic strategies for transplant recipients and reduce

    the risk of infection in this highly susceptible population.

    As a reflection of the challenges posed by a rapidly changing

    field, the editors and contributors of this text have identified both

    the advances and the gaps in our knowledge in transplant infec-

    tious diseases. The unique risk factors and epidemiology for in-fection have been characterized for each of the major transplant

    populations. Important shifts in the epidemiology that have been

    identified include those due to donor-derived pathogens and the

    introduction of transplantation into geographically diverse pop-

    ulations. The clinical utility of the text is enhanced by discussions

    of common and important presentations of infection including

    infections of the lungs, skin, central nervous system, and gas-

    trointestinal tract. Individual pathogens and therapies are ad-

    dressed in detail. Vaccination for the immunocompromised host

    and innovative therapies entering clinical practice are clearly

    presented and assessed, including adoptive immunotherapy. In

    each case, clinically important management issues are identifiedincluding infection control, immunosuppressive adjustments,

    and prophylactic and therapeutic antimicrobials. The authors

    have, in addition, identified important controversies and trends

    for each topic so as to clue the reader into areas in which change

    is ongoing. In sum, this volume is an important addition to the

    currently available literature in transplantation for infectious dis-

    ease and transplantation specialists, for both expert and novice

    alike. The availability of this information in a single volume will

    serve one group particularly wellour patients.

    Jay A. Fishman, MD

    Boston, Massachusetts, USA

    iv Foreword

  • 7/24/2019 Transplant Infections -3E

    5/798

    The success of both the first edition of Transplant Infections,

    published in 1998, and the second edition, published in 2003,

    as a reference work to bring together information directed at

    the management of the infectious complications occurring

    specifically in immunocompromised individuals undergoing

    transplantation has led to the creation of this third edition. No

    other text focuses solely on exogenously immunosuppressed

    transplant patients, and no text combines solid organ and

    hematopoietic stem cell transplantation (historically referred

    to as bone marrow transplantation). Many texts focus on im-

    munocompromised patients, but the field of transplant infec-

    tious diseases has evolved over the past 20 years as a field unto

    itself, with conferences devoted solely to this specialty, and

    guidelines, both national and international, being developedfor the management of such patients. In addition, peer re-

    viewed journals now exist which publish information on this

    specialized area, and training programs devoted to the subspe-

    cialty of transplant infectious diseases within the field of infec-

    tious disease are being developed.

    The field of transplant infectious diseases has continued to

    grow and expand since the second edition was published in

    2003. We have expanded the third edition to include a greater

    emphasis on surgical complications for each type of organ trans-

    planted. In addition, there are new chapters on organ donor

    screening, drug interactions after transplantation, and new im-

    munosuppressive agents. Chapters differentiating differences

    between solid organ and hematopoietic stem cell transplanta-tion have been expanded, as have chapters discussing fungal in-

    fections, as more data accumulate for improved diagnosis and

    treatment and many new antifungal agents are developed.

    There is a new section in the cardiac transplant chapter on

    ventricular assist device infections, a problem the transplant in-

    fectious disease specialist must wrestle with often in patients

    awaiting cardiac transplantation. We have also expanded some

    chapters on viral infections, such as the polyomaviruses and

    adenovirus since recognition of the importance of these

    pathogens has grown. A chapter on rare viral infections has

    been updated as well. Transplant tourism as a topic has also

    been added to a section on transplant travel medicine and vac-

    cines. A number of new authors have been added and chapters

    have been substantially revised or completely rewritten.

    This edition remains a globally inclusive product of lead-

    ing authors and investigators from around the world.

    Perspectives from Argentina, Brazil, Chile, New Zealand,

    Western Europe (Italy, Spain, Sweden, Germany, France, and

    Switzerland), Austria, the United States, Canada, and Israel

    have been synthesized in this edition.We continue to believe that much can be learned regard-

    ing an appreciation of both the similarities and the differences

    in the pattern of infections and the resulting morbidity and

    mortality in various transplant settings. Our goal with this

    textbook is to provide background and knowledge for all

    practitioners who work with transplant patients, in order to

    improve both the care and outcomes of transplant recipients

    and to provide a framework for education of physicians, and

    transplant coordinators, and trainees in the field. As success in

    the field continues to grow we hope that this text would pro-

    vide some small incremental knowledge base that would ad-

    vance the field and make transplantation safer for all who

    need this lifesaving intervention. We thank all the contribu-tors for their effort, and trust the reader will find this a valu-

    able reference text as they care for transplant recipients.

    Raleigh A. Bowden

    Per Ljungman

    David R. Snydman

    v

    PrefacePreface

  • 7/24/2019 Transplant Infections -3E

    6/798

    vi

    ContributorsContributors

    Tamara Aghamolla

    Immunocompromised Host Section

    Pediatric Oncology Branch

    Clinical Research Center

    National Cancer Institute

    National Institutes of Health

    Bethesda, Maryland

    Nora Al-mana, M.B.B.S.

    Tufts Medical Center

    Boston, Massachusetts

    Diana Averbuch, M.D.

    Department of Pediatrics

    The Hebrew University HadassahMedical School

    Infectious Diseases Consultant

    Department of Pediatrics

    Hadassah University Hospital

    Jerusalem, Israel

    Robin K. Avery, M.D.

    Professor of Medicine

    Cleveland Clinic Lerner College of Medicine

    of Case Western Reserve University

    Section Head

    Transplant Infectious Disease

    The Cleveland Clinic

    Cleveland, Ohio

    Emily A. Blumberg, M.D.

    Professor

    Department of Medicine

    University of Pennsylvania School of

    Medicine

    Division of Infectious Diseases

    Department of Medicine

    Hospital of the University of

    Pennsylvania

    Philadelphia, Pennsylvania

    Michael J. Boeckh, M.D.

    Associate Professor

    Department of Medicine

    University of Washington

    Member

    Vaccine and Infectious Disease Institute

    Fred Hutchinson Cancer Research Center

    Seattle, Washington

    Helen W. Boucher, M.D., F.A.C.P.

    Assistant Professor of Medicine

    Tufts University School of Medicine

    Director

    Fellowship Program

    Division of Geographic Medicine and

    Infectious Diseases

    Tufts Medical Center

    Boston, Massachusetts

    Emilio Bouza, M.D., Ph.D.

    Professor

    Clinical Microbiology

    University Complutense of Madrid

    ChiefClinical Microbiology and Infectious Diseases

    Hospital General Universitario Gregorio

    Maraon (HGUGM)

    Madrid, Spain

    Almudena Burillo, M.D., Ph.D.

    Physician

    Clinical Microbiology and Infectious Diseases

    Hospital de Mostoles

    Madrid, Spain

    Sandra M. Cockfield, M.D.

    Professor

    Department of Medicine

    University of Alberta

    Medical Director

    Renal Transplant Program

    Walter C. Mackenzie Health Science Center

    Edmonton, Alberta, Canada

    Jeffrey T. Cooper, M.D.

    Assistant Professor of Surgery

    Tufts University School of Medicine

    Attending Surgeon

    Tufts Medical Center

    Boston, Massachusetts

    Catherine Cordonnier, M.D.

    Professor of Hematology

    Hematology Oncology

    Universit Paris 12

    Head

    Clinical Hematology Department

    Henri Mondor University Hospital

    Crteil, France

    Isabel Cunningham, M.D.

    Adjunct Associate Research Scientist

    Hematology Oncology

    Columbia University College of Physicians

    and Surgeons

    New York, New York

    Mazen S. Daoud, M.D.

    Director

    Dermatopathology Laboratory

    Associates in Dermatology

    Fort Myers, Florida

    H. Joachim Deeg, M.D.

    Professor of MedicineMedical Oncology

    University of Washington Medical

    Center

    Member

    Transplantation Biology

    Fred Hutchinson Cancer Research

    Center

    Seattle, Washington

    David DeNofrio, M.D.

    Associate Professor of Medicine

    Cardiology/Medicine

    Tufts University School of Medicine

    Medical Director

    Cardiac Transplant Program

    Cardiology/Medicine

    Tufts Medical Center

    Boston, Massachusetts

    J. Stephen Dummer, M.D.

    Professor

    Departments of Medicine and Surgery

    Vanderbilt University School of

    Medicine

    Chief

    Transplant Infectious Diseases

    Vanderbilt University HospitalNashville, Tennessee

    Hermann Einsele

    Professor

    Department of Medicine

    University Wrzburg

    Director

    Department of Internal Medicine II

    University Hospital Wrzburg

    Wrzburg, Germany

  • 7/24/2019 Transplant Infections -3E

    7/798

    Dan Engelhard, M.D.

    Associate Professor

    Department of Pediatrics

    The Hebrew University Hadassah Medical

    School

    Chief

    Department of Pediatrics

    Hadassah University HospitalJerusalem, Israel

    Janet A. Englund, M.D.

    Professor

    Department of Pediatrics

    University of Washington

    Professor

    Pediatric Infectious Diseases

    Seattle Childrens Hospital

    Seattle, Washington

    Staci A. Fischer, M.D.

    Associate Professor

    Department of MedicineThe Warren Alpert Medical School of

    Brown University

    Director

    Transplant Infectious Diseases

    Rhode Island Hospital

    Providence, Rhode Island

    Richard Freeman, M.D.

    Professor and Chair

    Department of Surgery

    Dartmouth Medical School

    Hanover, New Hampshire

    ChairDepartment of Surgery

    Dartmouth-Hitchcock Medical Center

    Lebanon, New Hampshire

    Ed Gane, M.D., F.R.A.C.P.

    Associate Professor

    Faculty of Medicine

    University of Auckland

    Consultant Hepatologist

    Liver Unit

    Auckland City Hospital

    Auckland, New Zealand

    Joan Gavald, M.D., Ph.D.Senior Consultant

    Servei Malalties Infeccioses

    Vall dHebron

    Barcelona, Spain

    Juan Gea-Banacloche, M.D.

    Infectious Diseases Section

    Experimental Transplantation and

    Immunology Branch

    National Cancer Institute, National

    Institutes of Health

    Chief

    Infectious Diseases Consultation ServiceNational Institutes of Health Clinical

    Research Center

    Bethesda, Maryland

    Maddalena Giannella, M.D.

    PhD Course

    Clinical Microbiology

    University Complutense of Madrid

    Research Fellow

    Clinical Microbiology and Infectious

    Diseases

    Hospital General Universitario Gregorio

    Maraon (HGUGM)

    Madrid, Spain

    Lawrence E. Gibson, M.D.

    Professor of Dermatology

    Director of Dermatopathology

    Mayo Clinic

    Rochester, Minnesota

    John W. Gnann, Jr., M.D.

    Professor of Medicine, Pediatrics, and

    Microbiology

    Department of Medicine, Division of

    Infectious Diseases

    University of Alabama at Birmingham and

    Birmingham Veterans Administration

    Medical Center

    Birmingham, Alabama

    Michael Green, M.D., M.D.H.

    Professor

    Pediatrics and Surgery

    University of Pittsburgh School of Medicine

    Attending Physician

    Division of Infectious Diseases

    Childrens Hospital of Pittsburgh

    Pittsburgh, Pennsylvania

    Andreas H. Groll, M.D.Associate Professor

    Department of Pediatrics

    Wilhelms University

    Head

    Infectious Disease Research Program

    Center for Bone Marrow Transplantation

    and Department of Pediatric

    Hematology/Oncology

    Childrens University Hospital

    Muenster, Germany

    Morgan Hakki, M.D.

    Assistant Professor

    Division of Infectious Diseases

    Oregon Health and Science University

    Portland, Oregon

    John W. Hiemenz, M.D.

    Professor of MedicineDivision of Hematology/Oncology

    University of Florida College of Medicine

    Attending Physician

    Bone Marrow Transplant/Leukemia

    Program

    Shands at the University of Florida

    Gainesville, Florida

    Hans H. Hirsch, M.D., M.S.

    Professor

    Institute for Medical Microbiology

    Department of Biomedicine

    University of Basel

    Senior PhysicianInfectious Diseases & Hospital

    Epidemiology

    Department of Internal Medicine

    University Hospital Basel

    Petersplatz, Basel, Switzerland

    Jack W. Hsu, M.D.

    Assistant Professor

    Department of Medicine

    University of Florida

    Clinical Assistant Professor

    Department of Medicine

    University of Florida Shands Cancer

    Center

    Gainesville, Florida

    Abhinav Humar, M.D.

    Professor

    Department of Surgery

    University of Pittsburgh

    Chief of Transplant

    Starzl Transplant Institute

    University of Pittsburgh Medical Center

    Pittsburgh, Pennsylvania

    Atul Humar, M.D., M.SC., F.R.C.P. (C)

    Associate Professor of MedicineTransplant Infectious Diseases

    University of Alberta

    Director

    Transplant Infectious Diseases

    University of Alberta Hospital

    Edmonton, Alberta, Canada

    Contributors vii

  • 7/24/2019 Transplant Infections -3E

    8/798

    Michael G. Ison, M.D., M.S.

    Assistant Professor

    Divisions of Infectious Diseases & Organ

    Transplantation

    Northwestern University Feinberg School

    of Medicine

    Medical Director

    Transplant & Immunocompromised HostInfectious Diseases Service

    Northwestern Memorial Hospital

    Chicago, Illinois

    Barry D. Kahan, M.D., Ph.D.

    Professor Emeritus

    The University of Texas Medical School at

    Houston

    Houston, Texas

    Carol A. Kauffman, M.D.

    Professor

    Department of Internal Medicine

    University of Michigan Medical SchoolChief

    Infectious Diseases Section

    Veterans Affairs Ann Arbor Healthcare

    System

    Ann Arbor, Michigan

    Camille Nelson Kotton, M.D.

    Assistant Professor

    Department of Medicine

    Harvard Medical School

    Clinical Director

    Transplant Infectious Disease and

    Compromised Host Program

    Infectious Diseases Division

    Massachusetts General Hospital

    Boston, Massachusetts

    Sharon Krystofiak, M.S., M.T.(A.S.C.P.), C.I.C.

    Infection Preventionist

    Infection Control and Hospital

    Epidemiology

    University of Pittsburgh Medical Center

    Presbyterian

    Pittsburgh, Pennsylvania

    Deepali Kumar, M.D., M.SC.,F.R.C.P. (C)

    Assistant Professor of Medicine

    Transplant Infectious Diseases

    University of Alberta

    Staff Physician

    Transplant Infectious Diseases

    University of Alberta Hospital

    Edmonton, Alberta, Canada

    Shimon Kusne, M.D.

    Professor of Medicine

    Department of Medicine

    Mayo Medical School

    Chair

    Division of Infectious Diseases

    Department of Medicine

    Mayo Clinic ArizonaPhoenix, Arizona

    Roberta Lattes, M.D.

    Assistant Professor Infectious Diseases

    Department of Medicine

    University of Buenos Aires School of

    Medicine

    Chief

    Departement of Transplantation

    Transplant Infectious Disease

    Instituto de Nefrologa

    Buenos Aires, Argentina

    Kenneth R. Lawrence, B.S.,PHARM.D.

    Assistant Professor

    Department of Medicine

    Tufts University School of Medicine

    Senior Clinical Pharmacy Specialist

    Pharmacy

    Tufts Medical Center

    Boston, Massachusetts

    Ingi Lee, M.D., M.S.C.E.

    Instructor

    Department of Medicine

    University of Pennsylvania School of

    Medicine

    Division of Infectious Diseases

    Department of Medicine

    Hospital of the University of Pennsylvania

    Philadelphia, Pennsylvania

    Ajit P. Limaye, M.D.

    Associate Professor

    Department of Medicine

    University of Washington

    Director

    Solid-Organ Transplant Infectious Disease

    University of Washington Medical Center

    Seattle, Washington

    Per Ljungman, M.D., Ph.D.

    Professor of Hematology

    Karolinska Institutet

    Director

    Department of Hematology

    Karolinska University Hospital Stockholm

    Stockholm, Sweden

    Anna Locasciulli, M.D.

    Associated Professor

    Pediatric Hematology

    University of Medicine

    Director

    Pediatric Hematology

    San Camillo Hospital

    Rome, Italy

    David L. Longworth, M.D.

    Professor of Medicine and Deputy

    Chairman

    Department of Medicine

    Tufts University School of Medicine

    Chairman

    Department of Medicine

    Baystate Medical Center

    Springfield, Massachusetts

    Mitchell R. Lunn, B.S.

    Department of Medicine

    Stanford University School of MedicineStanford, California

    Clarisse M. Machado, M.D.

    Virology Laboratory

    So Paulo Institute of Tropical Medicine

    University of So Paulo

    So Paulo, Brazil

    Kieren A. Marr, M.D.

    Professor of Medicine

    Department of Medicine

    Johns Hopkins University

    Director

    Transplant and Oncology Infectious

    Disease

    Department of Medicine

    Johns Hopkins University

    Baltimore, Maryland

    Rodrigo Martino, M.D., Ph.D.

    Attending Senior Physician

    Hematology

    Hospital de la Santa Creu i Sant Pau

    Barcelona, Catalonia, Spain

    Susanne Matthes-Martin, M.D.

    Associate ProfessorHead of Unit

    St. Anna Childrens Hospital

    Stem Cell Transplant Unit

    Childrens Cancer Research Institute

    Vienna, Austria

    viii Contributors

  • 7/24/2019 Transplant Infections -3E

    9/798

    Lisa M. McDevitt, PHARM.D.,B.C.P.S.

    Assistant Professor

    Department of Surgery

    Tufts University School of Medicine

    Senior Clinical Specialist

    Organ Transplantation

    Department of PharmacyTufts Medical Center

    Boston, Massachusetts

    George B. McDonald, M.D.

    Professor

    Department of Medicine

    University of Washington

    Head and Member

    Gastroenterology, Hospital Section

    Fred Hutchinson Cancer Research

    Center

    Seattle, Washington

    Marian G. Michaels, M.D., M.D.H.Professor

    Department of Pediatrics and Surgery

    University of Pittsburgh

    Division of Pediatric Infectious Diseases

    Department of Pediatrics

    Childrens Hospital of Pittsburgh of

    University of Pittsburgh Medical

    Center

    Pittsburgh, Pennsylvania

    Barbara Montante, M.D.

    Resident

    Pediatric Hematology and Bone Marrow

    Transplant Unit

    San Camillo Hospital

    Rome, Italy

    Jose G. Montoya, M.D.

    Associate Professor of Medicine

    Division of Infectious Diseases and

    Geographic Medicine

    Stanford University School of Medicine

    Attending Physician

    Department of Medicine

    Stanford Hospital and Clinics

    Stanford, California

    David C. Mulligan, M.D., F.A.C.S.

    Professor

    Department of Surgery

    Mayo Clinic School of Medicine

    Director

    Transplant Center

    Mayo Clinic Arizona

    Phoenix, Arizona

    Patricia Muoz, M.D., Ph.D.

    Professor

    Clinical Microbiology

    University Complutense of Madrid

    Clinical Section

    Chief

    Clinical Microbiology and Infectious Diseases

    Hospital General Universitario GregorioMaran (HGUGM)

    Madrid, Spain

    Tue Ngo, M.D., M.D.H.

    Infectious Diseases Fellow

    Division of Infectious Diseases

    Vanderbilt University School of Medicine

    Nashville, Tennessee

    Albert Pahissa, M.D., Ph.D.

    Chair Professor

    Infectious Diseases Medicine

    Universitat Autnoma de Barcelona

    ChiefServei Malalties Infeccioses

    Vall dHebron

    Bellaterra, Barcelona, Spain

    Peter G. Pappas, M.D., F.A.C.P.

    Professor of Medicine

    Medicine and Infectious Diseases

    University of Alabama at Birmingham

    Birmingham, Alabama

    Maria Beatrice Pinazzi, M.D.

    Full-time Assistant

    Pediatric Hematology and Bone Marrow

    Transplant UnitSan Camillo Hospital

    Rome, Italy

    Jutta K. Preiksaitis, M.D.

    Professor of Medicine

    Department of Medicine

    University of Alberta

    Edmonton, Alberta, Canada

    Marcelo Radisic, M.D.

    Attending Physician

    Transplant Infectious Diseases

    Instituto De Nefrologa

    Buenos Aires, Argentina

    Raymund R. Razonable, M.D.

    Associate Professor of Medicine

    Department of Medicine

    Mayo Clinic College of Medicine

    Consultant Staff

    Division of Infectious Diseases

    Mayo Clinic

    Rochester, Minnesota

    Jorge D. Reyes, M.D.

    Professor

    Department of Surgery

    University of Washington

    Chief

    Division of Transplant Surgery

    University of Washington Medical

    CenterSeattle, Washington

    Andrew R. Rezvani, M.D.

    Research Associate

    Transplantation Biology Program

    Fred Hutchinson Cancer Research Center

    Acting Instructor

    Medical Oncology

    University of Washington Medical

    Center

    Seattle, Washington

    Jason Rhee, M.D.

    Transplant Research Fellow

    Department of Surgery

    Tufts Medical Center

    Boston, Massachusetts

    Stanely R. Riddell, M.D.

    Professor of Medicine

    Fred Hutchinson Cancer Research Center

    Seattle, Washington

    Antonio Romn, M.D., Ph.D.

    Senior Consultant

    Pneumology Department

    Vall dHebronBarcelona, Spain

    Brahm H. Segal, M.D.

    Assistant Professor of Medicine

    State University of New York at Buffalo

    Head of Infectious Disease

    Roswell Park Cancer Institute

    Buffalo, New York

    Maria Teresa Seville, M.D.

    Instructor

    Division of Infectious Diseases

    Mayo Clinic

    ChairInfection Prevention and Control

    Mayo Clinic Hospital

    Phoenix, Arizona

    Nina Singh, M.D.

    Associate Professor of Medicine

    University of Pittsburgh

    Pittsburgh, Pennsylvania

    Contributors ix

  • 7/24/2019 Transplant Infections -3E

    10/798

    David R. Snydman, M.D., F.A.C.P.

    Professor of Medicine

    Tufts University School of Medicine

    Chief, Division of Geographic Medicine

    and Infectious Diseases

    Hospital Epidemiologist

    Tufts Medical Center

    Boston, Massachusetts

    Gideon Steinbach, M.D., Ph.D.

    Associate Professor

    Department of Medicine

    University of Washington

    Associate Member

    Gastroenterology, Hospital Section

    Fred Hutchinson Cancer Research Center

    Seattle, Washington

    William J. Steinbach, M.D.

    Associate Professor

    Departments of Pediatrics and Molecular

    Genetics & Microbiology

    Duke University

    Durham, North Carolina

    W. P. Daniel Su, M.D.

    Professor of Dermatology

    Mayo Clinic

    Rochester, Minnesota

    Max S. Topp, M.D.

    Director

    Internal Medicine II

    University Medical Center II

    Wrzburg, Germany

    Thomas J. Walsh, M.D., F.A.C.P.,F.I.D.S.A., F.A.A.M.

    Adjunct Professor of Pathology

    The Johns Hopkins University School of

    Medicine

    Adjunct Professor of Medicine

    University of Maryland School of Medicine

    Senior InvestigatorChief, Immunocompromised Host Section

    National Cancer Institute

    Baltimore, Maryland

    Daniel J. Weisdorf, M.D.

    Professor & Director

    Adult Blood and Marrow Transplant

    Program

    Department of Medicine

    University of Minnesota

    Minneapolis, Minnesota

    Estella Whimbey, M.D.

    Associate Professor of MedicineUniversity of Washington

    Associate Medical Director

    Employee Health Center

    University of Washington Medical Center

    Medical Director

    Healthcare Epidemiology and Infection

    Control

    University of Washington Medical

    Center/Seattle Cancer Care Alliance

    (inpatients)

    Seattle, Washington

    John R. Wingard, M.D.

    Professor

    Department of Medicine

    University of Florida

    Director of Bone Marrow Transplant

    Program

    Department of Medicine

    University of Florida Shands CancerCenter

    Gainesville, Florida

    Jo-Anne H. Young, M.D.

    Associate Professor

    Department of Medicine

    University of Minnesota

    Director of the Program in Transplant

    Infectious Disease

    Department of Medicine

    University of Minnesota Medical Center

    Minneapolis, Minnesota

    x Contributors

  • 7/24/2019 Transplant Infections -3E

    11/798

    Foreword iii

    Preface v

    Contributors vi

    Section I

    Introduction to Transplant Infections1 Introduction to Hematopoietic Cell

    Transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1Andrew R. Rezvani and H. Joachim Deeg

    2 Introduction to Solid OrganTransplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . 13Barry D. Kahan

    3 Immunosuppressive Agents . . . . . . . . . . . . . . . . . 26Jason Rhee, Nora Al-Mana, Jeffery T. Cooper and Richard Freeman

    4 Common Drug Interactions Encountered inTreating Transplant-Related Infections . . . . . . . . 41Helen W. Boucher, Kenneth R. Lawrence

    and Lisa M. McDevitt

    Section II

    Risks and Epidemiology of Infectionsafter Transplantation5 Risks and Epidemiology of Infections

    after Allogeneic Hematopoietic StemCell Transplantation . . . . . . . . . . . . . . . . . . . . . . . 53Juan Gea-Banacloche

    6 Risks and Epidemiology of Infectionsafter Solid Organ Transplantation . . . . . . . . . . . . 67Ingi Lee and Emily A. Blumberg

    7 Donor-Derived Infections: Incidence,Prevention and Management . . . . . . . . . . . . . . . . 77Michael G. Ison

    8 Transplant Infections in DevelopingCountries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90Clarisse M. Machado

    9 Risks and Epidemiology of Infectionsafter Heart Transplantation . . . . . . . . . . . . . . . . 104David DeNofrio and David R. Snydman

    10 Risks and Epidemiology of Infectionsafter Lung or HeartLung Transplantation . . . . 114Joan Gavald, Antonio Romn and Albert Pahissa

    11 Infections in Kidney Transplant Recipients . . . . 138Deepali Kumar and Atul Humar

    12 Risks and Epidemiology of Infectionsafter Pancreas or KidneyPancreasTransplantation . . . . . . . . . . . . . . . . . . . . . . . . . . 150Atul Humar and Abhinav Humar

    13 Risks and Epidemiology of Infections

    after Liver Transplantation . . . . . . . . . . . . . . . . 162Shimon Kusne and David C. Mulligan

    14 Risks and Epidemiology of Infections afterIntestinal Transplantation . . . . . . . . . . . . . . . . . 179Jorge D. Reyes and Michael Green

    Section III

    Specific Sites of Infection15 Pneumonia after Hematopoietic Stem

    Cell or Solid Organ Transplantation . . . . . . . . . 187Catherine Cordonnier and Isabel Cunningham

    16 Skin Infections after Hematopoietic StemCell or Solid Organ Transplantation . . . . . . . . . 203Mazen S. Daoud, Lawrence E. Gibson and W. P. Daniel Su

    17 Central Nervous System Infectionsafter Hematopoietic Stem Cell or SolidOrgan Transplantation . . . . . . . . . . . . . . . . . . . . 214Diana Averbuch and Dan Engelhard

    18 Gastrointestinal Infections after Solid Organor Hematopoietic Cell Transplantation . . . . . . . 236George B. McDonald and Gideon Steinbach

    Section IV

    Bacterial Infections19 Gram-Positive and Gram-Negative

    Infections after Hematopoietic StemCell or Solid Organ Transplantation . . . . . . . . . . 257Dan Engelhard

    20 Typical and Atypical MycobacteriumInfections after Hematopoietic StemCell or Solid Organ Transplantation . . . . . . . . . 282Jo-Anne H. Young and Daniel J. Weisdorf

    21 Other Bacterial Infections after HematopoieticStem Cell or Solid Organ Transplantation . . . . . 295

    J. Stephen Dummer and Tue Ngo

    Section V

    Viral Infections22 Cytomegalovirus Infection after Stem

    Cell Transplantation . . . . . . . . . . . . . . . . . . . . . . 311Morgan Hakki, Michael J. Boeckh and Per Ljungman

    23 Cytomegalovirus Infection after SolidOrgan Transplantation . . . . . . . . . . . . . . . . . . . . 328Raymund R. Razonable and Ajit P. Limaye

    ContentsContents

    xi

  • 7/24/2019 Transplant Infections -3E

    12/798

    24 EpsteinBarr Virus Infection andLymphoproliferative Disorders afterTransplantation . . . . . . . . . . . . . . . . . . . . . . . . . . 362Jutta K. Preiksaitis and Sandra M. Cockfield

    25 Herpes Simplex and Varicella-ZosterVirus Infection after Hematopoietic StemCell or Solid Organ Transplantation . . . . . . . . . 391

    John W. Gnann, Jr

    26 Infections with Human Herpesvirus6, 7,and 8 after Hematopoietic Stem Cellor Solid Organ Transplantation . . . . . . . . . . . . . 411Nina Singh

    27 Community-Acquired RespiratoryViruses after Hematopoietic Stem Cellor Solid Organ Transplantation . . . . . . . . . . . . . 421Janet A. Englund and Estella Whimbey

    28 Adenovirus Infection in AllogeneicStem Cell Transplantation . . . . . . . . . . . . . . . . . 447Susanne Matthes-Martin

    29 Adenovirus Infection in Solid Organ

    Transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . 459Michael Green, Michael G. Ison and Marian G. Michaels

    30 Polyoma and Papilloma Virus Infectionsafter Hematopoietic Cell or Solid OrganTransplantation . . . . . . . . . . . . . . . . . . . . . . . . . . 465Hans H. Hirsch

    31 Hepatic Infections after Solid OrganTransplantation . . . . . . . . . . . . . . . . . . . . . . . . . . 483Ed Gane

    32 Hepatitis B and C in HematopoieticStem Cell Transplant . . . . . . . . . . . . . . . . . . . . . 498Anna Locasciulli, Barbara Montante and Maria Beatrice Pinazzi

    Section VI

    Fungal Infections33 Yeast Infections after Hematopoietic

    Stem Cell Transplantation . . . . . . . . . . . . . . . . . 507Tamara Aghamolla, Brahm H. Segal and Thomas J. Walsh

    34 Yeast Infections after Solid OrganTransplantation . . . . . . . . . . . . . . . . . . . . . . . . . . 525Peter G. Pappas

    35 Mold Infections after HematopoieticStem Cell Transplantation . . . . . . . . . . . . . . . . . 537

    William J. Steinbach and Kieren A. Marr36 Aspergillus and Other Mold Infections

    after Solid Organ Transplant . . . . . . . . . . . . . . . 554Patricia Muoz, Maddalena Giannella, Almudena Burillo

    and Emilio Bouza

    37 Infections Caused by UncommonFungi in Patients UndergoingHematopoietic Stem Cell or SolidOrgan Transplantation . . . . . . . . . . . . . . . . . . . 586John W. Hiemenz, Andreas H. Groll and Thomas J. Walsh

    38 Endemic Mycoses after HematopoieticStem Cell or Solid Organ Transplantation . . . . . 607Carol A. Kauffman

    Section VII

    Other Infections39 Toxoplasmosis Following Hematopoietic

    Stem Cell Transplantation . . . . . . . . . . . . . . . . . 617Rodrigo Martino

    40 Toxoplasmosis after Solid OrganTransplantation . . . . . . . . . . . . . . . . . . . . . . . . . 624Jose G. Montoya and Mitchell R. Lunn

    41 Parasites after Hematopoietic StemCell or Solid Organ Transplantation . . . . . . . . . 632Roberta Lattes and Marcelo Radisic

    Section VIII

    Infection Control42 Infection Control Issues after

    Hematopoietic Stem Cell Transplantation . . . . 653Robin K. Avery and David L. Longworth

    43 Infection Control Issues after SolidOrgan Transplantation . . . . . . . . . . . . . . . . . . . 667Maria Teresa Seville, Sharon Krystofiak and Shimon Kusne

    Section IX

    Immune Reconstitution Strategies forPrevention and Treatment of Infections44 Vaccination of Transplant Recipients . . . . . . . . 691

    Per Ljungman

    45 Growth Factors and OtherImmunomodulators after Transplantation . . . . 705Jack W. Hsu and John R. Wingard

    46 Adoptive Immunotherapy withHerpesvirus-specific T Cells afterTransplantation . . . . . . . . . . . . . . . . . . . . . . . . . . 724Hermann Einsele, Max S. Topp, Stanley R. Riddell

    Section X

    Hot Topics47 Emerging and Rare Viral Infections

    in Transplantation . . . . . . . . . . . . . . . . . . . . . . . 745Staci A. Fischer

    48 Travel Medicine, Vaccines andTransplant Tourism . . . . . . . . . . . . . . . . . . . . . . . .756Camille Nelson Kotton

    Index 768

    xii Contents

  • 7/24/2019 Transplant Infections -3E

    13/798

    The lymphohematopoietic system is the only organ system in

    mammals that has the capacity for complete self-renewal.

    Therefore, donation of lymphohematopoietic stem cells does not

    result in a permanent loss for the donor. Reports on the therapeu-

    tic use of bone marrow to treat anemia associated with parasitic

    infections date back a century (1,2), but not until the observations

    on irradiation effects in Hiroshima and Nagasaki and the ensuingsystematic research into hematopoietic cell transplantation (HCT)

    in animal models were the principles of HCT established (1,3,4).

    In 1957, the first clinical transplant attempts of the modern

    era were undertaken (1,5,6). As predicted from animal studies,

    patients who underwent transplantation from allogeneic donors

    (i.e., individuals who were not genetically identical) developed

    graft-versus-host disease (GVHD) (4). Patients transplanted

    from syngeneic (monozygotic twin) donors generally did not

    develop GVHD, but many of them died from progressive

    leukemia, apparently because of a lack of the allogeneic graft-

    versus-leukemia (GVL) effect which had been described by Barnes

    and Loutit (7) in murine models. These studies immediately estab-

    lished that allogeneic HCT functioned as immunotherapy.

    Beginning in the late 1950s and early 1960s, Dausset et al.

    characterized the first histocompatibility antigens in humans

    (8). Epstein et al. were the first to show the relevance of those

    histocompatibility antigens for the development of GVHD in

    an outbred species (9). Initially, the only source of hematopoietic

    stem cells (HSC) in clinical use was bone marrow. However,

    cells harvested from peripheral blood, either after recovery

    from chemotherapy or after the administration of hematopoi-

    etic growth factors such as granulocyte-colony stimulating fac-

    tor (G-CSF), were shown to result in accelerated hematopoieticrecovery after autologous transplantation. These cells, as well as

    cord blood cells, are now being used with increasing frequency

    in allogeneic transplantation (10,11).

    RATIONALE AND INDICATIONSFOR HEMATOPOIETIC CELLTRANSPLANTATION

    Current indications for HCT are summarized in Table 1.1. The

    majority of HCT is performed to treat malignant diseases.

    Myelosuppression is the most frequent dose-limiting toxicity of

    the chemoradiotherapy used to treat malignancies. Infusion ofHSCautologous or allogeneicas a rescue procedure al-

    lows the dose escalation of cytotoxic therapy, such that toxicity

    in the next most sensitive organs (intestinal tract, liver, or lungs)

    becomes dose-limiting. This strategy, often referred to as high-

    dose therapy with stem cell rescue, has been used extensively in

    the past. However, progressive dose intensification, although

    possibly effective in disease eradication, has resulted in minimal,

    if any, improvement in survival because of an increase in

    therapy-related toxicity and mortality. These observations, com-

    bined with an increasing appreciation of the central role of im-

    munologic graft-versus-tumor (GVT) reactions in the success of

    SECTION I Introduction to Transplant Infections

    Introduction to HematopoieticCell Transplantation

    ANDREW R. REZVANI, H. JOACHIM DEEG

    1

    CHAPTER

    1

    TABLE 1.1 Categories of Disease Treated withHematopoietic Cell Transplantation

    Malignant

    Hematologic malignanciesAcute leukemiasChronic leukemias

    Myelodysplastic syndromesMyeloproliferative syndromesNon-Hodgkin lymphomaHodgkin lymphoma

    Plasma cell dyscrasia (e.g., multiple myeloma)

    Selected solid tumorsRenal cell carcinomaEwing sarcoma

    NeuroblastomaBreast, colon, ovarian, and pancreatic cancer

    (investigational)

    Nonmalignant

    Acquired

    Aplastic anemia and red cell aplasiasParoxysmal nocturnal hemoglobinuria

    Autoimmune disorders (e.g., multiple sclerosis, lupuserythematosus, systemic sclerosis, rheumatoid arthritis)

    CongenitalImmunodeficiency syndromes (e.g., SCID)

    HemoglobinopathiesCongenital anemias (e.g., Fanconi anemia)Storage diseases (e.g., mucopolysaccharidoses)Bone marrow failure syndromes (e.g., dyskeratosis

    congenita)Osteopetrosis

  • 7/24/2019 Transplant Infections -3E

    14/798

    allogeneic HCT, have led to new concepts of transplant condi-

    tioning (see Modalities for Transplant Conditioning) (12).

    Replacement therapy in patients with congenital or ac-

    quired disorders of marrow function, immunodeficiencies, or

    storage diseases represents a second indication for HCT.

    Patients with autoimmune diseases (e.g., rheumatoid arthritis

    or systemic sclerosis) can also be considered part of this cate-gory (13). In contrast to the benefit from GVT alloreactivity in

    the malignant setting, patients with these nonmalignant disor-

    ders are not thought to derive any benefit from alloreactivity

    beyond its graft-facilitating effect.

    Finally, HSC (or their more mature progeny) may be effec-

    tive vehicles for gene therapy (14) and for immunotherapy.

    Objectives of gene therapy include the replacement of defective

    or missing enzymes (e.g., adenosine deaminase, glucocerebrosi-

    dase) or of the defective gene (15,16). Experience with the use of

    allogeneic cells, often T lymphocytes, as immunologic bullets is

    more extensive. Donor lymphocyte infusion (DLI) for reinduc-

    tion of remission in patients with chronic myelogenous leukemia(CML) who have relapsed after HCT has been remarkably suc-

    cessful, leading to broader application of this approach. A modi-

    fication of this strategy is the use of genetically modified donor

    lymphocytes expressing a suicide gene, which may be activated

    to abrogate the adverse effects of DLI, particularly GVHD (17).

    The principle of immunotherapy is also exploited in

    reduced-intensity conditioning (RIC), also referred to as non-

    myeloablative or mini transplants (both terms, however, are

    misleading, as the end result is intended to be ablation of the

    disease, and a mini-transplant is still a full transplant, albeit with

    a lower-intensity conditioning regimen). In this approach, the

    intensity of the conditioning regimen has been reduced with the

    objective of preventing early mortality, and donor antihost reac-

    tivity has been enhanced to eliminate host cells (Fig. 1.1) (18).

    SOURCES OF HEMATOPOIETIC STEMCELLS AND DONOR SELECTION

    HSC can be obtained from a variety of donors and cellular

    compartments, including the bone marrow, peripheral blood,

    cord blood, and the fetal liver. The choice of stem cell source is

    dependent upon several factors. Although autologous marrowor peripheral blood stem cells (PBSC) are theoretically avail-

    able for every patient (feasibility has been reported even for

    patients with severe aplastic anemia), these would not be use-

    ful without genetic manipulation for genetically determined

    disorders, and would be suboptimal for malignant disorders,

    because of the concern of contamination with malignant cells

    and the lack of an allogeneic antitumor effect. An HLA-

    haploidentical donor (e.g., parent, sibling, child) is available

    for most patients, and, while clearly investigational at this

    time, early results show surprisingly low rates of GVHD and

    graft rejection (19).

    Generally, each sibling has a 25% chance of sharing the

    HLA genotype of a patient. Phenotypically matched donors

    can be identified among family members in about 1% of pa-

    tients, and somewhat less than 1% of patients will have a syn-

    geneic (identical twin) donor. The lack of an HLA-identical

    related donor in more than 70% of patients has led to the de-

    velopment of (a) large data banks of volunteer unrelated

    donors; (b) research into alternative allograft sources such as

    HLA-haploidentical family members and umbilical cord

    blood, as indicated earlier; and (c) techniques to purge autol-

    ogous cells of tumor contamination.

    Supported by the efforts of the National Marrow DonorProgram in the United States, the Anthony Nolan Appeal in

    the United Kingdom, and other groups internationally, more

    than 10 million volunteer donors have been typed for HLA-A

    and HLA-B, and a rapidly increasing number also for HLA-C,

    HLA-DR (DRB1), and HLA-DQ antigens (20). The proba-

    bility of finding a suitably HLA-matched donor for a white

    patient in North America is about 70% to 80%. This probabil-

    ity is lower for other ethnic groups, in part because of lower

    representation in the data bank and in part because of greater

    polymorphism of the HLA genes (21).

    Cord blood cells, generally not matched for all HLA anti-

    gens of the patient, are being used with increasing frequency(22), while fetal liver cells have been used only very rarely in

    recent years.

    Autologous marrow or PBSC can be purged of contami-

    nating malignant cells by chemical means or by antibodies

    that recognize tumor cells. However, slow engraftment and

    residual tumor cells that resist the purging regimen limit

    the usefulness of this approach. A complementary approach is

    aimed at purifying stem cells using specific antibodies

    to positively select cells bearing CD34, which is the closest

    that the research community has come to characterizing

    human HSC.

    2 Section I Introduction to Transplant Infections

    FIGURE 1.1. Commonly used conditioning regimens forhematopoietic cell transplantation, stratified by intensity, toxicity,and relative reliance on immunological graft-versus-tumoreffects. Abbreviations: GVT, graft-versus-tumor; CY, cyclophos-

    phamide; TBI, total body irradiation; Gy, gray; FLU, fludarabine;BU, busulfan; ATG, antithymocyte globulin; araC, cytarabine.

  • 7/24/2019 Transplant Infections -3E

    15/798

    TRANSPLANT PROCEDURE

    Transplant Conditioning

    Rationale for Conditioning

    1. To eradicate (ablate) the patients disease, or at least to re-

    duce the number of malignant or abnormal cells to below

    detectable levels (this applies to allogeneic, syngeneic, and

    autologous donors).

    2. To suppress the patients immunity and to prevent rejection

    of donor cells (this applies to allogeneic, but not to auto-

    logous, HCT). Immunosuppression is also needed in pre-

    paration for some syngeneic transplants, apparently to

    eliminate autoimmune reactivity which may interfere with

    sustained hematopoietic reconstitution.

    The notion that conditioning is necessary to generate

    space in the transplant recipient has essentially been aban-

    doned. Recent data show that donor cells, given in sufficient

    numbers, create their own space and proceed to repopulate the

    recipients marrow (23).

    Exceptions to the conditioning requirement exist in chil-

    dren with severe combined immunodeficiency (SCID), be-

    cause of the nature of the underlying disease, which does not

    allow them to reject transplanted donor cells, and in patients

    in whom even partial donor engraftment can completely cor-

    rect the genetic defect (24).

    Modalities for Transplant Conditioning

    Modalities used to prepare patients for HCT have been re-

    viewed extensively elsewhere (25,26); commonly used regi-mens are listed in Figure 1.1. In principle, conditioning for

    HCT may include the following approaches:

    1. Irradiation is in the form of total body irradiation (TBI),

    total lymphoid irradiation (27), or modifications thereof.

    Many conventional TBI regimens deliver 1200 to 1400 cGy

    over 3 to 6 days. In addition, bone-seeking isotopes (e.g.,

    holmium) and isotopes (e.g., 131I, 92Y) conjugated to mono-

    clonal antibodies (MAbs) directed at lymphoid or myeloid

    antigens (e.g., anti-CD20, CD45) are in use (28). TBI may

    also be a component of RIC regimens, usually at lower

    doses of 2 Gy (29).2. Chemotherapy (e.g., cyclophosphamide, 120 to 200 mg/kg

    over 2 to 4 days) is included in many conventional regimens.

    Busulfan (available in oral and intravenous formulations) at

    16 mg/kg (or lower doses), targeted to predetermined plasma

    levels, is often used in combination with cyclophosphamide.

    Other agents, including etoposide, melphalan, thiotepa, cy-

    tarabine, and more recently treosulfan (30), may be used ei-

    ther alone or in combination (with or without irradiation).

    3. Biologic reagents (e.g., antithymocyte globulin (ATG)) or

    MAbs directed at T-cell antigens or adhesion molecules sup-

    press recipient immunity. Others are directed at antigens

    expressed on the recipients malignant cells; in addition, cy-

    tokines or cytokine antagonists are being investigated. Anti-

    T-cell therapy predisposes the individual to viral infections,

    in particular cytomegalovirus (CMV) and the development

    of EpsteinBarr virus (EBV)-related lymphoproliferative

    disorders (PTLD) after transplantation (31).

    4. T-cell therapy is based on the observation that broad T lym-phocyte depletion of donor marrow resulted in graft fail-

    ure. This has led to protocols of selective T-cell add-back to

    ensure engraftment. The observation that DLI was effec-

    tive in inducing remission in a proportion of patients who

    had experienced relapse after HCT renewed the interest in

    exploiting T-cell therapy for the treatment of leukemia.

    Other indications for T-cell therapy are viral infections

    such as CMV (32) or EBV, especially with the development

    of PTLD in the latter (33).

    Other procedures involve plasmapheresis of the recipi-

    ents blood to remove isoagglutinins directed against thedonors ABO blood group or the removal of plasma from the

    donor marrow to remove the isoagglutinins directed at recipi-

    ent cells. Alternatively, the donor red blood cells with which

    recipient antibodies may react can be removed, thus minimiz-

    ing transfusion reactions. Due to the procedure by which they

    are obtained, these additional manipulations are generally not

    required with PBSC.

    Marrow Harvest

    The marrow donor receives general or regional (e.g., epidural,

    spinal) anesthesia, and, under sterile conditions, multiple aspi-

    rates of marrow are obtained from both posterior iliac crests

    (34). Additional potential aspiration sites are the anterior iliac

    crests and the sternum. Approximately 10 to 15 mL/kg of

    donor weight is collected. If no ABO incompatibility exists

    and if the marrow is not to be subjected to any in vitro purging

    procedure, the resulting cell suspension is infused intra-

    venously without manipulation.

    Alternative Stem Cell Sources

    HSC circulate at low concentrations in blood (35). Their fre-

    quency increases dramatically during the recovery phase fol-

    lowing cytotoxic therapy, and after the administration ofrecombinant hematopoietic growth factors such as G-CSF

    which dislodge cells from the marrow. Peak blood concentra-

    tions of CD34 cells are typically reached on day 4 to 5 after

    initiating G-CSF. A single leukapheresis may be sufficient to

    harvest the number of HSC required for a transplant. For au-

    tologous procedures, the goal is to collect at least 2 to 5

    106 CD34 cells/kg recipient weight; for allogeneic trans-

    plants, the goal is 5 to 8 106 CD34 cells/kg, although the

    optimum dose has not been determined (36).

    Umbilical cord blood represents a segment of the periph-

    eral circulation of the fetus and is easily accessible (37). Also,

    Chapter 1 Introduction to Hematopoietic Cel l Transplantation 3

  • 7/24/2019 Transplant Infections -3E

    16/798

    cord blood cells are less immunocompetent than adult cells,

    and might therefore carry a lower risk of inducing GVHD

    than adult cells. The concentration of HSC in umbilical cord

    blood is high, but the small volume that is usually available

    (80150 mL) initially limited the use of these cells to children

    and smaller adults. In larger adults, approaches have included

    the use of two cord blood units to ensure adequate cell doseand engraftment (11), as well as ex vivo expansion of

    hematopoietic precursors in umbilical cord blood units for in-

    fusion together with an unmanipulated cord blood unit (38).

    Purging

    Several rationales exist for purging collected donor cells or

    fractionating them into subpopulations. In the autologous set-

    ting, the goal is to eliminate contaminating tumor cells, either

    by negative selection (removal of tumor cells with antibodies

    or physicochemical means) or by positive selection (purifica-

    tion of CD34 cells from the graft). Conversely, one may want

    to retain certain populations (e.g., CD4 cells) with potentialfor later uses such as posttransplant DLI.

    Hematopoietic Stem Cell Infusion:

    The Actual Transplant

    Donor cells are infused intravenously via an indwelling cen-

    tral line, often a Hickman catheter. Directed by surface mole-

    cules which interact with receptors on endothelial cells, HSC

    home to the marrow cavity. The actual infusion of stem cells is

    generally uneventful, though it can occasionally cause tran-

    sient mild hypotension or hypersensitivity reactions.

    CARE AFTER TRANSPLANTATION

    Complications of HCT, including infections, are related to sev-

    eral factors: the underlying disease, the preparative regimen,

    and the interactions of donor cells with recipient tissue (GVHD

    with immunosuppression and end-organ damage). All patients

    experience at least transient pancytopenia, although this may be

    mild with RIC regimens. Patients undergoing high-dose condi-

    tioning generally develop severe pancytopenia, including neu-

    tropenia, within days after completion of conditioning. This

    period may last 2 to 4 weeks with marrow allografts, 10 to12 days with mobilized PBSC grafts, or 4 to 6 weeks with

    umbilical cord blood grafts. The period of neutropenia ends

    with engraftment of the donor cells, clinically defined by stable

    increases in the white blood cell count. Cytopenias are less

    pronounced after RIC, and the pattern of engraftment may be

    less apparent in the peripheral white blood cell count.

    Engraftment in these patients is generally documented by

    demonstrating donor chimerism by cytogenetic or molecular

    means in peripheral blood leukocytes and bone marrow.

    Most patients prepared with high-dose regimens require

    transfusion support with platelets, red blood cells, or both.

    Transfusion requirements are substantially reduced in patientsprepared with RIC regimens, because the nadir of cells often

    is in a range in which no transfusions are required (39).

    Erythropoietin administration after HCT accelerates reticulo-

    cyte recovery and moderately reduces red blood cell transfu-

    sion requirements in patients undergoing allogeneic (though

    not autologous) HCT (34).

    Quantitative and functional deficiencies of granulocytes

    and T-lymphocytes for various periods after HCT are respon-

    sible for most of the infectious complications seen after HCT

    (Fig. 1.2). Although all patients receive prophylactic antimi-

    crobials, granulocyte transfusions are not routinely given.Laminar air flow (LAF) rooms and gastrointestinal deconta-

    mination may reduce the frequency of infections and the du-

    ration of febrile episodes, but neither is used routinely because

    of the high cost of LAF and the availability of effective broad-

    spectrum antibiotics (40).

    The most widely used modality of GVHD prophylaxis is

    the in vivo administration of immunosuppressive agents, such

    as methotrexate, cyclosporine (CSP), glucocorticoids, tacrolimus

    (FK506), mycophenolate mofetil (MMF), sirolimus and others,

    either alone or in combination (41). At many institutions, the

    current standard is a combination of a calcineurin inhibitor

    with methotrexate or MMF, but several other combinations areused. Due to the nonselectivity of these agents, recipients are

    broadly immunosuppressed and thus susceptible to infections.

    In vitro T-lymphocyte depletion of donor marrow may obviate

    the need for immunosuppressive treatment after HCT; how-

    ever, the elimination of mature T-cells is associated with a risk

    of rejection, delayed immunologic reconstitution, an increased

    risk of PTLD, and, for some disorders, disease recurrence.

    Both immunodeficiency and therapeutic immunosuppression

    predispose the patient to infections. Whether the selective re-

    moval of naive T-cells will lead to successful transplants with-

    out GVHD remains under investigation.

    4 Section I Introduction to Transplant Infections

    0Weeks Months

    Neutrophils, monocytes,NK cells

    B cells, CD8 T cells

    CD4 T cells

    Plasma cells, dendritic cells

    Upper normal limit

    Lower normal limit

    Years posttransplantImm

    unecellcounts(%n

    ormal)

    20

    40

    60

    80

    100

    120

    140 Graft infusion

    FIGURE 1.2. Approximate trends in immune cell counts aftermyeloablative hematopoietic cell transplantation. With the useof reduced-intensity conditioning, nadirs are higher and occurlater. These recovery rates may be influenced by clinical vari-ables such as graft-versus-host disease, stem cell source, and

    patient age. (Adapted from Storek J. Immunological reconsti-tution after hematopoietic cell transplantationits relation tothe contents of the graft (Review). Expert Opin Biol Ther.2008;8:583597.)

  • 7/24/2019 Transplant Infections -3E

    17/798

    HEMATOLOGIC RECOVERY

    A high-risk period for infections in patients conditioned with

    high-dose regimens exists early after transplantation, when gran-

    ulocytopenia develops due to the decline in endogenous marrow

    function while donor cells are not yet proliferating. If the donor

    marrow is T-cell depleted, the recovery may be even more pro-tracted. If the granulocyte count at day21 after transplantation

    is less than 200 cells/L, patients are generally given G-CSF or

    granulocyte-macrophage colony-stimulating factor. After PBSC

    transplants, engraftment (defined by 500 granulocytes/L)

    occurs as early as day 9 or 10, thus clearly shortening the

    length of granulocytopenia. Rather slower recovery (over several

    weeks) may be seen with cord blood transplants (42).

    One advantage of RIC regimens is the slow decline of pa-

    tient cells, so that donor cells begin to recover before patient

    cells reach their nadir. Consequently, several groups have re-

    ported lower rates of early infection after RIC conditioning as

    compared with high-dose conditioning (43,44).

    IMMUNOLOGIC RECOVERY

    All components of the innate and adaptive immune systems

    are deficient after HCT. Cell-mediated immunity, chemo-

    taxis, and neutrophil function are severely impaired even after

    autologous transplants. The development of GVHD substan-

    tially impairs immune reconstitution, through a combination

    of direct graft-versus-host toxicity to the thymus resulting in

    altered T-cell selection and use of immunosuppressive thera-

    pies to treat GVHD. Thus, optimal immune reconstitution

    can only occur in the absence of GVHD (45).

    Uncomplicated Recovery

    Shortly after HCT, damaged epithelial barriers facilitate the

    penetration of pathogenic bacterial or fungal organisms.

    Mucosal surfaces begin to heal within a week or two of comple-

    tion of high-dose conditioning, helped by the recovery of granu-

    locytes and their scavenging function, even though phagocytosis

    and superoxide production may still be impaired. After the

    transplant, the volume and immunoglobulin content of salivaalso improve with time. Even with uncomplicated recovery,

    T- and B-cell-mediated immune responses against viral, bac-

    terial, fungal, and other organisms are broadly suppressed; nat-

    ural killer (NK) cells recover more quickly. To some extent, the

    pattern of immune recovery is dependent on the immunity of

    the donor from whom the transplanted cells originated. The

    pattern of immunocompetence is also influenced by the recipi-

    ents prior antigen exposure, whether to the pathogen itself or in

    the form of a vaccine. Much of the literature on immune recon-

    stitution after allogeneic HCT describes patients who were

    prepared with high-dose conditioning. While the use of RIC

    regimens has increased rapidly in recent years, there are fewer

    data available on immune recovery in this setting, and the im-

    pact of RIC on immune reconstitution remains somewhat un-

    clear. Preliminary studies suggest that the tempo of immune

    recovery may be faster after RIC (46), but late immune function

    may be similar to that seen after high-dose conditioning (47).

    B Cells

    B-cell numbers are undetectable or very low shortly after

    high-dose conditioning, but may rise to supranormal levels by

    1 to 2 years (48). Recovery is faster with autologous than with

    allogeneic HCT; memory B cells lag behind naive cells. Early,

    but not late, recovery for both populations is faster for PBSC

    recipients than for marrow recipients (49). The B-cell compart-

    ment is generally replaced completely by donor-derived cells,

    except in patients with TB SCID, in whom the recipients

    B cells tend to persist (50). Nonetheless, some antibodies of host

    origin (e.g., isoagglutinins) that are derived from long-livedplasma cells may be detectable for months or even years after

    HCT. Persistently low B-cell counts after HCT may predict

    a high risk of infection (51). In the era of targeted therapy,

    treatment with B-cell-directed monoclonal antibodies such

    as rituximab prior to HCT may also impair B-cell reconstitu-

    tion, though relatively little is known about the long-term

    implications (52).

    During recovery, fewer B cells express CD25 and CD62L;

    more express CD9c, CD38, IgM, and IgD; and the antigen

    density is increased (as in neonatal B cells). CD5 cells may or

    may not be increased. Immunoglobulin gene usage appears to

    be restricted shortly after HCT and to be skewed toward the

    V-segments that are frequently used in neonatal B cells (e.g.,

    VH6). Concordantly, the antibody repertoire is restricted (53).

    IgG and IgA production may be abnormal for 1 to 2 years

    after HCT. Serum isotype levels after grafting recover in the

    same sequence as they evolve in neonates (i.e., IgM, IgG1, and

    IgG3 recover early, but IgG2, IgG4, and IgA may not follow

    until much later) (54). Many of the early antibodies are autoan-

    tibodies, or else have irrelevant specificities (55). Antibodies

    with relevant specificities recover only if the antigen is en-

    countered, and they recover more quickly if both patient and

    donor are immune (56). At 3 months after HCT, total IgG lev-els in recipients of allogeneic PBSC tend to be lower than those

    in marrow recipients. Antibodies to polysaccharide antigens

    tend to recover later than those directed at proteins. B-cell

    counts and IgM levels may recover more quickly after RIC as

    compared to high-dose conditioned patients, though IgA

    recovery is delayed in both groups (46). In addition to quanti-

    tative deficits in B-cell number and immunoglobulin levels,

    the B-cell pool early after HCT is marked by qualitative

    functional impairment. Isotype switching is deficient in the

    absence of effective T-cell help. Additionally, B cells from

    transplant recipients have a decreased capacity for somatic

    Chapter 1 Introduction to Hematopoietic Cel l Transplantation 5

  • 7/24/2019 Transplant Infections -3E

    18/798

    hypermutation independent of T-cell help, suggesting an

    intrinsic or environmental defect (57). Thus, B-cell deficits

    after HCT are comprised of at least three factors: low B-cell

    numbers, decreased T-cell help, and intrinsic defects such as

    impaired somatic hypermutation (54).

    Antibody responses to vaccination are almost universally

    lower than are those of normal controls, and repeated boostersare required (58). Responses are better in younger individuals

    and in those with T-cell-replete grafts; this may be related to

    CD4 recovery, which is faster in younger individuals. The pol-

    icy has been to delay revaccination until 1 to 2 years after HCT

    to minimize the risk of potential side effects and to increase

    the probability of antibody responses.

    T Cells

    CD4 Cells

    The number of CD4 cells is low for 1 to 3 months after high-

    dose conditioning, and rises slowly toward normal over several

    years. This rise is faster in children than in adults (58). The

    kinetics are similar following both autologous and allogeneic

    transplants. Early in the process, most cells are memory T cells;

    naive T cells follow quite gradually, particularly in older

    patients. This might be related to diminished thymic function,

    although the thymus appears to play some role in T-cell reconsti-

    tution even in older patients (59). After PBSC transplantations,

    both naive and memory CD4 T cells are more abundant than

    after marrow transplantations. Early after HCT, most CD4

    T cells are derived from transplanted mature T cells and T-cell

    precursors; later, they are stem-cell-derived, at least in pediatricpatients. CD4 T-cell reconstitution may occur more rapidly

    after RIC as compared to high-dose conditioning (46).

    CD4 T cells generally express CD11a, CD29, CD45RO,

    and HLA-DR and less CD28, CD45RA, and CD62L, consis-

    tent with the prominence of memory cells (58). Responses to

    polyclonal stimuli are low. Proliferative responses to fre-

    quently encountered antigens (e.g., Candida species) tend to

    normalize over 1 to 5 years, whereas responses to unlikely

    antigens (e.g., tetanus) remain subnormal. Responses to

    neoantigens (e.g., dinitrochlorobenzene) and recall antigens

    (e.g., mumps) are abnormally low for 2 to 3 years after HCT.

    CD8 Cells

    CD8 T cells are low for 2 to 3 months after HCT; subse-

    quently, they rise quickly, resulting in an inversion of the typi-

    cal CD4:CD8 ratio (58). These CD8 cells are largely memory

    cells expressing CD11a, CD11b, CD29, CD57, HLA-DR, and

    CD45RO but little CD28, CD45RA, and CD62L. The pres-

    ence of a CD11bCD57CD28 phenotype suggests anergic

    or suppressive CD8 cells. CD8 cells appear to be derived

    from transplanted T cells and stem cells.

    CMV-specific or EBV-specific CD8 cells can be trans-

    ferred successfully to a recipient, and they may persist for at

    least 18 months (60,61). Even established and refractory CMV

    infections can be treated effectively by the infusion of ex-

    panded CMV-specific CD8 donor cells (62). The logistical

    difficulty of generating CMV-specific cells for clinical use has

    been a barrier to the wide application of this approach.

    However, several groups have reported progress in developing

    simpler and more scalable means of producing virus-specificdonor T cells for infusion (63,64).

    The role of immunoregulatory CD4CD25 T cells

    (Treg) in clinical transplantation remains to be fully estab-

    lished (65).

    Antigen-Presenting Cells

    Monocytes reach normal levels within 1 month after high-dose

    conditioning, although their function may remain impaired for

    a year (66). G-CSF-mobilized PBSC contain large numbers of

    monocytes with altered cytokine profiles that may suppress al-

    logeneic T-cell responses. G-CSF-mobilized monocytes appearto settle in tissue in the early posttransplantation period.

    The reconstitution of dendritic cells (DC), their matura-

    tion, and the development of DC1 and DC2 have been incom-

    pletely characterized. DC precursors in the blood recover

    within 6 months, and DC reconstitution appears to be a clini-

    cally important event. Low numbers of DC at 1 month after

    reduced-intensity HCT have been associated with an in-

    creased risk of mortality and disease relapse; CD16 DC

    counts at 3 months were also strongly prognostic (67). A sepa-

    rate investigation found that low numbers of plasmacytoid

    DC at 3 months after HCT were associated with higher risks

    of infection and death (68). Langerhans cell levels are low in

    the early posttransplantation period, but return to normal by

    6 months. Follicular DC are reconstituted rather slowly,

    which may contribute to the delayed return to function of the

    germinal centers and memory B cells (58).

    Natural Killer Cells

    NK cells recover rapidly after HCT. With the recognition of

    the killer inhibitory receptor, renewed interest in these cells

    has been seen because of their possible function in engraftment

    and the prevention of relapse (69). Robust NK-cell reconstitu-tion after allogeneic HCT has been associated with reduced

    relapse and improved survival (70).

    GRAFT-VERSUS-HOST DISEASE ANDGRAFT-VERSUS-LEUKEMIA EFFECT

    Acute and chronic GVHD occur in 10% to 50% and 20% to

    50%, respectively, of patients after HLA-identical sibling

    HCT, and in 50% to 90% and 30% to 70%, respectively, of

    patients who undergo transplantation from alternative donors

    6 Section I Introduction to Transplant Infections

  • 7/24/2019 Transplant Infections -3E

    19/798

    (71,72). Without prophylaxis, virtually all recipients of allo-

    geneic transplants develop GVHD (73). Acute GVHD is the

    strongest risk factor for the development of chronic GVHD

    (74,75). Acute GVHD may occur within days (e.g., among

    HLA-nonidentical recipients) or by 3 to 5 weeks after HLA-

    identical transplantation following high-dose conditioning.

    The main target organs are the immune system, skin, liver,and intestinal tract. Only the skin, liver, and intestinal tract are

    generally considered in GVHD grading systems (76).

    Importantly, after RIC, classic manifestations of acute GVHD

    may develop several months after HCT and may overlap con-

    siderably with those of chronic GVHD (77). Features of

    chronic GVHD may be present as early as 50 to 60 days after

    HCT. Therefore, recently developed NIH consensus criteria

    distinguish acute from chronic GVHD on the basis of pathol-

    ogy and biology, rather than time of onset (78).

    HCT is unique in that donor/host tolerance frequently

    develops over time, to the point that maintenance immuno-

    suppression can often be discontinued. In patients withoutGVHD, all immunosuppressive medication is generally

    stopped by 6 months to 1 year after HCT. Even chronic

    GVHD is not necessarily a lifelong condition; many patients

    with chronic GVHD develop tolerance and resolution of

    GVHD over time, and are ultimately able to discontinue im-

    munosuppressive treatment (79,80).

    The immunopathophysiology of GVHD is complex.

    The initial damage to host tissue is induced by the transplant-

    conditioning regimen (81). The subsequent development of

    acute GVHD requires antigen presentation; Shlomchik et al.

    showed that host DC play a pivotal role in this process (82).

    Interactions of major histocompatibility complex (MHC)

    antigens (with bound peptides derived from minor histocom-

    patibility antigens) and T-cell receptors lead to activation, clonal

    expansion, and differentiation of donor T cells. Accessory T-cell

    surface molecules, such as CD4 or CD8, also contribute to the

    immunologic synapsis between T cells and antigen-presenting

    cells. The effector phase leads to host-cell destruction via in-

    flammatory signals, cytolytic effects, and programmed cell

    death (apoptosis). Inflammatory cytokines, which are primarily

    released from the gut, allow the transfer of endotoxins and

    lipopolysaccharides (LPS) into the circulation, triggering

    macrophage activation. The result is the further production ofcytokines, such as tumor necrosis factor (TNF) and inter-

    leukin 1 (IL-1) (83), leading to target cell death and the expres-

    sion of costimulatory molecules, such as CD80, CD86, and

    MHC class II antigens, on DC; T-cell stimulation; and the re-

    lease of T helper-1 (Th1) cytokines (IL-2, interferon- (IFN-)).

    Recent experiments also emphasize the role of other cy-

    tokines, particularly TNF, IL-15, and IL-18 (84). In mouse

    mod


Recommended