You are on page 1of 476

Antibodies

for Infectious Diseases


Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
Antibodies
for Infectious Diseases
Antibodies
for Infectious Diseases
Edited by

James E. Crowe, Jr.


Vanderbilt University Medical Center
Nashville, Tennessee

Diana Boraschi
National Research Council
Napoli, Italy

AND

Rino Rappuoli
Novartis Vaccines
Siena, Italy

Washington, DC
Copyright 2015 American Society for Microbiology. All rights reserved. No part of this publication
may be reproduced or transmitted in whole or in part or reused in any form or by any means,
electronic or mechanical, including photocopying and recording, or by any information storage and
retrieval system, without permission in writing from the publisher.

Disclaimer: To the best of the publishers knowledge, this publication provides information concerning
the subject matter covered that is accurate as of the date of publication. The publisher is not providing
legal, medical, or other professional services. Any reference herein to any specific commercial
products, procedures, or services by trade name, trademark, manufacturer, or otherwise does not
constitute or imply endorsement, recommendation, or favored status by the American Society for
Microbiology (ASM). The views and opinions of the author(s) expressed in this publication do not
necessarily state or reflect those of ASM, and they shall not be used to advertise or endorse any
product.

Library of Congress Cataloging-in-Publication Data

Antibodies for infectious diseases / edited by James E. Crowe, Jr., Vanderbilt University Medical Center,
Nashville, Tennessee, Diana Boraschi, National Research Council, Napoli, Italy, and Rino Rappuoli,
Novartis Vaccines, Siena, Italy.
pages cm
Includes bibliographical references and index.
ISBN 978-1-55581-735-0 (alk. paper)
1. Immunoglobulins. 2. Communicable diseases--Immunological aspects. I. Crowe, James E., Jr., editor.
II. Boraschi, D. (Diana), editor. III. Rappuoli, Rino, editor.
QR186.7.A534 2015
616.0798--dc23
2015009356
eISBN: 978-1-55581-741-1
doi:10.1128/9781555817411
10 9 8 7 6 5 4 3 2 1

All Rights Reserved


Printed in the United States of America

Address editorial correspondence to ASM Press, 1752 N St., N.W.,


Washington, DC 20036-2904, USA
Send orders to ASM Press, P.O. Box 605, Herndon, VA 20172, USA
Phone: 800-546-2416; 703-661-1593
Fax: 703-661-1501
E-mail: books@asmusa.org
Online: http://estore.asm.org

Cover image: Cancer cell with high details (source: 123RF)


Contents

Contributorsix
Prefacexvii

Introduction
1 History and Practice: Antibodies in Infectious Diseases3
Adam Hey

General Features of Immunoglobulins


2 Functions of Antibodies25
Donald N. Forthal
3 Antibody Structure49
Robyn L. Stanfield and Ian A. Wilson
4 The Role of Complement in Antibody Therapy for Infectious Diseases63
Peter P. Wibroe, Shen Y. Helvig, and S. Moein Moghimi
5 Immunoglobulin E and Allergy: Antibodies in Immune Inflammation
andTreatment75
Sophia N. Karagiannis, Panagiotis Karagiannis, Debra H. Josephs, Louise Saul, Amy E. Gilbert,
Nadine Upton, and Hannah J. Gould

Antibody Discovery Approaches


6 Phage and Yeast Display105
Jared Sheehan and Wayne A. Marasco
7 Efficient Methods To Isolate Human Monoclonal Antibodies from Memory
B Cells and Plasma Cells129
Davide Corti and Antonio Lanzavecchia
8 Use of Human Hybridoma Technology To Isolate Human Monoclonal Antibodies141
Scott A. Smith and James E. Crowe, Jr.
9 Humanized Mice for Studying Human Immune Responses and Generating Human
Monoclonal Antibodies157
Ramesh Akkina
10 Antibodies: Computer-Aided Prediction of Structure and Design of Function173
Alexander M. Sevy and Jens Meiler

v
vicontents

Pathogen-Specific Antibodies
11 Antibodies Targeting the Envelope of HIV-1193
Luzia M. Mayr and Susan Zolla-Pazner
12 Committing the Oldest Sins in the Newest Kind
of WaysAntibodies Targeting the Influenza Virus Type A
Hemagglutinin Globular Head209
Jens C. Krause and James E. Crowe, Jr.
13 Prevention of Respiratory Syncytial Virus Infection:
From Vaccine to Antibody221
Kelly Huang and Herren Wu
14 Human Metapneumovirus237
Jennifer E. Schuster and John V. Williams
15 Dengue Antibody-Dependent Enhancement:
Knowns and Unknowns249
Scott B. Halstead
16 Immunotherapeutic Approaches To Prevent Cytomegalovirus-
Mediated Disease273
Edith Acquaye-Seedah, Zachary P. Frye, and Jennifer A. Maynard
17 Rotavirus289
Manuel A. Franco and Harry B. Greenberg
18 Bacterial ToxinsStaphylococcal Enterotoxin B303
Bettina C. Fries and Avanish K. Varshney

Technical Advances
19 Antibody Engineering321
Kin-Ming Lo, Olivier Leger, and Bjrn Hock
20 High-Throughput DNA Sequencing Analysis of Antibody
Repertoires345
Scott D. Boyd and Shilpa A. Joshi
21 Antibody Informatics: IMGT, the International ImMunoGeneTics
Information System363
Marie-Paule Lefranc
22 Probing Antibody-Antigen Interactions381
Guocheng Yang, Stefanie N. Velgos, Shanta P. Boddapati,
and Michael R. Sierks
23 Radiolabeled Antibodies for Therapy of Infectious Diseases399
Ekaterina Dadachova and Arturo Casadevall
contents vii

Alternate Systems for Expression


24 Plant-Derived Monoclonal Antibodies for Prevention and Treatment
of Infectious Disease413
Andrew Hiatt, Kevin J. Whaley, and Larry Zeitlin
25 Vector-Mediated In Vivo Antibody Expression427
Bruce C. Schnepp and Philip R. Johnson

Index441
Contributors

Edith Acquaye-Seedah
Department of Biochemistry
University of Texas at Austin
Austin, TX 78712
Ramesh Akkina
Department of Microbiology, Immunology, and Pathology
Colorado State University
1619 Campus Delivery
Fort Collins, CO 80523
Shanta P. Boddapati
Department of Biomedical Engineering
Oregon Health and Science University
3181 SW Sam Jackson Park Road
Portland, OR 97239
Scott D. Boyd
Department of Pathology
Stanford University
Stanford, CA 94305
Arturo Casadevall
Departments of Microbiology and Immunology, and Medicine
Albert Einstein College of Medicine of Yeshiva University
1695A Eastchester Road
Bronx, NY 10461
Davide Corti
Humabs BioMed SA
via Mirasole 1
Bellinzona, 6500
Switzerland
James E. Crowe, Jr.
School of Medicine
Vanderbilt University
Nashville, TN 37232

ix
xCONTRIBUTORS

Ekaterina (Kate) Dadachova


Departments of Radiology, Microbiology and Immunology
Albert Einstein College of Medicine of Yeshiva University
1695A Eastchester Road
Bronx, NY 10461
Donald N. Forthal
Department of Infectious Diseases
3044 Hewitt Hall
University of California, Irvine
Irvine, CA 92617
Manuel A. Franco
Facultad de Ciencias y MedicinaPontificia
Universidad Javeriana
Oficina 306, Edificio 50
Carrera 7 # 40-62
Bogot
Colombia
Bettina C. Fries
Department of Medicine/Infectious Diseases and
Department of Microbiology and Immunology
Albert Einstein College of Medicine
Bronx, NY 10461
Zachary P. Frye
Department of Chemical Engineering
University of Texas at Austin
Austin, TX 78712
Amy E. Gilbert
Cutaneous Medicine and Immunotherapy Unit
St. Johns Institute of Dermatology
Division of Genetics and Molecular Medicine & NIHR
Biomedical Research Centre at Guys and St. Thomass Hospitals and
Kings College London School of Medicine
Guys Hospital
London SE1 9RT
United Kingdom
Hannah J. Gould
Randall Division of Cell and Molecular Biophysics
Division of Asthma, Allergy and Lung Biology
MRC and Asthma UK Centre for Allergic Mechanisms of Asthma
Kings College London
New Hunts House
Guys Campus
London SE1 1UL
United Kingdom
CONTRIBUTORS xi

Harry B. Greenberg
Departments of Medicine and Microbiology and Immunology
Stanford University
School of Medicine and the VAPAHCS
Palo Alto, CA 94305
Scott B. Halstead
Department of Preventive Medicine and Biometrics
Uniformed Services University of the Health Sciences
Bethesda, MD 20814
Shen Y. Helvig
Department of Pharmacy
Center for Pharmaceutical Nanotechnology and Nanotoxicology
Faculty of Health and Medical Sciences
University of Copenhagen
Copenhagen, DK-2100
Denmark
Adam Hey
Preclinical Safety, Biologics
Novartis AG
Basel
Switzerland
Andrew Hiatt
Mapp Biopharmaceutical, Inc.
6160 Lusk Blvd. #C105
San Diego, CA 92121
Bjrn Hock
Department of Protein Engineering and Antibody Technologies
Merck Serono, Merck KgaA
Frankfurter Str. 250
D-64293 Darmstadt
Germany
Kelly Huang
Department of Infectious Disease
MedImmune, LLC
One MedImmune Way
Gaithersburg, MD 20878
Philip R. Johnson
The Childrens Hospital of Philadelphia
Abramson Research Center
Philadelphia, PA 19104
xiiCONTRIBUTORS

Debra H. Josephs
Cutaneous Medicine and Immunotherapy Unit
St. Johns Institute of Dermatology
Division of Genetics and Molecular Medicine & NIHR
Biomedical Research Centre at Guys and St. Thomass Hospitals
Kings College London School of Medicine
Guys Hospital
London SE1 9RT
United Kingdom
Shilpa A. Joshi
Department of Pathology
Stanford University
Stanford, CA 94305
Sophia N. Karagiannis
Cutaneous Medicine and Immunotherapy Unit
St. Johns Institute of Dermatology
Division of Genetics and Molecular Medicine & NIHR Biomedical Research
Centre at Guys and
St. Thomass Hospitals
Kings College London School of Medicine
Guys Hospital
London SE1 9RT
United Kingdom
Panagiotis Karagiannis
Cutaneous Medicine and Immunotherapy Unit
St. Johns Institute of Dermatology
Division of Genetics and Molecular Medicine & NIHR Biomedical Research
Centre at Guys and
St. Thomass Hospitals
Kings College London School of Medicine
Guys Hospital
London SE1 9RT
United Kingdom
Jens C. Krause
Childrens Hospital
University of Freiburg Medical Center
79106 Freiberg
Germany
Antonio Lanzavecchia
Institute for Research in Biomedicine
via Vincenzo Vela 6
Bellinzona, 6500
Switzerland
CONTRIBUTORS xiii

Marie-Paule Lefranc
Laboratoire dImmunoGntique Molculaire LIGM
IMGT, the international ImMunoGeneTics information system
Institut de Gntique Humaine IGH
Universit Montpellier 2
UPR CNRS 1142
Montpellier, 34396
cedex 5, 40202
France
Olivier Leger
Department of Protein Engineering and Antibody Technologies
Merck Serono S.A.--Geneva
9, chemin des Mines
1202 Geneva
Switzerland
Kin-Ming Lo
Department of Protein Engineering and Antibody Technologies
EMD Serono Research Institute
45A Middlesex Turnpike
Billerica, MA 01821
Wayne A. Marasco
Department of Cancer Immunology and AIDS
Dana-Farber Cancer Institute
Harvard Medical School
Boston, MA 02215
Jennifer A. Maynard
Department of Chemical Engineering
University of Texas at Austin
Austin, TX 78712
Luzia M. Mayr
INSERM U1109
Universit de Strasbourg
3 Rue Koeberl
67000 Strasbourg
France
Jens Meiler
Department of Chemistry and Center for Structural Biology
Vanderbilt University
Nashville, TN 37212
xivCONTRIBUTORS

S. Moein Moghimi
Department of Pharmacy
Center for Pharmaceutical Nanotechnology and Nanotoxicology
Faculty of Health and Medical Sciences
University of Copenhagen
Copenhagen, DK-2100
Denmark
Susan Zolla-Pazner
New York University School of Medicine
550 First Avenue
New York, NY 10016
Louise Saul
Cutaneous Medicine and Immunotherapy Unit
St. Johns Institute of Dermatology
Division of Genetics and Molecular Medicine & NIHR Biomedical Research
Centre at Guys and
St. Thomass Hospitals
Kings College London School of Medicine
Guys Hospital
London SE1 9RT
United Kingdom
Bruce C. Schnepp
The Childrens Hospital of Philadelphia
Abramson Research Center
Philadelphia, PA 19104
Jennifer E. Schuster
Department of Pediatrics
Childrens Mercy Hospital
Kansas City, MO 64108-4619
Alexander M. Sevy
Department of Chemistry and Center for Structural Biology
Vanderbilt University
Nashville, TN 37212
Jared Sheehan
Department of Cancer Immunology and AIDS
Dana-Farber Cancer Institute
Harvard Medical School
Boston, MA 02215
Michael R. Sierks
Department of Chemical Engineering
Arizona State University
Tempe, AZ 85287-6006
CONTRIBUTORS xv

Scott A. Smith
The Vanderbilt Vaccine Center and the Department of Medicine
Vanderbilt University Medical Center
Nashville, TN 37232
Robyn L. Stanfield
Department of Molecular Biology
The Scripps Research Institute
10550 North Torrey Pines Road
La Jolla, CA 92037
Nadine Upton
Randall Division of Cell and Molecular Biophysics
Division of Asthma, Allergy and Lung Biology
MRC and Asthma UK Centre for Allergic Mechanisms of Asthma
Kings College London
New Hunts House
Guys Campus
London SE1 1UL
United Kingdom
Avanish K. Varshney
Department of Medicine/Infectious Diseases and
Department of Microbiology and Immunology
Albert Einstein College of Medicine
Bronx, NY 10461
Stefanie N. Velgos
Mayo Clinic Arizona
5777 East Mayo Boulevard
Phoenix, AZ 85054
Kevin J. Whaley
Mapp Biopharmaceutical, Inc.
6160 Lusk Blvd. #C105
San Diego, CA 92121
Peter P. Wibroe
Centre for Pharmaceutical Nanotechnology and Nanotoxicology
Department of Pharmacy
Faculty of Health and Medical Sciences
University of Copenhagen
Copenhagen, DK-2100
Denmark
John V. Williams
Department of Pediatrics
School of Medicine
Vanderbilt University
Nashville, TN 37232-2581
xviCONTRIBUTORS

Ian A. Wilson
Department of Molecular Biology and Skaggs Institute for Chemical Biology
The Scripps Research Institute
10550 North Torrey Pines Road
La Jolla, CA 92037
Herren Wu
Department of Antibody Discovery and Protein Engineering
MedImmune, LLC
One MedImmune Way
Gaithersburg, MD 20879
Guocheng Yang
Department of Chemical Engineering
Arizona State University
Tempe, AZ 85287-6006
Larry Zeitlin
Mapp Biopharmaceutical, Inc.
6160 Lusk Blvd. #C105
San Diego, CA 92121
Preface

Antibodies form the principal foundation for modern intervention against in-
fectious diseases. Emil Adolf von Behring was awarded the first Nobel Prize in
Physiology or Medicine in 1901 for his work on serum therapy, especially its appli-
cation against diphtheria, by which he has opened a new road in the domain of med-
ical science and thereby placed in the hands of the physician a victorious weapon
against illness and deaths. Antibodies now provide the focus for understanding
mechanisms of immunity to most infectious diseases, and they play a central role
in passive immunotherapy and active vaccination as mechanisms or correlates of
immunity. For most of the 20th century, immunotherapy was based on passive
transfer of polyclonal hyperimmune animal serum, immune human serum, or
even hyperimmune human serum. Georges J.F. Khler and Csar Milstein re-
ported the generation of monoclonal antibodies in 1979, for which they shared
the 1984 Nobel Prize in Physiology or Medicine for . . .the discovery of the princi-
ple for production of monoclonal antibodies. Since that time, entire fields related
to antibodies for infectious diseases, including antibody gene cloning, engineer-
ing, and expression; antibody libraries; and high-throughput antibody gene rep-
ertoire sequence analysis have extended our capabilities to explore the diversity
of antibody specificity and function with unprecedented depth and breadth.
This book provides a broad survey of many of the most important aspects of
the field of antibodies for infectious diseases. The book begins with a general
introduction, followed by chapters 2 through 5 on general features pertaining
to structure, function, isotype, and the role of complement in antibody function.
Chapters 6 through 10 review contemporary approaches for antibody discovery
using phage and yeast display, plasma cell and memory B cell cloning, human
hybridomas, humanized mice, and computational methods. Chapters 11 through
18 review in depth the biology of antibodies specific for particular pathogens,
including viruses and bacterial toxins, to illustrate the role of antibodies in anti-
microbial immunity with specific targets. These chapters reveal that attempts
to raise effective antibody responses to each of these pathogens faces unique and
pathogen-specific challenges. Chapters 19 to 23 cover major technical advances
pertaining to antibody engineering, repertoire sequencing and analysis, and new
methods for study or therapeutic use of antibodies, including radiotherapy. Fi-
nally, chapters 24 and 25 cover new methods for expression of monoclonal anti-
bodies, in plants or by transfer of antibody genes for in vivo expression in treated
subjects.
Recent literature is exploding with new antibody-related techniques and re-
ports of antimicrobial antibodies with unprecedented potency, breadth of activ-

xvii
xviiiPreface

ity, and therapeutic potential. We hope that this timely compilation of state-of-
the-art reviews of major aspects of this field will be of interest to both antibody
cognoscenti and those new to this exciting field. We thank the authors for their
dedication in producing definitive reviews of the topics at hand.

James E. Crowe, Jr.


Diana Boraschi
Rino Rappuoli
INTRODUCTION
History and Practice:
Antibodies in Infectious Diseases

ADAM HEY1
1
Antibodies in Infectious Diseases aims to inform, update, and inspire students,
teachers, researchers, pharmaceutical developers, and health care professionals
on the status of the development of antibody-based therapies for treating
infectious diseases and the potential for these in times of growing antibiotic
resistance to provide alternative treatment solutions to the currently used
antibiotics and new treatments for infectious diseases where no proper
treatments are available.
This introductory article will provide a historical perspective on the use
of antibody-based therapies, followed by a high-level overview of what
makes antibodies attractive tools for this purpose. This will include the pros
and cons of such therapies compared to the use of antibiotics and the
practical and strategic considerations involved in selecting the best format
and development path for new antibody-based therapies targeting specific
infectious agents. Then, examples of antibody-based therapies in the
development of treatments for infectious diseases will be presented, and
finally a look into the future will summarize the different aspects that will
influence what the future might bring for this type of treatments for
infectious diseases.

1
Preclinical Safety, Biologics, Novartis AG, Basel, Switzerland.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0026-2014

3
4 HEY

HISTORICAL PERSPECTIVE above, serum therapy applied in these early


days (late 19th century and early 20th century)
Antibodies and the use of passive antibody involved preparations of serum from rabbits
therapy in the treatment of infectious dis- and horses immunized with the infectious
eases is the story of a treatment concept agent or in live and/or neutralized versions or
which dates back more than 120 years, to the toxins from these (1). The costs of keeping the
late 19th century, and which originally, by immunized animals and the production and
the use of serum from immunized animals, potency testing of the materials made this a
provided the first effective treatment options relatively expensive treatment. In 1891, data
against severe bacterial infections (1, 2). By from Klemperer (4) showed serum therapy to
immunizing horses with bacterial toxins protect rabbits from Streptococcus pneumoniae
from Clostridium tetani and Corynebacteri- infection and paved the way for this type of
um diphtheriae, Emil A. Von Behring and treatment and for development of similar
Shibasaburo Kitasato (3) generated serum serum-based treatments of streptococcal in-
containing antibodies capable of neutraliz- fections in humans. When treating humans,
ing the effects of the toxins produced by early administration of serum could reduce
these bacteria and successfully provided mortality significantly down to around 5%
treatment for these serious diseases where compared to when administered 4 to 5 days
the pathogenesis is driven by the effects of after onset of symptoms, when serum treat-
the bacterial toxins. For his work on provid- ment was largely without effect. This strongly
ing treatment for diphtheria, Behring re- indicated the need for quick diagnosis and
ceived the Nobel Prize in Physiology or quick treatment to control the infection before
Medicine in 1901. These radical treatment it got out of control. Consequently, in the
results quickly prompted development of absence of a specific diagnosis, mixtures of
multiple additional serum therapies for the serum from immunizations with different
treatment of infectious diseases caused by, serotypes were used to circumvent this need
e.g., Neisseria meningitidis, Haemophilus in- for early treatment without having a serotype-
fluenza, and group A Streptococcus. Since specific diagnosis. The understanding that
serum therapy involved administration of different serotypes existed for pneumococci
large amounts of crude mixtures of animal and that efficient treatment relied on using
proteins including antibodies, they were serotype-specific serum was being built up
associated with side effects in the form of during the 1920s and 1930s through experience
hypersensitivity and serum sickness (2). from extensive clinical trials.
Due to the crude and unpurified nature of By the end of the 1930s serum therapy was
these products, side effects were seen even the standard of care for treatment of pneu-
when administering human serum prepa- mococcal pneumonia. At that time, the
rations. Side effects were observed in up to efficacy and potency of the derived sera
50% of patients and were considered to be were assessed in mice, in the mouse pro-
caused by immune complex formations that tection test by testing survival after a
resulted in symptoms such as rash, itching, concomitant intraperitoneal injection of a
joint pain, fever, and in serious cases hypoten- lethal dose of pneumococci and the serum to
sion and shock. However, due to the lack of be tested. Due to the inherent variation in
alternative options, these treatments were, this test, efficacy and survival in two thirds of
despite their side effects, widely used. Serum the animals was the acceptance criteria, and
was normally administered by intravenous 10 times the lowest dose providing this was
infusion in patients after a test for hypersensi- used for defining a unit of the serum. This
tivity where a small amount of serum was allowed for large batch-to-batch variation,
injected subcutaniously (1). As described and the use of different strains of bacteria for
CHAPTER 1 History and Practice: Antibodies in Infectious Diseases 5

immunization probably explains part of the tridium botulinum, vaccinia virus, echovirus,
missing responses observed (1). In the early and enterovirus. For the most part, these
20th century a pandemic of meningitis in treatments consist of pooled immunoglobulin,
Europe and the United States, with mortality also known as IVIG (intravenous immunoglob-
rates up to 80%, spurred the development of ulin), from several postexposure donors. This
serum therapy treatment options. Although results in both batch-to-batch variation, in the
in the 1930s this became the recommended need for relatively large amounts of serum due
treatment in children assumed to be suffer- to low specificity and to restricted supplies due
ing from meningitis, failure to reduce mor- to reliance on exposed donors.
tality in several meningitis epidemics during However, several challenges have resulted in
that time raised doubts about the general the need for new tools in the treatment and
applicability of serum treatment. In those prevention of infectious diseases. The broad and
days serum therapy often involved quite general use of antibiotics in human and veter-
extensive procedures and infusion of large inary medicine for many years has resulted in
volumes of serum. The following example the development of multi-resistant strains of
clearly illustrates this. Data from Flexner bacteria with limited to no response to existing
and Jobling (5) from treating meningitis in treatments such as methicillin-resistant Staph-
monkeys resulted in the development of sera ylococcus aureus (MRSA), vancomycin-resistant
from immunized horses for treatment in S. aureus, and others. This has resulted in
humans. The treatment protocol included patients needing screening and treatment with
lumbar puncture and withdrawal of more several antibacterial agents and longer treat-
than 30 ml of spinal fluid representing an ment time, causing extra strain on patients and
amount slightly larger than the expected health care providers (6; http://www.cdc.gov/
amount of horse serum to be injected subse- drugresistance/threat-report-2013/index.
quently. This treatment involved such daily html). According to the WHO and CDC more
slow infusions of up to 30 ml of serum until than 25,000 people in European Union
the patients condition improved. This treat- countries and similar numbers in the United
ment was used in outbreaks in New York in States die every year as a result of antibiotic-
1905 and 1906 and did markedly decrease resistant infections. This together with the
mortality. emergence of new pathogens (e.g., severe
After the discovery of penicillin by Fleming acute respiratory syndrome, Middle East respi-
in 1928, and the subsequent introduction of ratory syndrome), the re-emergence/epidemics
antibiotics in the 1930s, serum therapy was of old/known pathogens (e.g., Ebola), and the
largely abandoned over a period of 10 years due difficulties in treating infections in immune-
to the availability of these new, more broadly deficient patients (e.g., HIV patients) has high-
effective and cheaper treatment options, which lighted the need for new solutions. The 2014
also had fewer side effects. Although improve- Ebola epidemic in West Africa (Liberia, Sierra
ments in the purification of antibodies had Leone, Guinea, Nigeria, and Senegal) has fur-
resulted in preparations with better safety and ther highlighted this. No treatment or prophy-
side effect profiles, high manufacturing costs lactic vaccine is available to treat or prevent the
and narrow specificity resulted in antibody spread of Ebola infections, which have an
therapy being mostly restricted to a smaller average mortality of >50%. Local health author-
number of selected treatments for snake ities in the affected countries are struggling to
venoms, bacterial toxins, and some viral infec- contain and handle the disease, which is
tions (1, 2). Currently, antibody administration threatening to go out of control and spread
is used for treatment and prevention of more widely. Various products, mainly antibody
hepatitis B virus, rabies virus, respiratory cocktails from previously recovered patients,
syncytial virus (RSV), Clostridium tetani, Clos- are being used despite a lack of clinical data on
6 HEY

their safety and efficacy, and those are the only Antibodies. Antibody-based programs cur-
sporadically available treatment options and rently at different stages of development
only in small amounts and for a few patients. include investigations into the potential use of
Ebola is an example of a disease which single antibody preparations, combinations of
normally affects only a small number of antibodies (to avoid survival of escape mu-
individuals and which normally burns out tants), fragments of antibodies, and antibodies
when disease outbreaks are contained. There- carrying radioactive isotopes or cytotoxic drugs
fore, given the small number of potential or antibody-like frameworks (e.g., fibronectin)
patients affected by previous Ebola infections either alone, as a first-line treatment, or as an
to date, there was no incentive for big pharma adjunct to existing treatments. This multitude
companies to do research and development of of possible formats, the ability to raise anti-
drugs for Ebola. With the increasing number of bodies to almost any target and the ability to
infected (13,567) and a death toll of 4,951 (7) and engineer both size, effector functions, and half-
the lack of the ability to contain the epidemic, it life are now considered by many to provide
will be interesting to follow the aftermath of very valuable tools for designing specific
this outbreak and see whether there will be antibody-based treatments to eradicate specific
requests for new ways to ensure that vaccines targeted infectious agents. However, although
and treatment options are available for Ebola several antibody-based therapies have been
and similar high-mortality and potential bio- approved for oncology and anti-inflammatory
warfare infections that have no available indications (see Table 1), only one monoclonal
treatments or procedures to for mass-produc- antibody (mAb) is approved against an infec-
tion upon the first signs/reports of active tious disease agentSynagis (palivizumab,
infections. Although both the CDC and the MedImmune) for the prevention and treat-
WHO have special programs focusing on these ment of respiratory syncytical virus (RSV) in
types of infections, the Ebola outbreak in 2014 high-risk children. Economic obstacles such as
clearly shows that more financial support for the high cost of mAb therapies and relatively
research and development of new diagnostics small markets have resulted in less interest
and treatments is needed. This is one example from pharmaceutical companies in developing
where antibody-based treatments would have these. Also highlighting the challenges in
the potential to play a major role. To put this in developing these types of antibody-based
the right perspective, one should, however, not treatments are the difficulties encountered in
forget that other infectious diseases such as developing a follow-up higher-affinity candi-
tuberculosis, influenza, and malaria kill hun- date to Synagis. Poor translatability of data
dreds of thousands each year. There is there- obtained in the available animal disease mod-
fore plenty of room for improvement in els for RSV in cotton rats has resulted in non-
developing treatments for these diseases as approval due to inferiority relative to Synagis.
well, but the high attention drawn to, e.g., the In other viral diseases such as cytomegalovirus
Ebola outbreak creates a special niche and (CMV) the high species specificity of the
opportunity where antibody treatments could strains of CMV makes it impossible to test
gain extraordinary development funding and human antibody therapies in vivo. Therefore,
support and prove their value and treatment any real data on the efficacy of treatments for
potential. CMV will not be obtained until testing in
That antibody-based therapies could take clinical trials. As described later in this article
this role is supported by the revolution in several new antibody-based therapies for in-
technologies for the development, selection, fectious diseases are in development, and
generation, and purification of fully human hopefully this will gradually open the field
antibodies described in more detail later in the and result in better treatment options and
section Methods and Platforms for Generating outcomes for patients.
CHAPTER 1 History and Practice: Antibodies in Infectious Diseases 7

TABLE 1 Approved and pending antibody-based therapies


Name: antibody Target: antibody type Indication Company Approval date
a
OKT3 : Muronomab-CD3 CD3: murine, IgG2a Autoimmune Johnson & 1986 (U.S.)
Johnson
ReoPro: abciximab PIIb/IIIa: chimeric, IgG1, Homeostasis Johnson & 1984 (U.S.)
Fab Johnson
Rituxan: rituximab CD20: chimeric, IgG1 Cancer Genentech 1997 (U.S.) 1998 (E.U.)
Zenapaxa: daclizumab CD25: humanized, IgG1 Autoimmune Roche 1997 (U.S.) 1999 (E.U.)
Simulect: basiliximab CD25: chimeric, IgG1 Autoimmune Novartis 1998 (U.S., E.U.)
Synagis: palivizumab RSV: humanized, IgG1 Infections MedImmune 1998 (U.S.) 1999 (E.U.)
Remicade: iniximab TNF: chimeric, IgG1 Autoimmune Johnson & 1998 (U.S.) 1999 (E.U.)
Johnson
Herceptin: trastuzumab HER2: humanized, IgG1 Cancer Genentech/ 1998 (U.S.) 2000 (E.U.)
Roche
Mylotarga: gemtuzumab CD33: humanized, IgG4, Cancer Wyeth/Pzer 2000 (U.S.)
ozogamicin immunotoxin
Campath: alemtuzumab CD52: humanized, IgG1 Cancer Genzyme 2001 (U.S.) 2001 (E.U.)
Zevalin: ibritumomab CD20: murine, IgG1, Cancer Biogen Idec 2002 (U.S.) 2004 (E.U.)
tiuxetan radiolabeled (yttrium 90)
Humira: adalimumab TNF: human, IgG1 Autoimmune Abbott 2002 (U.S.) 2003 (E.U.)
Xolair: omalizumab IgE: humanized, IgG1 Autoimmune Genentech/ 2003 (U.S.)
Roche
Bexxar: tositumomab-I-131 CD20: murine, IgG2a, Cancer Corixa/GSK 2003 (U.S.)
radiolabeled (iodine
131)
Raptivaa: falizumab CD11a: humanized, IgG1 Autoimmune Genentech/ 2003 (U.S.) 2004 (E.U.)
Roche
Erbitux: cetuximab EGFR: chimeric, IgG1 Cancer Imclone/Lilly 2004 (U.S.) 2004 (E.U.)
Avastin: bevacizumab VEGF: humanized, IgG1 Cancer Genentech/ 2004 (U.S.) 2005 (E.U.)
Roche
Tysabri: natalizumab 4-Intergrin: humanized, Autoimmune Biogen Idec 2004 (U.S.)
IgG4
Actemra: tocilizumab Anti-IL-6R: humanized, Autoimmune Chugai/ 2005 (Japan) 2010 (U.S.)
IgG1 Roche
Vectibix: panitumumab EGFR: human, IgG2 Cancer Amgen 2006 (U.S.)
Lucentis: ranibizumab VEGF: humanized IgG1 Macular Genentech/ 2006 (U.S.)
Fab degeneration Roche
Soliris: eculizumab C5: humanized IgG2/4 Blood disorders Alexion 2007 (U.S.)
Cimzia: certolizumab pegol TNF: humanized, Autoimmune UCB 2008 (US)
pegylated Fab
Simponi: golimumab TNF: human IgG1 Autoimmune Johnson & 2009 (U.S., E.U., Canada)
Johnson
Ilaris: canakinumab IL1b: human IgG1 Infalmmatory Novartis 2009 (U.S., E.U.)
Stelara: ustekinumab IL-12/23: human IgG1 Autoimmune Johnson & 2008 (E.U.) 2009 (U.S.)
Johnson
Arzerra: ofatumumab CD20: human IgG1 Cancer Genmab 2009 (E.U.)
Prolia: denosumab RANK ligand: human Bone loss Amgen 2010 (U.S.)
IgG2
Numax: motavizumab RSV: humanized IgG1 Anti-infective MedImmune Pending
ABthrax: Raxibacumab B. anthrasis PA: human Anti-infection GSK 2012 (U.S.)
IgG1
Benlysta: belimumab BLyS: human IgG1 Autoimmune HGS 2011 (U.S.)
Yervoy: ipilimumab CTLA-4: human IgG1 Cancer BMS 2011 (U.S.)
Adcetris: brentuximab CD30: chimeric, IgG1, Cancer Seattle 2011 (U.S.)
vedotin drug- conjugate Genetics
(Continued on next page)
8 HEY

TABLE 1 Approved and pending antibody-based therapies (Continued)


Name: antibody Target: antibody type Indication Company Approval date

Perjeta: pertuzumab Her2: humanized, IgG1 Cancer Genentech/ 2012 (U.S.)


Roche
Kadcyla: ado-trastuzumab Her2: humanized, IgG1, Cancer Genentech/ 2013 (U.S.)
emtansine Drug-conjugate Roche
Raxibacumab Anti-B anthrasis PA: Anthrax Not 2012
human IgG1 infection approved
Entyvio: vedolizumab Integrin 47: Crohns disease, Takeda 2014 (U.S.)
humanized IgG1 ulcerative colitis
Cyramza: ramuciruumab Anti-VEGFR2: Gastric cancer Lilly 2014 (U.S.)
Human IgG1
Gazyva: obinutuzumab Anti-CD20: humanized Cancer/CLLb Genentech/ 2014 (U.S.)
IgG1 glucoengineered Roche
Sylvant: situximab Anti IL-6: chimeric IgG1 Castleman Janssen 2014 (U.S.), 2014 (E.U.)
disease
Cosentyx: sekukinumab Anti IL-17a: human IgG1 Psoriasis Novartis 2015 (U.S.), 2015 (E.U.)
Nivolumab Anti PD-1: human IgG4 Melanoma BMS Not approved
Keytruda: pembrolizuumab Anti-PD-1: humanized Melanoma Merck 2014 (U.S.)
IgG4
a
Antibodies approved but later withdrawn.
b
CLL, chronic lymphocytic leukemia.

INTRODUCTION TO ANTIBODIES Antibodies consist of two pairs of heavy and


light chains which, as described above, are held
Antibodies, also called immunoglobulins based together in a Y-shaped arrangement. Each of
on their combined structure and function in the heavy and light chains in these pairs is
immune responses, are produced by B cells of separated into constant and variable chains.
the immune system. They are part of the The upper arms of the Y each contain a variable
adaptive immune response and are specially and a constant section of the light and the heavy
designed for neutralizing and eliminating the chain, where the upper variable parts of the
infectious agents and toxins produced by these. heavy and light chains contain the antigen-
Antibodies are present in blood, plasma, and binding site, and the constant parts are
extracellular fluids, and since these fluids were connected via disulfide bonds. The lower part
formerly known as humors, they were said to of the Y, called the constant part, consists of two
be part of the humoral immune response. or three constant segments (immunoglobulin
Antibodies are Y-shaped structures consisting domains) from each of the two heavy chains
of two main parts: the upper arms of the Y, interacting and also linked via disulfide bonds.
which contain two identical variable-region The sections of the heavy chains connecting the
antigen binding sites, and the lower region, constant part of the upper arms to the constant
called the constant region, which is responsible parts of the constant region contain special
for the initiation of effector functions that lead hinge regions that provide flexibility to the
to the removal and destruction of the pathogen different sections of the antibody to bind to
or cells harboring the pathogen (see Fig. 1). The antigens and effector cells. The constant re-
antigen-binding sites on an antibody can by gions of the heavy chain also contain attached
themselves bind to and neutralize bacterial oligosaccharide moieties which provide func-
toxins and viruses, thereby preventing them tional specialization to the antibodies (8).
from binding to their target cells or receptors The variable domains of the heavy and light
causing toxic effects or spread of the infection. chains form the antigen-binding sites and
CHAPTER 1 History and Practice: Antibodies in Infectious Diseases 9

FIGURE 1 (Left panel) Model of antibody structure exemplied by IgG. On top the antigen-binding sites
in orange each contain one variable light and variable heavy domain with the three complementarity
determining regions (CDRs) that are responsible for the specic binding of the antibody to its target. For
each arm of the antibody, an additional set of variable heavy and light domains, together with the CDR-
containing domains, represent the two fragment antigen binding (Fab) regions. The two Fabs are held
together via two disulde bridges. Below the Fabs is the Fc region, which contains four constant heavy
domains. On the upper pair of these domains are binding sites for oligosaccharides, which have major
importance for the ability of the antibody Fc part to trigger effector functions when the Fc portion is
bound to Fc gamma receptors on natural killer cells, neutrophil granulocytes, monocytes/macrophages,
dendritic cells, and B cells. (Right panel) Examples of some of the antibody-derived alternative formats
used to exploit the specic features of the CDRs, the Fabs, and the Fc parts of the antibodies. ScFv: The
single chain fragment variable consists of the variable domains of the heavy and light chains held
together by a exible linker. This can also be used as a carrier of a cytotoxic drug in a so-called antibody
drug complex (ADC) where the specicity of the ScFv is used to target the cytotoxic drug to, e.g., a
tumor. Bite (bi-specic T cell engager): Fusion proteins consisting of two ScFvs, one directed against the
target on a tumor cell and the other against the T cell receptor (CD3). Diabody: ScFv dimers where short
linker peptides (ve amino acids) ensure dimerization, and not folding, of the ScFvs. Fab and F(ab)2
fragments: Single Fab fragments or fragments containing two Fabs linked via disulde bridges. This is
used where effector functions related to the Fc part of the antibody are unwanted and where a smaller
size is desired to obtain better tissue penetration in, e.g., tumors. Due to the lack of the FcRn binding via
the Fc part, Fab and F(ab)2 fragments have much shorter half-lives (hours or days) than full-size
antibodies (weeks). These can also be used as carriers of cytotoxic payloads or cytotoxic radioactive
isotopes and for the F(ab)2 fragments can be constructed as bi-specics which can cross-link immune
cells and target cells. Fc fusion protein: Fusion protein containing the Fc domain of an immunoglobulin
bound to a peptide. The peptide can be a ligand for a specic receptor on a target cell or a blocking
peptide for a soluble ligand. The Fc part provides a longer half-life to the construct and the potential to
bind to and engage effector functions in the killing of, e.g., tumor cells or infected cells. ADCs/RIAs and
bi-specics: Full-size IgG antibodies carrying either a cytotoxic chemical or radioactive payload, which
may also carry different CDRs, enabling cross-linking of effector and target cells for increased killing.
doi:10.1128/microbiolspec.AID-0026-2014.f1
10 HEY

contain special hyper-variable segments called cells to kill infected cells via binding to Fc
complementarity-determining regions (CDRs), receptors on these cells and antibody-depen-
which allow the B cells, via genetic recom- dent cellular cytotoxicity; and (iv) neutraliza-
bination, to generate antibodies to all the tion of bacterial toxins and blocking of binding
different antigens specific to amino acid se- and uptake of bacteria or viruses to target cells
quences or three-dimensional structural motifs (see Fig. 2) (9).
(polysaccharides, DNA, and RNA) found on The Fc part of IgG antibodies also contains a
pathogens. region that interacts with the neonatal Fc
Based on the structure of their heavy chain receptor (FcRn) expressed on most immuno-
constant regions, antibodies, or immuno- competent cells and in many other cells such as
globulins, are separated into five classes endothelial cells in kidney, liver, placenta, lung,
IgM, IgG, IgE, IgD, and IgA. Differences in and breast (10), which protects antibodies from
the sequences of the constant regions provide degradation by phagocytotic/exocytotic cy-
distinct characteristics to these different clas- cling into FcRn-expressing cells, thereby pro-
ses. These characteristics include the number longing their half-life. This results in half-lives
of heavy chain constant segments, the number of 20 to 21 days for most IgG1, 2, and 4
and location of interconnecting disulfide antibodies, whereas half-lives are 10, 6, and 2
bridges, the length of the hinge region, and days, respectively, for IgM, IgA, and IgE. The
the number and location of oligosaccharide highly specific binding sites on the variable
moieties. For IgG, four heavy chain versions regions of the upper arms of the antibody allow
exist, creating IgG1, 2, 3, and 4 isotypes, each for several important features of antibodies
with different characteristics in their serum binding to and neutralization of, e.g., bacterial
half-lives and ability to trigger different effec- toxins, binding to pathogens and blocking their
tor functions. Because of the combined ad- binding to or interaction with target cells (e.g.,
vantage from ability to trigger effector cyncytia formation of CMV-infected cells); or
mechanisms, long [21 days] half-life, their blocking of binding and interactions of infec-
ability to be transported over the placenta, tious agents with target cells via blocking
and their stability in the production process, receptors on the target cells.
antibodies of the IgG class and of the IgG1
isotype are preferred as the basis for new
antibody-based therapies. The effector func- METHODS AND PLATFORMS FOR
tions are triggered by the Fc part of an antibody GENERATING ANTIBODIES
when the variable parts of the antibody are
bound directly to an infectious agent or to As described above, the early use of passive
proteins from an infectious agent expressed on antibody transfer as a therapy in infectious
an infected cell. This binding leaves the Fc part diseases involved the use of immune serum
free to interact with Fc receptors on phagocytic from immunized sheep and horses. These pre-
cells and neutrophils, eosinophils, and natural parations were crude and contained a broad
killer cells capable of inducing phagocytosis or mixture of neutralizing and non-neutralizing
lysis of infectious agents or infected cells. antibodies and both the foreign nature of the
These effector functions include (i) the ability antibodies and the numerous host serum
to activate the complement system for lysis proteins. This resulted in less specific and
(complement-dependent cellular cytotoxicity); thus less efficient therapies due to a high
(ii) triggering of uptake and destruction by variability in efficacy between batches derived
phagocytic cells such as macrophages and from different animals immunized with dif-
neutrophils via interaction with Fc receptors ferent strains of bacteria in different labor-
on these cells; (iii) activation of macrophages, atories and resulted in multiple unwanted
eosinophils, neutrophils, and natural killer side effects related to hypersensitivity and
CHAPTER 1 History and Practice: Antibodies in Infectious Diseases 11

FIGURE 2 Effector functions of antibodies. (a) Antibodies bind to pathogen-derived or endogenous


antigens expressed on the surface of an infected cell, which triggers binding to Fc receptors on natural
killer cells and lysis of the infected cell by antibody-dependent cellular cytotoxicity. (b) Antibodies bind
to pathogen-derived or endogenous antigens expressed on the surface of infected cells, which triggers
activation of complement through binding of complement factor C1q. (c) Neutralization. Top: Bacterial
toxin neutralized by bound antigen. Bottom: Antibody bound to either receptor for the virus or to the
virus itself, which blocks virus binding and entry into the cell. (d) Antibody bound to viral surface
proteins binds to Fc receptors on phagocytic cells, e.g., macrophages, and triggers endocytosis and
destruction of virus in endolysosome. doi:10.1128/microbiolspec.AID-0026-2014.f2

anaphylactic reactions to foreign proteins. Rituxan [rituximab, Genentech]). Next stage


These days, antibodies being developed for was then the humanized antibodies where
treatment of diseases in humans are highly the only remaining murine part of the anti-
purified and are mostly fully human mono- body is the antigen-binding CDR (e.g., Xolair
clonal antibodies. The term monoclonal anti- [omalizumab, Novartis/Genentech]). Both the
body refers to that cell cultures used for first chimeric and humanized antibodies were
generating these antibodies each originates approved in the late 1990s. Then finally 16
from a single cell and thus produces only years later, the first fully human antibody
one specific antibody. Development of fully Humira (adalimumab, Abbott) was approved
human monoclonal antibodies happened in in 2002. Since the time of the crude and low-
stages over a time span of more than 15 years. scale manufacturing of the original antibody
First stage in this process was generation of serum products, much has changed, and
monoclonal murine IgG antibodies (e.g., OKT3 therapeutic antibodies are now produced by
[muronomab-CD3, Johnson and Johnson] ap- fermentation in 5,000 liter scale using highly
proved in 1986). Next stage was generation of controlled and documented yeast or mamma-
so called chimeric antibodies consisting of lian cell lines that are genetically engineered to
human heavy and light chain constant sec- express a single specific antibody. This has
tions but with murine variable sections (e.g., resulted in a pure and highly specific antibody
12 HEY

product, and thanks to great improvements in tration of cells in the subsequent seeding
cell line design and fermentation efficiency, cultures are less than one. This is done to
monoclonal antibody therapeutics can now be ensure that each cell culture is derived from one
produced at more reasonable, although still cell only (monoclonal) and that only identical
high, costs. antibodies from a single clone is harvested from
The price of antibody-based therapies is, each of the cell cultures.
however, still much higher than chemically An essential technological step on the way to
synthesized antibiotics. Although they do the current antibody products was the method
provide very welcome and needed treatment to produce monoclonal antibodies by immortal-
options, the cost of antibody-based treat- izing B cells, which is the basis for the
ments approved for cancer and inflammatory hybridoma technology (13). This allowed the
diseases is creating major economic chal- production of large amounts of homogenous
lenges for health care systems and health antibodies with defined specificity and a single
care providers around the world. Targeted Ig class and isotype (14). This was quickly
immune therapy in, e.g., colorectal cancer adapted for clinical use, and in the 1980s the
with 8 weeks of treatment with a monoclonal OKT3 anti-CD3 monoclonal murine IgG2 anti-
antibody like Erbitux (cetuximab, Imclone/ body was the first one in the class approved for
Lilly) cost up to $31,790 in the 1990s. In prevention of organ transplant rejection (14).
comparison, $63 for an 8-week treatment The ability to generate monoclonal antibodies in
with fluorouracil, which was the standard unlimited amounts and against almost any target
treatment until the mid-1990s, puts the cost protein provided extremely valuable tools for
of antibody treatments in perspective. detecting, locating, inhibiting, and blocking
Highly specific antibodies monoclonal anti- specific markers and pathways in general
bodies (each produced by culture of cells all biological and medical research and in setting
derived from the same single cell), can be up analyses for numerous markers via enzyme-
selected for and generated from immunized linked immunosorbent assay or immunohisto-
humans or animals in several different ways: (i) chemistry. It also played a major role in basic
phage display of a human variable light segment research into the mechanisms of antibody
library from several donors and B cells, followed action.
by cycles of panning against target antigen,
selection of phages with the desired target STRATEGIC CONSIDERATIONS IN
specificity, and subsequent cloning and expres- DEVELOPING ANTIBODY-BASED
sion in a cell line for expression; (ii) the use of THERAPIES FOR INFECTIOUS DISEASES
transgenic mice carrying the genes for human
IgG and immortalization of the mouse B cells by As mentioned above, the past 10 to 15 years saw
fusing them with myeloma cells (the hybridoma a revolution in development and approvals of
technology); and (iii) isolation of memory B monoclonal antibody-based therapies for in-
cells or activated memory B cells (plasmablasts) flammatory and neoplastic diseases. This rev-
from patients that have been or are exposed to olution, however, did not include treatments
the infectious agent. After isolation the memory for infectious diseases. As can be seen in Table
B cells are then first screened for reaction to the 1, more than 40 antibody-based treatments
relevant antigen and positive cells are then have been approved or are under final evalu-
immortalized by transformation with Epstein ation. Although many products for infectious
Barr virus transformation in the presence of a diseases are in different stages of development,
oligodeoxynucleotide (CpG) and irradiated pe- still only one monoclonal antibody-based prod-
ripheral blood mononuclear cells (11, 12). uct, Synagis, is currently approved for use in
Finally the cell cultures are subjected to several infectious diseases. The main reason for this is
steps of limiting dilution where the concen- the availability of the much cheaper, easier to
CHAPTER 1 History and Practice: Antibodies in Infectious Diseases 13

administer, and more broadly acting anti- therapy in the United Kingdom not working
microbials or antibiotics. However, the in- in one in seven patients (15). In the United
creasing development of resistant strains of States this situation has resulted in multiple
bacteria (e.g., MRSA) and viruses has opened cases with the need for dosing up to 10 different
the door for a re-emergence of antibody-based antibiotics before proper control of infection
therapies. Several features of monoclonal anti- can be obtained. Table 2 lists the pros and cons
bodies are, however, working against them of antibody-based therapies and antibiotics.
when comparing with antimicrobial therapies. To make antibody therapeutics more
First, and most importantly, is the very high efficacious and more convenient for patients
cost of production already mentioned above. In and treating physicians, several approaches
addition, since antibodies are proteins, they have been and are being taken.
need to be treated carefully, kept refrigerated,
and they are administered by intravenous or
Strategy To Avoid Escape Mutants
subcutaneous injection. In contrast, antibiotics
usually come in the form of tablets that can be Although antibody treatments do not induce
taken orally and can be kept in a bag or in a resistance in nontarget bacteria or viruses, both
closet at room temperature. Antibiotics nor- bacteria and viruses have the ability to escape
mally target general mechanisms, e.g., cell wall the host immune system and specific antibody
formation in bacteria, and can act against, e.g., a treatments via mutations that change their
broad spectrum of bacteria, whereas antibodies surface proteins or structure, creating so-called
are very specific to a single virus, bacteria, or escape mutants that are no longer neutralized
bacterial subtype, and a clear diagnosis should by the specific antibody. The best way to
be made before initiating treatment with a circumvent this is by using a combination of
monoclonal antibody. This highlights the need antibodies directed at different viral targets. The
for improving or developing diagnostics in use of cocktails of two or more antibodies was
parallel with developing antibody therapies. shown to provide synergism or additive effects
The specificity also has an economical angle in neutralizing, e.g., hepatitis B virus (HBV) and
since it results in a smaller market compared to RSV infections (16). Combinations of antibodies
broad-spectrum antibiotics, making antibody- have also been used in HIV, targeting GP41 and
based therapies less attractive for pharmaceu- GP120 viral proteins (17), and rabies (18) and are
tical companies to develop. part of the strategy in most pharmaceutical
The specificity is, however, also a strength companies that are developing antiviral anti-
in the sense that the antibody works only on a bodies. Therefore, part of the early preclinical
specific infectious agent, and although muta- development of such antibody combinations
tions in that agent can render the antibody consists of serial passage of virally infected cells
ineffective, this does not affect other similar (>20 generations) to ensure continued efficacy
agents and thus does not cause the spread of and the absence of escape mutants. This is
resistance. The specificity also results in very combined with testing against known patient
low off-target binding and therefore very few isolates, when available. Cocktails of antibodies
side effects, including the gastrointestinal ef- would also be an interesting approach to target
fects often observed with antibiotics due to groups of infectious agents often seen in parallel
broad effects on bacterial flora in the gut. The in, e.g., burn wounds.
balance between antibody therapies and anti-
biotics with respect to costs and ease of use in
Antibody Engineering
the clinic should, however, be seen in a broad
and more realistic context, including the Another strategy to increase efficacy, increase
current situation around the development of the ability of the treatment to reach the
resistance, which has resulted in antibiotic intended target, and avoid unwanted side
14 HEY

TABLE 2 Pros and cons of antibody based therapies related to serum therapy and antibiotics
Parameter Serum therapy Antibiotics mAbs Comments

Source Animals Humans Chemical synthesis Tissue culture


Fermentation Bioreactor
Fermentation
Lot variation High Low Low
Specicity Narrow Broad Narrow Narrow specicity
prevents development of
resistance in non-target
species but results in the
requirement for a specic
diagnosis before treatment.
Restricts treatment to single
species.
Toxicity High Low Low Original crude antibodies
from animals had poor
tolerability, but current
human- and animal-derived
immunoglobulins have been
proven safe
Ease of Difcult (intravenous Easy/difcult Easy Many antibiotics can be
administration [i.v.], intramuscular (oral/i.v.) (i.v., i.m., s.c.) administered orally,
[i.m.], subcutaneous whereas serum or mAbs
[s.c.]) are given by i.v or s.c
injection
Pharmacokinetics Variable Consistent Consistent
Cost/dose High (in the Very high (in the Low (in the
hundreds) thousands) single digits)

effects resulting from the killing of nontarget lular cytotoxicity (22). This is achieved by
cells is antibody engineeringmore precisely, manufacturing the antibodies in cell lines
genetic manipulation of the Fc domain (mainly lacking the enzyme fucosyl transferase, which
in the CH2 domain) or changes to the gluco- renders them unable to add fucose to the
sylation pattern of the N-linked oligosaccha- oligosaccharide moieties (22).
ride moieties attached at antibody N297 in the Similarly, ways to reduce or ablate the
Fc part of the heavy chain. For generating ability of antibodies to trigger effector func-
antibodies with enhanced effector functions, tions have been described and are being used
different mutations have been identified that broadly in cases where the aim is to block
have increased affinity to the FcgIIIa receptor specific membrane-bound receptors/targets
and a significant enhanced cellular cytotoxicity and where killing of the cell harboring the
(S239D/A330L/I332E, also known as 3M [19, target is not desired. Again, mutations in the Fc
20], F243L [21], and G236A). These antibodies part, e.g., the mutations L234A and L235A, also
either directly or indirectly enhance binding of called the LALA mutation, greatly reduce but
Fc receptors and thus significantly enhance do not completely remove effector functions by
cellular cytotoxicity. Enhanced effector func- removing amino acids important for the C1q
tion can also be achieved by modulating the factor of complement (23, 24). Modulation of
oligosaccharide moieties. Removal of fucose the glucosylation pattern, in this case creating
from the A297 linked oligosaccharide moietites, completely aglucosylated antibodies, has also
which creates so-called afucosylated Fc do- been shown to remove the ability to properly
mains, has been shown to greatly increase the bind Fc receptors on effector cells and trigger
potency for inducing antibody-dependent cel- effector functions. One alternative approach
CHAPTER 1 History and Practice: Antibodies in Infectious Diseases 15

used especially when developing immuno- treatments. Difficulties in handling and dispos-
modulatory agonistic antibodies is the use of ing of such products have also contributed to
antibodies of the IgG4 isotype, which does not this.
trigger effector functions. Finally, mutations in With a similar approach but with antibodies
the Fc part that increase the affinity to the FcRn aimed at delivering cytotoxic drugs via anti-
receptor have also been used to create anti- bodies also known as antibody-drug complexes
bodies with an increased half-life. Introduction (ADCs) there is currently great enthusiasm and
of three mutations in the Fc domain (M252Y, activity, especially in oncology. The first anti-
S254T, and T2556E, also called the YTE) has body of this kind, Myoltarg (gemtuzumab
been shown to provide a half-life extension of ozogamicin), was approved by the FDA in
3- to 4-fold (25). From a convenience point of 2000 but was later withdrawn due to major
view, a long half-life is obviously attractive, but toxicities in patients, caused by instability and
it can be a down-side in the case of severe heterogenicity. Two other ADC antibodies have
adverse effects due to the long duration of been approved for the treatment of cancer:
action. Adcetris (brentuximab vedotin) and Kadcyla
(ado-trastuzumab emtansine), which were ap-
proved in 2011 and 2013, respectively. The ADC
Creating Magic Bullets
field is expanding, and many products are in
In oncology, to enhance the efficacy of thera- development in cancer and inflammatory
peutic antibodies, the tactic of using the diseases, but has not resulted in any approvals
specific binding capacity of the antibody to for products in infectious diseases. A major
deliver a cargo in the form of a cell toxin or a challenge in this approach is the selection of the
radioactive isotope to kill tumor cells has long right linker to create a stable complex, as well as
been recognized. The first treatments using ensuring minimal off-target exposure and, most
radiolabeled antibodies were the anti-CD20 importantly, achieving internalization and re-
antibodies Zevalin (ibritumomab tioextan; lease in the proper compartment of the target
yttrium 90 labeled) and Bexxar (tositumumab; cells. The antibody CDRs should ensure spe-
iodine 131 labeled), which were approved in the cific binding and delivery of the cytotoxic drugs,
United States in 2002 and 2003, respectively. but in animal studies using a specific ADC and
These demonstrated the potential for develop- an ADC without target but labeled with same
ing such antibodies. However, they are not toxin, the same broad pattern of toxicity was
widely used in cancer therapy. Successful observed with both products. This indicates
generation of labeled antibodies against fungal that the desired specific targeting to mainly
infections with Cryptococcus neoformans and affect target cells or tissues is currently not
Histoplasma capsulatum and bacterial infec- achievable. So far, this concept is used only in
tions with S. pneumoniae was reported by cancer treatment; for any successful develop-
Saylor et al. (26), and efficacy was shown in ment of such treatments in infectious diseases, a
vitro and in animal models. However, clinical critical case-by-case risk/benefit evaluation
efficacy still needs to be demonstrated. One combined with considerations of availability
opportunity with this approach is that these of other effective drugs will be needed.
antibodies can be directed at infected cells and
not the pathogen itself, killing the infected cells
Antibody Formats
and thereby potentially removing reservoirs of
infected cells (26). The challenges in selecting In the era of antibody-based therapies, full-size
the right isotopes, achieving distribution and antibodies were and are still the main format.
uptake into tumors and tissues, and minimizing There is, however, an increase in companies
exposure of non-target tissues have slowed the offering/researching into new variants of anti-
pace of development of these types of antibody bodies or new combinations of antibody-de-
16 HEY

rived structures. Examples of these can be seen becomes obvious that the vast majority of
in Fig. 1 and include both bispecific full-size these focus on viral and some on bacterio-
antibodies with antigen-binding sites with logical infections. Around 70% of these
different targets, Fab or F(ab)2 fragments, focus on five main pathogens: hepatitis C
single-chain variable fragments (ScFv), pairs virus (HCV), HIV, Bacillus anthracis, Esch-
of ScFvs linked in different ways (diabodies and erichia coli, and S. aureus. The following
bites), fusion-proteins containing, e.g., a recep- section describes examples of projects tar-
tor fused to an antibody Fc part, and many geting infections with these and a few
others not shown. Much creativity has been additional treatments divided into anti-
demonstrated in this field of research, and every bacterials, antivirals, and antifungals.
imaginable variation and combination of anti-
body structures is being created and tested. This
Antibacterials
is done to test how these different formats and
the attributes of the different parts of antibodies Due to their ability to cause a multitude of
potentially can overcome the challenges facing serious infections and due to their high propen-
the development of antibody-based therapies. sity for developing resistance, MRSA infections
These challenges include the high costs of are a major problem in hospitals and in local
production and finding optimal combinations community settings. However, despite several
of size, stability, half-life, efficacy, and very clinical trials with antibodies targeting the
importantly, safety. These new formats also clumping factor A, e.g., Aurexis (tefibazumab,
introduces new structures foreign to the im- Inhibitex) (27) and Aurograb (Novartis), a
mune system and thus immunogenicity is likely single-chain variable fragment (ScFv) targeting
going to be a major factor in these developments an S. aureus surface protease has failed to show
with the potential risk of loss of efficacy, loss of efficacy of any of these treatments (28). Some
exposure, potential hypersensitivity reactions, hope has arisen from the work on pagibaximab
and potential cross-reactivity to endogenous (Biosynexus), which is an antibody for the
receptors or immunoglobulins. As described prevention of staphylococcal sepsis in prema-
above, a lot of work was put into reducing ture infants with low birth weight. Clinical
immunogenicity and immune reactions to the phase I and II trials have shown good safety and
animal-derived or chimeric antibodies by turn- no cases of treatment related adverse events
ing them into more and more fully human when doses of 90 mg/kg were given. A phase III
antibodies. It is therefore a paradox that with study is ongoing (29).
the introduction of these new formats (e.g., new Research into mAbs targeting the adhesins
scaffolds with fibrinogen carrying antigen- SA I and II from Streptococcocus mutans, which
binding sites), the risk of immunogenicity and aims at recolonizing the bacterial flora in the
immune reactions is being reintroduced. Time oral cavity to prevent caries, did not show the
will show whether the safety and efficacy of expected effects in the first human trials, and
these new formats will actually support their further development work was stopped (30).
registration and use in patients with infectious Some optimism has been generated in
diseases. finding an antibody-based treatment of Pseu-
domonas aeruginosa. Two antibody-based treat-
ments are in development for the prevention
EXAMPLES OF ANTIBODIES THAT ARE and treatment of infections with Pseudomonas
OR WERE IN DEVELOPMENT FOR in ventilated patients: a full-size antibody, pano-
INFECTIOUS DISEASES bacumab (Kenta Biotech), directed against
LPS 011, and a pegylated Fab product target-
When searching for antibody-based treat- ing the type 3 secretion system. Both of these
ments in different stages of development, it have shown good tolerability and efficacy in
CHAPTER 1 History and Practice: Antibodies in Infectious Diseases 17

patients, and phase II studies are ongoing body treatment for infectious diseases. An
(31, 32). affinity maturation and half-life extended
B. anthracis causes highly lethal pulmonary third-generation monoclonal, Numax (mota-
infections and is recognized as a potential vizumab, AstraZeneca/MedImmune), is cur-
biological warfare weapon. Four antibodies, rently in clinical development but due to safety
raxibacumab (ABthrax, Human Genome issues and an inability to demonstrate superi-
Sciences), Valortim (PharmAthene/Medarex), ority has so far not been approved by the FDA.
Thravixa (Emergent Biosolutions), and Anthim This is an interesting example of a case where a
(Elusys Therapeutics), are human antibodies back-up for antibody, despite higher affinity
directed against the protective antigen which and potency, fails to show improved efficacy in
interacts with the toxins lethal factor and clinical trials (35).
edema factor to facilitate their toxic action. All A number of antibodies against HCV are in
are undergoing clinical trials for tolerability, but development. One of these (MBL-HCV-1) is
raxibacumab was approved in 2012 for treat- directed against an envelope protein on the
ment of inhalation anthrax. It was, however, virus (36). The others are directed against host
not a traditional approval based on phase II and cell proteins, and one of these, bavituximab
III clinical trials showing efficacy but was the (Peregrine Pharmaceuticals) (37), is directed
first treatment approved under the so-called against phosphatidylserine, which normally is
animal rule, where the efficacy, for good expressed only on the inner surface of the cell
reasons, is approved based on animal studies. membrane but which in cancer- and virus-
For treatment of infections with Clostridium infected cells flips out and gets expressed on
difficile a cocktail of two antibodies, CDA-1 and the external part of the membrane, allowing its
CDB-1 MK3415A (Merck), for neutralization of use as a target for mAbs. Another approach
toxins A and B are in phase II development (33). adopted from cancer treatment is immuno-
Clinical trial data with MK3415A given on top of modulation. Another example of a mAb being
a standard of care showed a lower recurrence tested in HCV is the two anti-PD-1 receptor
rate compared to the standard of care alone. antibodies, CTZ-11 and BMS-936558, where
This was an example of positive data where a blocking the inhibitory receptor PD-1 on CD8 T
therapeutic antibody treatment was given as an cells restores the ability of these cells to lyse
adjunct to standard of care antibiotics. HCV-infected cells.
Another cocktail of two antibodies (caStx1, Shifting the focus to HIV, a number of mAbs
caStx2; Shigamabs [LFB Biotechnologies/ which targeted the GP120 envelope protein
Thallion Pharmaceutics]) is being developed were being tested but suffered from escape
to treat infections with strains of E. coli mutants causing a rebound of viral loads. A
producing Shiga toxin. There is currently no different strategy targeting conserved receptors
treatment for the hemolytic uremic syndrome such as CCR5 or CXCR4, which serve
caused by infections with these strains of E. coli coreceptors for the virus on target CD4 T
which affects around 300,000 people every cells, has had some success in reducing viral
year. Phase I trials showed the cocktail to be loads (38, 39). Two such antibody therapies,
well tolerated in healthy volunteers, and a phase Ibalizumab (WuXi Pharma Tech) and PRO140
II study is looking at pharmacokinetics and (CytoDyn), are in late-stage clinical develop-
efficacy of treatment (34). ment and have caused some modest optimism.
In rabies the current treatment practice
after potential exposure is vaccination and
Antivirals
administration of immunoglobulin from vacci-
Synagis (palivizumab, MedImmune), for the nated humans (human rabies immunoglobulin
prevention of RSV infection in infants, is [HRIG]) or from horses. In Asia >50,000 people
currently the only approved monoclonal anti- annually die from rabies (35), and a shortage of
18 HEY

specific immunoglobulins has resulted in the Antibody-based treatments have been


testing of a polyclonal antibody product (CL- going through a time of great excitement and
184, foravirumab, Sanofi/Crucell); virus-neu- development in generating treatments for
tralizing effects similar to what is observed with cancer and inflammatory diseases, and many
immunoglobulin treatment in dosed humans new antibody-based products are now ap-
has been observed (40). proved for clinical use (Table 1). This devel-
opment passed by the field of infectious
diseases, and despite multiple programs in
Antifungal
multiple diseases only one treatment, the anti-
Fungal infections with yeasts such as Candida RSV antibody Synagis, has been approved.
albicans and C. neoformans cause morbidity Inherent challenges such as high cost, the
and mortality in immunocompromised individ- need for parallel development of specific
uals. For candidiasis one antibody, efungumab diagnostics, and ease of storage and dosing
(Mycograb, Novartis), which targets heat- have been difficult to overcome in a field
shock protein 90 (HSP90), was in late-stage where cheap and functioning antibiotics are
clinical development for treatment on top of available. For cancer and inflammatory dis-
antifungal therapy. However, safety concerns ease there is usually no alternative treatment,
and lack of proven efficacy compared to which increases the need and demand for a
antifungal treatment alone resulted in discon- proper treatment and acceptance of, e.g., high
tinuation of the program. For Cryptococcus a costs of treatment. Furthermore, in many
murine antibody, MAb18b7, which targets programs a lack of predictive animal models
capsular glucuronoxylomannan and acts has resulted in many disappointments in
through complement activation, was tested in translating apparent efficacy in animals to
HIV patients, but due to generation of anti- humans. In addition, in diseases with large
mouse antibodies, this program was halted (41). potential markets the vaccine and antibiotic
development approach will generally be used.
It is therefore time to take a new and revised
FUTURE PERSPECTIVES FOR ANTIBODY look into the future to find appropriate niches
TREATMENTS IN INFECTIOUS DISEASES in infectious diseases where new antibody-
based treatments could prove their value and
There is no questioning the fact that new make a major difference. In this, the ability to
treatment options are needed in infectious provide treatments for infections in immuno-
diseases. Increasing multidrug resistance, in- compromised subjects such as HIV patients or
ability to treat immunocompromised patients, very young or elderly people, as well as
risk of bioterrorism, and new emerging diseases treatments for some of the severe potential
call for alternatives to the current armament. bioterror infections such as Ebola, could open
For that purpose, antibodies and antibody- the eyes of health care providers, researchers,
derived treatments offer very attractive tools and pharmaceutical developers and provide
and attributes to kill or neutralize infectious stepping stones for broader acceptance of the
agents, lyse infected cells, or modulate the potential for these treatments.
immune system to enable effector cells to In addition, the general approach to the
escape immunosuppressed conditions and con- format and use of these treatments should
tribute to the elimination of infections. The be reconsidered. The use of cocktails of
ability to raise antibodies to any target, and the multiple antibodies more and more seems to
ability to modulate effector functions, half-life, be required to avoid escape mutations and
and size of the treatment units makes anti- to more efficiently neutralize several toxins
bodies ideal for tailoring treatoments for spe- or target infection with different mecha-
cific infectious agents. nisms. Regulatory pathways for developing
CHAPTER 1 History and Practice: Antibodies in Infectious Diseases 19

such cocktails now seem to be in place CITATION


where mostly only the actual cocktail is
Hey A. 2015. History and practice: antibodies
tested and only limited additional clinical
in infectious diseases. Microbiol Spectrum
data demonstrating the contribution from
3(1):AID-0026-2014.
each element is needed. What is also clear is
that to generate efficacious antibody treat-
ments, further research into specific mech- REFERENCES
anisms of pathogenesis is needed, which
1. Casadevall A, Sharff MD. 1994. Serum therapy
will assist in choosing the right target revisited: animal models of infection and develop-
antigens. In this respect the vast flexibility ment of passive antibody therapy. Antimicrob
that can be achieved by modulating format, Agents Chemother 38:16951702.
effector functions, half-life, and other fea- 2. Casadevall A. 1996. Antibody-based therapies
tures provides many opportunities but also for emerging infectious diseases. Emerg Infect
Dis 2:200208.
challenges in conducting the necessary
3. Behring EA, Kitasato S. 1890. Ueber das
testing to enable selection of the product zustandekommen der diptherie-immunitt
with the optimal combination of these und der tetanus-immunitt bei thieren. Deutch
features. Med Woch 49:11131114.
Another consideration for the future is 4. Klemperer G, Klemperer F. 1891. Versuche
uber immunisirung und heilung bei der pneu-
to focus more on developing treatments
mokokkeninfection. Berlin Klin Wochenschr
that are adjunct to existing ones, e.g., 28:833835.
antibiotic treatment instead of a stand- 5. Flexner S, Jobling JW. 1908. Serum treatment
alone treatment. This might provide that of epidemic cerebro-spinal meningitis. J Exp
extra efficacy in patients with failing im- Med 10:141195.
mune response and potentially also com- 6. Rosington B. 2013. The drugs wont work: top
doctor warns of apocalypse in 20 years as bugs
pensate for and reduce the development of become resistant to antibiotics. Daily Mirror,
resistance, thereby ensuring available effi- January 24.
cacious treatment options. On the cost side, 7. WHO. 2014. Ebola response roadmap update
research into the use of the smaller and report. http://www.who.int/csr/resources/
cheaper antibody-based fragments could publications/ebola/response-roadmap/en/.
8. Chan CEZ, Chan AHY, Hanson BJ, Ooi EE.
contribute to making antibody-based treat-
2009. The use of antibodies in the treatment
ments more attractive and more efficient, of infectious diseases. Singapore Med J 50:
and cheaper ways of manufacturing anti- 663672.
bodies would contribute to this. In summa- 9. Berry JD, Gaudet RG. 2011. Antibodies in
ry, there is no doubt that antibody-based infectious diseases: polyclonals, monoclonals
treatments, through their great flexibility to and niche biotechnology. New Biotechnol
28:489501.
be designed for specific targets on infec- 10. Cauza K, Hinterhuber G, Dingelmaier-
tious agents or the host immune cells, will Hovorka R, Brugger K, Klosner G, Horvat
and should play a major role in designing R, Wolff K, Foedinger D. 2005. Expression of
new treatments for infectious diseases in FcRn, the MHC class I-related receptor for
IgG, in human keratinocytes. J Invest Dermatol
the future. Hopefully, several of the pro-
124:132139.
grams currently in development will show 11. Marasco WA, Sui J. 2007. The growth and
clinical efficacy and through their approval potential of human antiviral monoclonal anti-
provide the basis and enthusiasm for this body therapeutics. Nat Biotechnol 25:1421
process. 1434.
12. Wrammert J, Koutsonanos D, Li G-M,
Edupganti S, Sui J, Morrissey M, McCausland
ACKNOWLEDGMENT M, Skountzou I, Hornig M, Lipkin WI, Metha
A, Razave B, DelRio C, Zheng N-Y, Lee J-H,
Conflicts of interest: I declare no conflicts. Huang M, All Z, Kaur K, Andrews S, Amara RR,
20 HEY

Wang Y, Das SR, ODonnel CD, Yewdell JW, broadly neutralizing antibody against human
Subbarao K, Marasco WA, Mulligan M, immunodeficiency virus type 1. J Virol 75:
Compans R, Ahmed R, Wilson PC. 2011. Broadly 1216112168.
cross-reactive antibodies dominate the human B 24. Hessell AJ, Hangartner L, Hunter M,
cell response against 2009 pandemic H1N1 influ- Havenith CD, Beursken FJ, Bakker JM,
enza virus infection. J Exp Med 208:181193. Lanigan CM, Landucci G, Forthal DN,
13. Khler G, Milstein C. 1975. Continuous cultures Parren PW, Marx PA, Burton DR. 2007.
of fused cells secreting antibody of predefined Fc receptor but not complement binding is
specificity. Nature 256:495497. important in antibody protection against HIV.
14. Casadevall A, Dadachova E, Pirofski LA. Nature 449(7158):101104.
2004. Passive antibody therapy for infectious 25. DallAcqua WF, Kiener PA, Wu H. 2006.
diseases. Nat Rev Microbiol 2:695703. Properties of human IgG1s engineered for
15. Triggle N. Antibiotic treatments from GPs enhanced binding to the neonatal Fc receptor
fail 15% of the time. BBC News, Health, (FcRn). J Biol Chem 281:2351423524.
September 25. 26. Saylor C, Dadachova E, Casadevall A. 2009.
16. Bregenholt S, Jensen A, Lantto J, Hyldig S, Monoclonal antibody-based therapies for
Haurum J. 2006. Recombinant human poly- microbial diseases. Vaccine 275:G38G46.
clonal antibodies: a new class of therapeutic 27. Hall AE, Domanski PJ, Vernachio JH,
antibodies against viral infections. Curr Pharm Syrbeys PJ, Gorovits EL, Johnson MA, Ross
Des 12:20072015. JM, Patti JM. 2003. Characterization of a
17. Flego M, Ascione A, Cianfriglia M, Vella S. protective monoclonal antibody recognizing
2013. Clinical development of monoclonal anti- Staphylococcus aureus MSCRAMM protein
body-based therapy drugs in HIV and HCV clumping factor A. Infect Immun 71:68646870.
diseases. BMC Med 11:117. 28. Bebbington C, Yarranton G. 2008. Antibodies
18. de Kruif J, Bakker ABH, Marissen WE, Arjen for the treatment of bacterial infections: cur-
Kramer R, Throsby M, Rupprecht CE, rent experience and future prospects. Curr
Goudsmit J. 2007. A human monoclonal anti- Opin Biotechnol 19:613619.
body cocktail as a novel component of rabies 29. Weisman LE, Thackray HM, Steinhorn RH,
post-exposure prophylaxis. Annu Rev Med 50: Walsh WF, Lassiter HA, Dhanireddy R,
359368. Brozanski BS, Palmer KG, Trautman MS,
19. Lazar GA, Dang W, Karki S, Vafa OPeng JS, Escobedo M, Meissner HC, Sasidharan P,
Hyun L, Chan C, Chung HS, Eivazi A, Yoder Fretz J, Kokai-Kun JF, Mond JJ. 2011. A
SC. 2006. Engineered antibody Fc variants with randomized study of a monoclonal antibody
enhanced effector function. Proc Natl Acad Sci (pagibaximab) to prevent staphylococcal sep-
USA 103:40054010. sis. Pediatrics 128:271279.
20. Shields RL, Namenuk AK, Hong K, Meng YG, 30. Weintraub JA, Hilton JF, White JM, Hoover CI,
Rae J,Briggs J, Xie D, Lai J, Stadlen A, Li B. Wycoff KL, Yu L, Larrick JW, Featherstone JD.
2001. High resolution mapping of the binding 2005. Clinical trial of a plant-derived antibody on
site on human IgG1 for FcgRI, FcgRII, FcgRIII, recolonization of mutans streptococci. Caries Res
and FcRn and design of IgG1 variants with 19:241250.
improved binding to the FcgR. J Biol Chem 31. Baer M, Sawa T, Flynn P. 2009. An engineered
276:65916604. human antibody Fab fragment specific for Pseudo-
21. Stewart R, Thom G, Levens M, Gler-Gane G, monas aeruginosa PcrV antigen has potent anti-
Holgate R, Rudd PM, Webster C, Jermutus bacterial activity. Infect Immun 77:10831090.
L, Lund J. 2011. A variant human IgG1-Fc 32. Secher T, Fauconnier L, Szade A. 2011. Anti-
mediates improved ADCC. Protein Eng Des Sel pseudomonas aeruginosa serotype 011 LPS immu-
24:671678. noglobulin M monoclonal antibody panobacumab
22. Niwa R, Hatanaka S, Shoji-Hosaka E, (KBPA101) confers protection in a murine model
Sakurada M, Kobayashi Y, Uehara A, Yokoi of acute lung infection. J Antimicrob Chemother
H, Nakamura K, Shitara K. 2004. Enhance- 66:11001109.
ment of the antibody-dependent cellular cyto- 33. Migone T, Subramanian GM, Bolmer SD.
toxicity of low-fucose IgG1 is independent of 2009. Raxibacumab for the treatment of inha-
Fc-q gammaRIIIa functional polymorphism. lational anthrax. N Engl J Med 361:135144.
Clin Cancer Res 10:62486255. 34. Lowy I, Molrine DC, Leav BA. 2010. Treat-
23. Hezareh M, Hessell AJ, Jensen RC, van de ment with monoclonal antibodies against Clos-
Winkel JGJ, Parren PWHI. 2001. Effector tridium\ difficile toxins. N Engl J Med 362:197
function activities of a panel of mutants of a 205.
CHAPTER 1 History and Practice: Antibodies in Infectious Diseases 21

35. Ter Meulen J. 2011. Monoclonal antibodies in 39. Li L, Sun T, Yang K. 2010. Monoclonal CCR5
infectious diseases: clinical pipeline in 2011. antibody for treatment of people with HIV
Infect Dis Clin N Am 25:798802. infection (review). Cochrane Database Syst Rev
36. Broering TJ, Garrity KA, Boatright NK. 8:CD008439.
2009. Identification and characterization of 40. Quiambao B, Bakker A, Bermal NN. 2009.
broadly neutralizing human monoclonal anti- Evaluation of the safety and neutralizing activity
bodies directed against the E2 envelope glyco- of CL184, a monoclonal antibody cocktail against
protein of hepatitis C virus. J Virol 83:12473 rabies, in a phase II study in healthy adolescents
12482. and children. Quebec, Canada: Presentation at
37. Soares MM, King SW, Thorpe PE. 2008. RITA XX, Rabies in the Americas.
Targeting inside-out phosphatidylserine as a 41. Larsen RA, Pappas PG, Perfect J, Aberg JA,
therapeutic strategy for viral diseases. Nat Med Casadevall A, Cloud GA. 2005. Phase I
14:13571362. evaluation of the safety and pharmacokinetics
38. Bruno BJ, Jacobson JM. 2010. Ibalizumab: an of murine-derived anticryptococcal antibody
anti-CD4 monoclonal antibody for the treatment of 18b7 in subjects with treated cryptococcal
HIV-1 infection. J Antimicrob Chemother 65:1839 meningitis. Antimicrob Agents Chemother 49:
1841. 952958.
GENERAL FEATURES OF
IMMUNOGLOBULINS
Functions of Antibodies

DONALD N. FORTHAL1
2
INTRODUCTION

In the setting of infectious diseases, antibody function refers to the biological


effect that an antibody has on a pathogen or its toxin. Thus, assays that measure
antibody function are differentiated from those that strictly measure the ability
of an antibody to bind to its cognate antigen. Examples of antibody functions
include neutralization of infectivity, phagocytosis, antibody-dependent cellular
cytotoxicity (ADCC), and complement-mediated lysis of pathogens or of
infected cells.
Antibodies can impact pathogens in the presence or in the absence of
effector cells or effector molecules such as complement, and experiments
can often sort out with precision the mechanisms by which an antibody
inhibits a pathogen in vitro. In addition, in vivo models, particularly those
engineered to knock in or knock out effector cells or effector molecules are
excellent tools for understanding antibody functions. However, it is highly
likely that multiple antibody functions occur simultaneously or sequentially
in the presence of an infecting organism in vivo.
The most critical incentive for measuring antibody functions is to provide a
basis for vaccine development and for the development of therapeutic antibodies.

1
Department of Infectious Diseases, University of California, Irvine, Irvine, CA 92617.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0019-2014

25
26 FORTHAL

In this respect, some functions, such as virus ficities can neutralize a given organism, at least
neutralization, serve to inhibit the acquisition of in vitro, through multiple mechanisms.
a pathogen or limit its pathogenesis. However,
an antibody can also enhance replication or Preattachment Neutralization
contribute to pathogenesis. This review will Some antibodies have been shown to inhibit
emphasize those functions of the antibody that infectivity by binding to organisms and causing
are potentially beneficial to the host; a separate them to aggregate. Aggregation or agglutination
article is devoted to a discussion of antibody- by immunoglobulin A (IgA) may allow more
dependent enhancement of infection. In addi- efficient entrapment of bacteria in mucus and
tion, this article will focus on the effects of subsequent clearance by peristalsis (2, 3).
antibodies on the organisms themselves, rather Although aggregation is more likely to occur
than on the toxins the organisms may produce. with polymeric IgA and IgM, some neutralizing
Finally, the role of antibody in modulating IgG antibodies can aggregate poliovirus; the
T-cell immunity is not discussed in detail. aggregation results in less infectivity, probably
by reducing the number of encounters between
virus and host cells (4, 5).
ANTIBODY FUNCTIONS INDEPENDENT Antibodies have also been shown to immo-
OF EFFECTOR CELLS OR EFFECTOR bilize or paralyze organisms, such as the
MOLECULES channel catfish pathogen Ichthyophthirius
multifiliis (6). The IgA monoclonal antibody
Antibodies are capable of having an impact on (mAb) Sal4 can render Salmonella enterica
organisms in the absence of effector cells or serovar Typhimurium immobile, independently
effector molecules such as complement. For the of agglutination, although Sal4 also specifically
most part, the impact of antibodies by them- interferes with uptake into epithelial cells.
selves can be measured in vitro as neutralization Antibodies directed against Pseudomonas
of organism infectivity. Neutralization is herein aeruginosa flagella inhibit motility of that
referred to as the ability of the antibody by itself organism (7). Polyclonal antibodies, induced
to inhibit the infection of susceptible cells or, in by immunizing mice with Vibrio cholerae outer
the case of some extracellular organisms, to membrane vesicles, protect suckling mice from
inhibit an initial pathogenic step. Importantly, oral V. cholerae challenge, likely by inhibiting
as described below, neutralization involves the motility of the organism (8). Antibody may
many potential mechanisms. Furthermore, it slow the random movement of human immu-
should be emphasized that other antibody nodeficiency virus type 1 (HIV-1) in vaginal
functions in addition to neutralization may mucus, presumably reducing the number of
ultimately be involved in the prevention or times the virus can make contact with the
clearance of infection, even by antibodies that epithelial surface; this antibody function
neutralize the relevant organism in vitro (1). appears to rely in part on Fc interactions with
components of the mucus (9).
Some antibodies appear to destabilize orga-
Neutralization of Infectivity
nisms, rendering them noninfectious. For ex-
In vitro, antibodies are capable of blocking the ample, the anti-foot-and-mouth-disease virus
infectivity or pathogenesis of viruses, bacteria, mAb 4C9 disrupts virion capsids, possibly by
parasites, and fungi. Neutralization generally mimicking the virus cell receptor (10). A neu-
occurs as a result of interfering with an tralizing antibody against the E1 glycoprotein
organisms attachment to host tissues. Howev- of Sindbis virus also induces conformational
er, it is now clear that several mechanisms changes (11). Binding of HIV-1 gp120 can result
account for neutralization and that a single in the shedding of gp120, leaving the trans-
antibody or antibodies with different speci- membrane glycoprotein on the surface. How-
CHAPTER 2 Functions of Antibodies 27

ever, the overall effect of such shedding on ization requires the engagement of more than
neutralization sensitivity is unclear (12). one antibody on the virion (27). Both antibody
mAbs binding to a surface protein of affinity and the accessibility of epitopes on the
Borrelia can kill the organism by inducing organism are the critical factors in determining
pores in the outer membrane (13). A mAb whether antibody binding will exceed the
directed against fungal heat shock protein threshold required for neutralization. Thus,
90, a component of yeast cell walls, directly for example, one cannot necessarily predict
inhibits the growth of Candida (14, 15) and neutralizing potency by measuring antibody
works in synergy with antifungal drugs to affinity alone or on the basis of epitope
inhibit Cryptococcus neoformans (16). IgG1 specificity. Antibody Fab or F(ab)2 fragments
and IgM mAbs that bind to the C. neo- are often capable of providing sufficient block-
formans capsule affect gene expression, ade of attachment to inhibit neutralization.
lipid biosynthesis, cellular metabolism, and These and other details regarding virus neu-
protein phosphorylation or susceptibility to tralization, including kinetics and requirements
amphotericin B (17). Other mechanisms by for steric hindrance can be found elsewhere (12,
which antibody inhibits bacterial and fungal 28, 29).
infections directly and prior to attachment Adhesion of bacteria to the surface of host
have been described (18, 19, 20). cells or tissue allows targeting of the organism
to a specific cell type and allows the bacteria to
Interference with Pathogen Attachment resist physical removal by hydrodynamic shear
Antibodies that bind to pathogen ligands forces (30). Thus, adhesion is a first step in
essential for attachment of the pathogen to its bacterial pathogenesis. The molecules respon-
host receptor have been described for many sible for bacterial adhesion are known as
pathogens. In the case of viruses, such anti- adhesins and are generally incorporated into
bodies generally inhibit infectivity without pili or fimbriae (30, 31). These adhesins are
altering their cognate antigen, thus strictly targets for antibodies that, in a manner some-
inhibiting by virtue of steric interference. what analogous to virus neutralization, can
This mechanism of virus inhibition has been inhibit attachment (32, 33, 34). Thus, vaccines
described for many enveloped and non- have been developed in order to elicit anti-
enveloped antibodies. Well-studied example bodies directed against adhesions. In most
are antibodies against HIV-1 gp120 that inter- cases, this strategy has failed because of
fere with binding of gp120 to CD4 (21). In sequence variation in the structural proteins
addition, antibodies that neutralize, among of fimbriae. Nonetheless, vaccination with
others, flaviviruses (22), Newcastle disease FimH was able to reduce bladder infection of
virus (23), papillomavirus (24), and rotavirus mice and monkeys with uropathogenic Esche-
(25) may do so by interfering with attachment. richia coli (35, 36, 37). The use of this vaccina-
Some antibodies that block virus attachment do tion strategy in humans is made difficult by a
not bind directly to the site on the virus shared epitope between FimH and human
involved with attachment. For example, an LAMP-2 and thus a fear of autoimmunity (38).
antibody against human rhinovirus type 14 Another example of adherence inhibition was
binds to surrounding viral structures but described by Manjarrez-Hernanadez et al., who
nonetheless sterically hinders interactions be- found that secretory IgA (sIgA) in breast milk
tween the virus and its intercellular adhesion was able to inhibit the adherence of entero-
molecule-1 host receptor (26). pathogenic E. coli to cells (39). An mAb against
The stoichiometry of antibody-antigen in- lipoarabinomannan, a surface lipoglycan of
teractions required for neutralization has been Mycobacterium tuberculosis, is able to prevent
studied for many viruses, and evidence supports adherence of M. tuberculosis to human mono-
a multiple hit phenomenon in which neutral- cyte-derived macrophages (40). Antibodies can
28 FORTHAL

also inhibit the attachment of bacteria to abiotic infection of a number of different organisms
surfaces (41). (50, 51, 52, 53, 54, 55, 56, 57).
Antibodies to merozoite surface protein 1
(MSP1) on Plasmodium spp. can protect
Postattachment Neutralization
rodents against infection (42). One mecha-
nism that might account for this is inhibition Inhibition of fusion/entry
of the attachment of the parasite to red blood Intracellular pathogens can be neutralized by
cells (43, 44). Plasmodium-infected red antibodies at postattachment steps in their life
blood cells express Plasmodium falciparum cycle. In the case of viruses, several studies have
erythrocyte membrane protein 1 (PfEMP1), identified antibodies that inhibit fusion of viral
which mediates binding to host endothelia and host membranes or entry into susceptible
and placenta. Antibodies have been elicited target cells. For enveloped viruses, antibodies
that can inhibit the interaction between can block an interaction between a viral protein
infected erythrocytes and chondroitin sul- necessary for fusion and its cellular receptor
fate proteoglycan, their ligand on placenta (58). mAb 2F5, an HIV-1-neutralizing antibody,
(45). Inhibition of binding in this manner may block fusion of HIV-1 by obstructing the
would not impact infection per se, but might juxtaposition of viral and cellular membranes
influence pathogenesis. (29, 59). 4E10, another HIV-1-neutralizing
As with other organisms, binding of fungi to mAb, may interfere with the formation of
host-cell surfaces is a first step in infection. fusion-competent complexes of gp41 (60). In
mAb 2G8, directed against 1,3-glucan, can the case of West Nile virus, a neutralizing
inhibit binding of Candida albicans to human monoclonal antibody likely sterically constrains
epithelial cells (49). It should be noted that mAb low-pH-mediated rearrangements of E pro-
2G8 also directly inhibits fungal growth and teins (61, 62). Similarly, anti-influenza virus
facilitates antifungal activity of human poly- hemagglutinin (HA) antibodies can hinder the
morphonuclear neutrophils (49, 50). Other low pH-induced structural changes necessary
antibodies can also inhibit adhesion of C. for fusion of viral and endosomal membranes
albicans to HEC cells (51) and of C. neoformans (63, 64). It is possible that anti-influenza HA
to a human lung epithelial cell line (52). As with antibodies can inhibit both attachment and
bacteria, antibodies can inhibit C. albicans postattachment steps (65).
adherence to abiotic surfaces (53). An interesting twist on fusion inhibition was
In the case of several parasites that infect described for an mAb against influenza virus
the gastrointestinal tract, the mechanisms HA. The mAb becomes internalized at acid pH
by which Ig, and, in particular, IgA, may through the Fc neonatal receptor (FcRn) and
function are unclear, but it is likely that the reduces viral replication following apical expo-
inhibition of attachment plays a role (46). sure of Madin-Darby canine kidney cells to
For example, mucosal anti-Giardia IgA anti- influenza virus. As virus, mAb, and FcRn
bodies may prevent infection by inhibiting colocalize within endosomes, it is possible
attachment of the organism to the intestinal that inhibition of infectivity occurs by interfer-
epithelium (47). Intestinal IgE antibodies ing with fusion of viral envelope and endosomal
might contribute to the elimination of Tri- membranes (66).
chinella spiralis in rats, possibly by blockade Nonenveloped viruses generally enter
of the attachment to intestinal epithelium cells by endocytosis, and escape from the
(48), and immune serum can block the attach- endocytic vesicle is mediated by capsid
ment of Cryptosporidium parvum to epithelial protein. Antibodies against polio virus may
cells (49). stabilize the capsid and prevent the struc-
Finally, antibodies generated against the tural rearrangements necessary for vesicle
host receptors themselves can also block escape (29, 67, 68).
CHAPTER 2 Functions of Antibodies 29

Inhibition of other steps in organism structural change results in transcriptionally


life cycles incompetent particles (78, 79).
A number of studies have revealed the ability A novel mechanism of intracellular virus
of antibodies to inhibit organisms once they inhibition was described by Mallery et al.,
have successfully entered cells. In order for wherein antibody bound to adenovirus inter-
intracellular neutralization to be accom- acted with cytosolic TRIM21; this interaction
plished, antibodies must be internalized by resulted in the antibody-bound virus being
host cells. Internalization of antibodies can targeted to the proteasome for degradation
occur as a result of coating of the organism, in (80). This mechanism of inhibition would not
which case, the coated organism must be be expected to work with enveloped virus,
capable of cell entry, or through Fc receptors. since the antibody would be shed along with
In addition, cells have been engineered to the envelope prior to internalization within the
express intracellular antibodies (intrabodies) cytoplasm.
for potential therapeutic purposes (69). Intra- An interesting example of intracellular
cellular neutralization can potentially inter- antibody function in a bacterial infection was
rupt an organisms life cycle by interfering described by Wang et al., who showed that a
with the release, replication, or expression of mAb against Anaplasma phagocytophilum in-
genomic material. As an example, adenovirus hibits morulae formation within HL-60 cells (a
type 5 antihexon mAb (9C12) allows viral human promyelocytic leukemia cell line) (81).
attachment, cell entry, and intracellular trans- A mAb against listeriolysin O, the pore-forming
port of the virus to the nuclear periphery (70). toxin of Listeria monocytogenes, blocks L.
Nonetheless, 9C12 neutralizes virus infectivity, monocytogenes infection within macrophages.
likely by interfering with capsid uncoating and Inhibition likely occurs as a result of intracel-
the release of viral genome (70). A rabbit anti- lular neutralization of a secreted Listeria
HPV16 L2 serum was able to neutralize virulence factor (82).
HPV16 pseudoviruses through a mechanism With respect to parasites, IgA is reported to
that appeared to involve, at least in part, inhibit the replication of Toxoplasma gondii in
blocking the transport of viral genome to the enterocytes (83). In addition, a mouse mono-
nucleus (71). clonal IgG2b antibody, which enters host
IgA directed against surface proteins or fibroblasts upon invasion of the antibody-
glycoproteins can mediate neutralization of treated organism, inhibits the intracellular
Sendai virus, influenza virus, and measles growth of T. gondii (84).
virus within susceptible target cells (72, 73,
74). In addition, IgA directed against measles
virus M and N proteins, which are internal to INHIBITION OF LATER STEPS
the membrane, can inhibit measles virus
replication within Vero cells (73, 75). Poly- Antibodies are capable of binding to nascent
meric IgA or sIgM can intracellularly block viruses and inhibiting their liberation from
the transcytosis of HIV-1 through epithelial infected cells. This function has been de-
cells. Although the epithelial cells are not scribed for antibodies directed against the
thought to be a target of HIV-1 infection and neuraminidase of influenza A virus (85). It
replication, such blocking of transcytosis could has also been suggested that the antibody
block access in vivo to subepithelial CD4+ cells directed against influenza A virus M2 pro-
(76). Similarly, IgA inhibits transcytosis of tein influences the efficiency of virus bud-
rotavirus through polarized Caco-2 cells (77). ding (86). An mAb against rubella virus E1
IgA can also introduce a conformational glycoprotein was reported to delay the
change in the rotavirus VP6 trimer, which is release of virus, perhaps by affecting virion
exposed after internalization of virus. The assembly (87).
30 FORTHAL

ANTIBODY FUNCTIONS DEPENDENT ing antibodies against several viruses, includ-


ON COMPLEMENT ing influenza viruses (96), Newcastle disease
virus (97), herpes simplex virus (98), and
Activation of the complement cascade by Japanese encephalitis virus (99). Paramyxo-
antibody can result in the lysis of organisms viruses represent an interesting case in terms
or of infected cells (88). In addition, orga- of the role of complement, since one study has
nisms bound by complement can be inter- shown that antibody can neutralize human
nalized by phagocytic cells, with resultant parainfluenza virus type 2 with little contri-
clearance of the organism. Internalization bution by complement, whereas neutralization
through complement receptors on antigen- of mumps virus and simian virus 5 was
presenting cells can also result in the dependent on complement (100).
processing of antigen for presentation to T In a mouse model of respiratory syncytial
lymphocytes. The details of complement virus, passive immunization of a nonneu-
activation have been reviewed elsewhere tralizing mAb was shown to protect animals
(88). It is important to note that antibodies from intranasal challenge. The mAb lost
that bind and activate complement may also protective activity as a Fab, and decom-
directly inhibit pathogens in the absence of plementation of mice reduced the degree of
complement. Complement activation may protection (101). Similarly, protective mAbs
also have an indirect effect on pathogens by against Semliki Forest virus lose some effect
recruiting and activating leukocytes to sites in complement-depleted mice (102).
of infection (89, 90). Similarly, comple- HIV-1 Env-specific antibodies are capable
ment-activating antibodies may engage Fc of lysing HIV-1-infected cells or virus in the
receptors (see below). The Ig subtype and presence of complement. However, such com-
IgG subclass of antibody are major deter- plement-mediated effects are inhibited by the
minants of complement activation (91). For presence of regulators of complement activa-
the most part, in this review, we limit the tion found on infected cells or on the virus itself
discussion to antibodies that affect patho- (103, 104). Recently, complement-mediated
gens in the presence of complement but phagocytosis of apoptotic, HIV-1-infected T
that, in the absence of complement, either cells by polyreactive antibodies has been
have no or reduced antimicrobial activity. reported (105). Another study has found that
A role for IgM and complement in limiting antibody from human immunodeficiency virus
West Nile virus infection in mice has been type 2 (HIV-2)-infected subjects is more potent
suggested (92). More recently, Vogt et al. than that from HIV-1-infected subjects in
determined that a nonneutralizing mouse complement-mediated inactivation of the re-
IgG1 mAb decreased West Nile virus load in spective virus. Given the multiple potential
mice in a manner that required C1q as well as consequences of complement, either directly
phagocytic cells and FcRIII (93). C1q, as well or indirectly, its role in human immunodefi-
as FcRs, contributed to the enhancement of ciency virus (HIV) infection in vivo remains
CD4+ T-cell responses mediated by non- unsettled (106, 107, 108).
neutralizing anti-respiratory syncytial virus Natural antibodies, generally of the IgM
(RSV) antibody during RSV infection (94). subtype, activate complement and can neutral-
Antibodies that both neutralize and medi- ize influenza virus (109). Moreover, natural
ate complement-dependent lysis of influenza IgM recognizing influenza virus or a surface
virus-infected cells may provide broader strain protein of Leishmania may be involved in
cross-reactivity than antibodies that only neu- regulating CD4+ or CD8+ T cells through
tralize (95). Furthermore, the addition of complement (110, 111).
complement has long been known to increase A unique function of antibody is to initiate
the infectivity-inhibiting activity of neutraliz- the clearance of pathogens via complement
CHAPTER 2 Functions of Antibodies 31

activation and binding to erythrocyte comple- through complement receptors (124). IgM also
ment receptor 1 (CR1); the result of such binding promotes complement deposition and PMN
sequesters the pathogen from invading suscep- phagocytosis of C. neoformans (125). Interest-
tible tissue and may facilitate the destruction of ingly, IgM, IgA, and IgG1 can promote the
the organisms by tissue macrophages (112). This phagocytosis of C. neoformans through comple-
phenomenon was first noted for bacteria by ment receptors in the absence of complement;
Nelson in 1953 (113). this occurs because of an antibody-mediated
Bacterial pathogens have developed strate- change in the organisms capsule that allows an
gies to evade the effects of complement. interaction with complement receptors (126).
However, in the presence of specific antibody, Nonspecific autologous antibodies can
effective activation of complement can result in opsonize P. falciparum-parasitized erythro-
the death or clearance of organisms such as cytes, activate complement, and clear the
Neisseria meningitidis, Neisseria gonorrhoeae, infected cells through phagocytosis (127). In-
and Haemophilus influenzae (114). Individuals terestingly, antibody and complement deposi-
with complement deficiencies are at higher risk tion and phagocytosis are increased with
of infection with these organisms, and, in the erythrocytes from individuals with glucose-6-
case of N. meningitidis and H. influenzae, phosphate dehydrogenase deficiency, sickle
vaccine-induced antibody may protect through trait, and -thalassemia; it has been proposed
complement-mediated bacterial killing (115, that this antibody-mediated phenomenon
116). However, even with late complement underlies the protection against falciparum
component deficiencies, C3b deposition allows malaria in individuals with certain genetic
antibodies to kill the organisms by complement- disorders of red blood cells (127, 128, 129,
mediated phagocytosis (115, 116, 117, 118, 119). 130). Antibody and complement can promote
Antibody-mediated complement lysis of the killing of P. falciparum blood forms by
Legionella pneumophila is ineffective; however, THP-1 cells (a myelomonocytic cell line) and
organisms opsonized with both antibody and neutrophils (131, 132). In addition, antibody-
complement are phagocytosed by polymorpho- dependent complement-mediated lysis of
nuclear leukocytes (PMNs), although killing of schizonts results in growth inhibition of P.
ingested bacteria is limited (120). Vaccination falciparum (133). Antibodies to P. falciparum
of humans with an oral typhoid vaccine, gametes can abolish infectivity of the gametes
M01ZH09, results in antibodies that are bacte- to mosquitoes; the gametes are lysed in the
ricidal to S. enterica serovar Typhi in the presence of complement and antibody, and
presence of complement; the antibodies also antibody that binds to gametes but does not lyse
promote phagocytosis of S. enterica serovar them does not abolish mosquito infectivity
Typhi by macrophages in a complement- (134, 135, 136, 137). Antibody has also been
independent manner (121). shown to clear experimental Trypanosoma
Antibody and complement augment proin- brucei infection in a manner dependent on C3
flammatory cytokine production of human and associated with uptake of organisms in the
peripheral blood mononuclear cells stimulated liver (138). Moreover, clearance of African
with C. albicans, which could be a factor in host trypanosomes by IgM, which is a major factor
defense against C. albicans infection (122). Han in controlling parasitemia, is mediated by
et al. found that protective IgM or IgG3 mAbs complement and CR3 (139). Complement may
more efficiently bind C3 to the yeast cell than also be involved in the pathogenesis of severe
does a nonprotective mAb and that protection is malaria (140, 141).
likely associated with enhanced phagocytosis Finally, it is important to note that antibodies
and killing (123). In C. neoformans, immune that neutralize in vitro only in the presence of
serum or an IgG1 mAb localize C3 at the edge of complement may protect in vivo through other
the organisms capsule, allowing phagocytosis means (142).
32 FORTHAL

ANTIBODY FUNCTIONS DEPENDENT ON through other receptors (including comple-


FC-FC RECEPTOR INTERACTIONS ment receptors) (144). Engagement of FcRs can
also inhibit intracellular pathogens without
Much of the biological activity of antibody is apparently killing the host cell (159). Modula-
mediated through interactions between Fc and tion of inflammation is another FcR-mediated
Fc receptors found on a number of cells antibody function that can impact several
important for host defense. The engagement pathogens (160, 161, 162, 163, 164). Finally,
of Fc receptors by immune complexes (ICs) studies have documented the impact of FcR
results in several downstream effects, de- engagement on assays used to measure the
pending on the Fc receptor-bearing cell, the neutralizing activity of antibodies (165, 166, 167,
form of the IC, the cytokine milieu, and the 168).
presence of complement. Fc receptor-mediated
antibody activity can impact virus, bacteria,
Antibody-Dependent Cellular
fungi, and parasites and can have beneficial or
Cytotoxicity
adverse consequences to the host.
Fc receptors have been identified for IgG ADCC occurs when antibody forms a bridge
(FcR), IgE (FcR), and IgA (FcR), and for between an infected target cell (or directly
both IgA and IgM (FcR/). Five FcRs with some pathogens) and an FcR-bearing
have been identified in humans: FcRI, effector cell. The result of this three-way
FcRIIa, FcRIIb, FcRIIIa, and FcRIIIb interaction is the death of the target cell,
that differ from one another in their cellular either by lysis or apoptosis. ADCC is likely to
distribution, function, and binding to IgG Fc play an important role in the clinical effects
(143, 144). There are two known forms of of antitumor mAbs, such as rituximab and
FcR and one expressed form each of FcR trastuzumab, but its role in infections is less
and FcR/ (144). FcRIIa, FcRIIIa, and clear and complicated by the multiple func-
FcRIIIb are each encoded by polymorphic tions of antibody (169, 170, 171).
genes that result in phenotypically different ADCC, first described against virus-infected
receptors with respect to binding to differ- cells by Shore et al. for HSV-1 (172), becomes
ent IgG subclasses (145, 146, 147, 148, 149). most interesting with regard to antibodies that
As a rule, IgG1 and IgG3 bind best to FcRs, protect animals but that poorly neutralize the
whereas IgG2 and IgG4 bind less well (150). pathogen in vitro. Nonneutralizing mAbs di-
Despite the similar magnitude of IgG1 and rected against HSV-2 glycoproteins can protect
IgG3 binding to FcRs, it has been reported mice after a footpad injection of a lethal dose of
that IgG3 mAbs are less able to mediate HSV-2. The mAbs are equally efficient in
phagocytosis of antibody-coated red blood protection in complement-sufficient and com-
cells than are IgG1 mAbs, whereas ADCC plement-deficient mice (173). More recently,
activity of IgG3 is greater than that of IgG1 Gorander et al. found that vaccination of mice
(151). In addition, glycosylation of the Fc with glycoprotein G of HSV-2 plus CpG could
segment of antibody can impact binding to protect animals from vaginal challenge with
FcRs (152, 153, 154, 155). HSV-2. The protective vaccine was associated
Interactions between Fc and FcRs can result with CD4+ T-cell interferon- responses. In
in the death of pathogens or of cells infected addition, the vaccine resulted in nonneu-
with pathogens by a process known as anti- tralizing antibodies that mediated ADCC and
body-dependent cellular cytotoxicity (156, 157, might have been involved in protection (174).
158). Fc-FcR interactions are also important for Chu et al. found that passive infusion of IgG
phagocytosis of pathogens or of infected cells, antibodies decreased symptoms and mortality
although phagocytosis can occur in the absence and decreased vaginal viral quantity in normal
of antibody or in the presence of antibody mice infected with HSV-2; although the anti-
CHAPTER 2 Functions of Antibodies 33

body had neutralizing activity, protection was ADCC has been documented in vitro for a
significantly diminished in mice lacking FcR number of parasites. IgG opsonized T.
expression (175). Influenza A virus M2 vacci- spiralis larvae are susceptible to ADCC by
nation results in partial protection of mice from eosinophils, neutrophils, and monocytes
influenza A infection that is mediated by (187, 188). Schistosomula are killed by
nonneutralizing antibodies; ADCC is likely eosinophils, macrophages, or platelets in
involved, since protection is not dependent on the presence of specific antibody, including
complement, whereas NK cells depletion IgE and IgG (189, 190, 191, 192). In vivo
reduces the protective effect (176). protection of rats from Schistosoma mansoni
The role of ADCC in HIV-1 and other infection is likely the result of such IgE-
lentivirus infections has been reviewed re- mediated ADCC (193). Antibodies that bind
cently (156, 177). A great deal of correlative poorly to FcRs, such as IgM, IgG2, and IgG4,
evidence in monkeys, as well as in humans, can inhibit ADCC against schistosomula and
suggests a role for ADCC or other FcR- have been epidemiologically linked to in-
mediated antibody activities in preventing creased susceptibility of infection in humans
or modulating lentivirus infections. A more (194, 195, 196). ADCC mediated by IgG or IgE
definitive study has demonstrated that mu- and by macrophages, eosinophils, or neutro-
tations in the Fc segment of antibody that phil effector cells has also been shown in
abrogate FcR binding render a neutralizing vitro to kill larval or adult filaria (197, 198,
mAb (IgG1b12) less protective in vivo 199, 200, 201, 202). In vivo, clearance of
against vaginal SHIV challenge than the Brugia malayi microfilaria is very likely
unmutated mAb (1). However, whether the mediated through ADCC (203). ADCC activ-
decreased protection is due to a lack of ity against trypanosomes and other parasites
ADCC or to some other FcR-mediated activ- has also been documented (204, 205, 206,
ity remains unknown. 207, 208, 209, 210).
There is scant literature on the role of It is important to note that many of the
ADCC in bacterial infections. However, com- effector cells mediating ADCC against parasites
plement-independent killing of bacteria in vitro are capable of antibody-mediated phagocytosis
in the presence of killer lymphocytes has been as well. In both ADCC and phagocytosis,
described for N. meningitidis and Shigella organisms are killed and radioisotopes or dyes
flexneri (178, 179). IgA, as well as IgG, in are released, and distinguishing between these
combination with lymphocytes from murine two antibody functions requires careful con-
gut-associated lymphoid tissue, is reported to sideration (204, 205).
mediate ADCC against S. flexneri and Salmo- Finally, antibodies may also inhibit infec-
nella spp. (180, 181). Similarly, the same group tions in a manner that requires the components
found antibacterial activity against Streptococ- of ADCC (i.e., infected target cells, antibody,
cus pneumoniae by mouse lung lymphocytes in and FcR-bearing effector cells) but does not
conjunction with IgA (182). ADCC has also necessarily rely on target cell lysis. Thus,
been described for Ehrlichia risticii and antibody-dependent cell-mediated inhibition
Coxiella burnetii-infected cells and for Brucella of P. falciparum has been described, where the
abortus, although, as with other bacteria, the development of intracellular parasites is
role, if any, of ADCC in these infections in vivo blocked in a manner dependent on blood
is unknown (183, 184, 185). In the case of C. monocytes and antibody; triggering of both
burnetii, passive antibody treatment can pro- FcRIIa and FcRIIIa may be required, but
tect mice from C. burnetii infection in common erythrocyte target cells do not appear to be
-chain knockout mice, suggesting that Fc- killed (159, 211, 212). Forthal et al. have
FcR interactions were not required for pro- described antibody-dependent cell-mediated
tection (186). virus inhibition (ADCVI) with measles virus
34 FORTHAL

HIV, simian immunodeficiency virus (SIV) survival. A particularly interesting point


and simian-human immunodeficiency virus about this study was the need for PMNs
(SHIV) (1, 160, 213, 214). ADCVI is a measure despite the high neutralizing titer of the
of virus inhibition occurring as a result of infused immune serum; this finding is consis-
antibody-FcR interactions and is likely depen- tent with that of Hessell et al. where maximal
dent on combinations of ADCC, phagocytosis, protection against SHIV was afforded by a
and chemokine/cytokine production. neutralizing antibody that engaged Fc recep-
tors (1). Huber et al., using passive antibody
transfer in FcR/ mice, also concluded that
Phagocytosis
phagocytosis is important in the clearance of
The internalization and degradation of anti- influenza virus (220). In all of these studies,
body-coated pathogens by phagocytes via FcRs however, it is not possible to precisely define
has been well described for a number of the antibody function responsible for protec-
organisms and is likely a critical antibody func- tion, because phagocytosis, ADCC, or soluble
tion for clearance of pathogens in vivo. Since factors could have contributed.
phagocytosis and ADCC often require the same An interesting phenomenon related to
components (antibody and effector cells), it may phagocytosis was described by Chan et al.
be difficult to definitively and specifically (221) who showed that inhibition of dengue
demonstrate a role for phagocytosis in pre- virus phagocytosis, by aggregating virus and
venting or modulating infections in animals or cross-linking of FcRIIb, resulted in neu-
humans. tralization of virus infectivity.
With respect to viruses, passive infusion of Phagocytosis of antibody-coated infected
antibodies results in the rapid elimination of cells, in addition to phagocytosis of immune
cell-free organisms from the blood of animals complexed cell-free virus, could be a contrib-
(215, 216). This is consistent with information utor to protection, although virus-infected
indicating that the rate of clearance of antigens cells can be phagocytosed in the absence of
by the reticuloendothelial system is greatly antibody (222, 223). Surprisingly, two studies
increased in the presence of specific anti- indicated that neither human monocytes nor
bodies (217, 218). human neutrophils were able to phagocytose
A recent example comes close to demon- IgA or IgG immune complexes formed with
strating a key role for phagocytosis in pre- influenza virus in vitro (224, 225).
venting a viral infection (93). In that study, a FcR-mediated phagocytosis and clearance
poorly neutralizing antibody against the West of Bordetella pertussis has been demonstrated
Nile virus envelope could reduce viremia in in vitro and in a mouse model (226). Similarly,
mice via an FcRIII- and C1q-dependent natural and vaccine-induced antibodies medi-
mechanism that required phagocytic cells. ate phagocytosis of S. pneumoniae, and such
Since NK cells did not seem to be involved, it FcR-mediated phagocytosis may play a role in
is less likely that ADCC played a significant protection (227). In FcRIIb-deficient mice,
role in protection. However, it remains possi- phagocytosis and survival after S. pneumoniae
ble that the lysis of infected cells mediated by challenge are both improved relative to control
the phagocytes, in addition to or instead of mice, although the survival advantage is re-
phagocytosis of antibody-coated virus, was versed after immunization followed by chal-
involved in protection. A possible role for lenge with a high dose of bacteria (227). In the
phagocytosis in clearing influenza virus from later case, it is likely that inflammatory cyto-
the lungs of mice was suggested by Fujisawa kines triggered by interactions between
(219). In that study, both PMNs and the passive antipneumococcal antibody and activating
infusion of neutralizing antibody were re- FcRsin the absence of the inhibitory
quired for maximum viral clearance and FcRIIbwere responsible for the higher
CHAPTER 2 Functions of Antibodies 35

mortality. Other studies in mice have found malaria-endemic areas can mediate the phago-
that antibody-mediated protection from S. cytosis of P. falciparum-infected erythrocytes
pneumoniae does not depend on FcRs (228). by monocytes (245). In addition, the PfEMP1 is
In humans, IgG2, a relatively inefficient acti- the major target of antibodies that mediate
vator of complement, is thought to be impor- phagocytosis, and anti-PfEMP1 antibodies are
tant in protection against S. pneumoniae. associated with a reduced risk of developing
Although IgG2 is also relatively poor at symptomatic malaria (246).
engaging FcRs, it binds best to the H isoform
of FcRIIa and to the V isoform of FcRIIIa
(150). Consistent with a role for FcR-mediat- OTHER ANTIBODY FUNCTIONS
ed phagocytosis, there appears to be an
association between homozygosity for the R Apart from specific effects on organisms,
isoform of FcRIIa and severe or invasive antibodies may modulate inflammation and
pneumococcal disease (229, 230). Moreover, thereby indirectly affect pathogenesis. Such
PMNs from FcRIIa HH homozygous donors immune modulation is well described for FcR
have higher phagocytic activity against anti- triggering by immune complexes, which
body-opsonized S. pneumoniae (231). In the results in the generation, secretion, or repres-
case of N. meningitidis, complement-mediated sion of various pro- or anti-inflammatory sub-
clearance or bactericidal activity appears to be stances (247, 248, 249, 250, 251, 252, 253, 254,
more important than FcR-mediated phagocy- 255, 256). In addition to modulation of cyto-
tosis (see above). However, FcR-mediated kines by FcRs themselves, internalization of
phagocytosis can be demonstrated in vitro immune complexes via FcRs can result in the
(232, 233). Furthermore, some studies, but engagement of toll-like receptors, adding a
not all, have found associations between further layer of complexity and control over
FcRIIa genotypes and susceptibility to or inflammation (255, 257, 258, 259, 260).
severity of meningococcal infection (233, 234, An example of the role of FcRs in
235, 236, 237, 238). Finally, T cells capable of inflammation was the demonstration that
phagocytosing antibody-opsonized E. coli via soluble FcRII, by competing with IC binding
FcRIIIa have been described (239). to cellular FcRs, can limit the inflammation
Opsonization and phagocytosis by IgG due to the IC (i.e., the Arthus reaction) (261).
subclass-switched mouse mAbs has been Soon thereafter, it was established that the
described for C. neoformans (240). Passive Arthus reaction was markedly attenuated in
infusion of the mAbs had some effect on FcR-chain knockout mice (262). These types
clearing yeast from mice, but the phagocytic of studies have important implications for
activity in vitro did not correlate well with autoimmune diseases (263).
clearing of organisms in vivo (241). A study Engagement of FcRIIb by IgG immune
of C. neoformans phagocytosis has sug- complexes serve to regulate B-cell activity
gested a specific receptor for IgG3 in mice and survival and may serve as a means of
different from the known FcRs (242). A maintaining peripheral tolerance for B cells
recent study using X-linked immunodefi- (264, 265, 266). Immune complex binding
cient mice indicated that IgM promotes and internalization via FcRs also result in
containment of C. neoformans in the lungs dendritic cell maturation and in efficient
by augmenting phagocytosis (243). major histocompatibility complex class I-
In many cases, parasites may be too large for restricted presentation of the exogenous
phagocytosis: lysosomal and parasitic mem- peptides making up the immune-complexed
branes fuse after Fc-FcR (, , or ) interactions, antigen (267).
resulting in the lysis of parasites extracellularly Another important mechanism of im-
(244). However, IgG from individuals living in mune modulation by antibodies is through
36 FORTHAL

the activation of complement components, REFERENCES


which can then serve as chemotactic agents 1. Hessell AJ, Hangartner L, Hunter M,
(268). Moreover, C5a anaphylatoxin is in- Havenith CE, Beurskens FJ, Bakker JM,
volved in immune complex-mediated injury Lanigan CM, Landucci G, Forthal DN,
in part because it results in a shifting of the Parren PW, Marx PA, Burton DR. 2007. Fc
balance between activating and inhibitory receptor but not complement binding is im-
portant in antibody protection against HIV.
FcRs toward a more inflammatory pheno- Nature 449:101104.
type (269). Activation of C3bi on immune 2. Brandtzaeg P. 2007. Induction of secretory
complexes can result in blunting of the immunity and memory at mucosal surfaces.
inflammatory response by diverting inter- Vaccine 25:54675484.
actions of the IC away from FcRs and 3. Phalipon A, Cardona A, Kraehenbuhl JP,
Edelman L, Sansonetti PJ, Corthesy B. 2002.
toward CR3 (270).
Secretory component: a new role in secretory
IgA-mediated immune exclusion in vivo. Immu-
nity 17:107115.
CONCLUSIONS 4. Brioen P, Dekegel D, Boeye A. 1983. Neutral-
ization of poliovirus by antibody-mediated
The inhibitory effects of antibodies on patho- polymerization. Virology 127:463468.
5. Thomas AA, Vrijsen R, Boeye A. 1986.
genic organisms have been documented
Relationship between poliovirus neutraliza-
since the late 1800s (271). Since that time, tion and aggregation. J Virol 59:479485.
much has been learned regarding the mech- 6. Lin TL, Clark TG, Dickerson H. 1996. Passive
anisms that underlie the antimicrobial activ- immunization of channel catfish (Ictalurus
ity of antibodies. However, antibodies often punctatus) against the ciliated protozoan
parasite Ichthyophthirius multifiliis by use of
have multiple functions in vitro and in vivo,
murine monoclonal antibodies. Infect Immun
either directly or through interactions with 64:40854090.
FcRs or complement. Modern tools, such as 7. Campodonico VL, Llosa NJ, Grout M, Doring
knockout mice or antibodies engineered to G, Maira-Litran T, Pier GB. 2010. Evaluation of
abrogate or enhance functions have proven flagella and flagellin of Pseudomonas aeruginosa
useful for more precise explorations of anti- as vaccines. Infect Immun 78:746755.
8. Bishop AL, Schild S, Patimalla B, Klein B,
body function. Nonetheless, major questions
Camilli A. 2010. Mucosal immunization with
regarding the way in which an antibody Vibrio cholerae outer membrane vesicles pro-
functions in vivo remain, and multiple ac- vides maternal protection mediated by anti-
tivities are likely to contribute to the anti- lipopolysaccharide antibodies that inhibit
microbial effect. bacterial motility. Infect Immun 78:44024420.
9. Hope T. 2010. Defining the interaction of HIV
with the mucosal barriers to gain insights into
ACKNOWLEDGMENTS the mechanisms of sexual transmission. Abstr.
AIDS Vaccine Conference 2010, Atlanta, GA.
I received financial support from the U.S. 10. McCullough KC, Smale CJ, Carpenter WC,
Public Health Service (grants AI084136, Crowther JR, Brocchi E, De Simone F. 1987.
AI0797775, and AI102715). Conformational alteration in foot-and-mouth
disease virus virion capsid structure after com-
I acknowledge the editorial assistance of
plexing with monospecific antibody. Immunol-
Gabriel J. A. Forthal. ogy 60:7582.
Conflicts of interest: I declare no 11. Hernandez R, Paredes A, Brown DT. 2008.
conflicts. Sindbis virus conformational changes induced
by a neutralizing anti-E1 monoclonal antibody.
J Virol 82:57505760.
CITATION 12. Klasse PJ, Sattentau QJ. 2002. Occupancy
and mechanism in antibody-mediated neutral-
Forthal DN. Functions of antibodies. 2014. ization of animal viruses. J Gen Virol 83:2091
Microbiol Spectrum 2(4):AID-0019-2014. 2108.
CHAPTER 2 Functions of Antibodies 37

13. LaRocca TJ, Holthausen DJ, Hsieh C, Renken human neutralizing antibody response against
C, Mannella CA, Benach JL. 2009. The bacte- dengue virus type 2. J Med Virol 45:451461.
ricidal effect of a complement-independent 23. Iorio RM, Glickman RL, Riel AM, Sheehan JP,
antibody is osmolytic and specific to Borrelia. Bratt MA. 1989. Functional and neutralization
Proc Natl Acad Sci USA 106:1075210757. profile of seven overlapping antigenic sites on
14. Matthews RC, Rigg G, Hodgetts S, Carter T, the HN glycoprotein of Newcastle disease virus:
Chapman C, Gregory C, Illidge C, Burnie J. monoclonal antibodies to some sites prevent
2003. Preclinical assessment of the efficacy viral attachment. Virus Res 13:245261.
of mycograb, a human recombinant antibody 24. Booy FP, Roden RB, Greenstone HL, Schiller
against fungal HSP90. Antimicrob Agents JT, Trus BL. 1998. Two antibodies that neu-
Chemother 47:22082216. tralize papillomavirus by different mechanisms
15. Pachl J, Svoboda P, Jacobs F, Vandewoude show distinct binding patterns at 13 A resolu-
K, van der Hoven B, Spronk P, Masterson G, tion. J Mol Biol 281:95106.
Malbrain M, Aoun M, Garbino J, Takala J, 25. Ruggeri FM, Greenberg HB. 1991. Antibodies
Drgona L, Burnie J, Matthews R. 2006. A to the trypsin cleavage peptide VP8 neutralize
randomized, blinded, multicenter trial of lipid- rotavirus by inhibiting binding of virions to
associated amphotericin B alone versus in target cells in culture. J Virol 65:22112219.
combination with an antibody-based inhibitor 26. Smith TJ, Olson NH, Cheng RH, Liu H, Chase
of heat shock protein 90 in patients with ES, Lee WM, Leippe DM, Mosser AG, Rueckert
invasive candidiasis. Clin Infect Dis 42:1404 RR, Baker TS. 1993. Structure of human rhino-
1413. virus complexed with Fab fragments from a
16. Nooney L, Matthews RC, Burnie JP. 2005. neutralizing antibody. J Virol 67:11481158.
Evaluation of Mycograb, amphotericin B, caspo- 27. Della-Porta AJ, Westaway EG. 1978. A multi-
fungin, and fluconazole in combination against hit model for the neutralization of animal
Cryptococcus neoformans by checkerboard and viruses. J Gen Virol 38:119.
time-kill methodologies. Diagn Microbiol Infect 28. Platt EJ, Gomes MM, Kabat D. 2012. Kinetic
Dis 51:1929. mechanism for HIV-1 neutralization by anti-
17. McClelland EE, Nicola AM, Prados-Rosales body 2G12 entails reversible glycan binding
R, Casadevall A. 2010. Ab binding alters gene that slows cell entry. Proc Natl Acad Sci USA
expression in Cryptococcus neoformans and 109:78297834.
directly modulates fungal metabolism. J Clin 29. Reading SA, Dimmock NJ. 2007. Neutralization
Invest 120:13551361. of animal virus infectivity by antibody. Arch Virol
18. Watanabe M, Blobel G. 1989. Site-specific 152:10471059.
antibodies against the PrlA (secY) protein of 30. Klemm P, Vejborg RM, Hancock V. 2010.
Escherichia coli inhibit protein export by Prevention of bacterial adhesion. Appl Microbiol
interfering with plasma membrane binding of Biotechnol 88:451459.
preproteins. Proc Natl Acad Sci USA 86:1895 31. Cegelski L, Marshall GR, Eldridge GR,
1899. Hultgren SJ. 2008. The biology and future
19. Gregory RL, Michalek SM, Shechmeister prospects of antivirulence therapies. Nat Rev
IL, McGhee JR. 1984. Function of anti- Microbiol 6:1727.
Streptococcus mutans antibodies: anti-ribosomal 32. Smani Y, McConnell MJ, Pachn J. 2012. Role
antibodies inhibit acid production, growth, and of fibronectin in the adhesion of Acinetobacter
glucose phosphotransferase activity. Infect Immun baumannii to host cells. PLoS One 7:e33073.
45:286289. doi:10.1371/journal.pone.0033073.
20. Casadevall A, Pirofski LA. 2012. Immuno- 33. Tsang TM, Annis DS, Kronshage M, Fenno
globulins in defense, pathogenesis, and therapy JT, Usselman LD, Mosher DF, Krukonis ES.
of fungal diseases. Cell Host Microbe 11:447 2012. Ail protein binds ninth type III fibronec-
456. tin repeat (9FNIII) within central 120-kDa
21. Klasse PJ, Sanders RW, Cerutti A, Moore JP. region of fibronectin to facilitate cell binding
2012. How can HIV-type-1-Env immunogenic- by Yersinia pestis. J Biol Chem 287:1675916767.
ity be improved to facilitate antibody-based 34. Khan MN, Pichichero ME. 2012. Vaccine
vaccine development? AIDS Res Hum Retro- candidates PhtD and PhtE of Streptococcus
viruses 28:115. pneumoniae are adhesins that elicit functional
22. He RT, Innis BL, Nisalak A, Usawattanakul antibodies in humans. Vaccine 30:29002907.
W, Wang S, Kalayanarooj S, Anderson R. 35. Langermann S, Ballou WR. 2003. Development of
1995. Antibodies that block virus attachment a recombinant FimCH vaccine for urinary tract
to Vero cells are a major component of the infections. Adv Exp Med Biol 539:635648.
38 FORTHAL

36. Langermann S, Palaszynski S, Barnhart M, 46. Yun CH, Lillehoj HS, Lillehoj EP. 2000.
Auguste G, Pinkner JS, Burlein J, Barren P, Intestinal immune responses to coccidiosis.
Koenig S, Leath S, Jones CH, Hultgren SJ. Dev Comp Immunol 24:303324.
1997. Prevention of mucosal Escherichia coli 47. Eckmann L. 2003. Mucosal defences against
infection by FimH-adhesin-based systemic Giardia. Parasite Immunol 25:259270.
vaccination. Science 276:607611. 48. Negrao-Correa D. 2001. Importance of immu-
37. Langermann S, Mollby R, Burlein JE, noglobulin E (IgE) in the protective mechanism
Palaszynski SR, Auguste CG, DeFusco A, against gastrointestinal nematode infection:
Strouse R, Schenerman MA, Hultgren SJ, looking at the intestinal mucosae. Rev Inst
Pinkner JS, Winberg J, Guldevall L, Soderhall Med Trop Sao Paulo 43:291299.
M, Ishikawa K, Normark S, Koenig S. 2000. 49. Cevallos AM, Zhang X, Waldor MK, Jaison S,
Vaccination with FimH adhesin protects cyno- Zhou X, Tzipori S, Neutra MR, Ward HD.
molgus monkeys from colonization and infection 2000. Molecular cloning and expression of a
by uropathogenic Escherichia coli. J Infect Dis gene encoding Cryptosporidium parvum glyco-
181:774778. proteins gp40 and gp15. Infect Immun 68:4108
38. Kain R, Exner M, Brandes R, Ziebermayr R, 4116.
Cunningham D, Alderson CA, Davidovits A, 50. Lopalco L, Barassi C, Pastori C, Longhi R,
Raab I, Jahn R, Ashour O, Spitzauer S, Burastero SE, Tambussi G, Mazzotta F,
Sunder-Plassmann G, Fukuda M, Klemm P, Lazzarin A, Clerici M, Siccardi AG. 2000.
Rees AJ, Kerjaschki D. 2008. Molecular CCR5-reactive antibodies in seronegative partners
mimicry in pauci-immune focal necrotizing of HIV-seropositive individuals down-modulate
glomerulonephritis. Nat Med 14:10881096. surface CCR5 in vivo and neutralize the infectivity
39. Manjarrez-Hernandez HA, Gavilanes-Parra S, of R5 strains of HIV-1 In vitro. J Immunol 164:
Chavez-Berrocal E, Navarro-Ocana A, Cravioto 34263433.
A. 2000. Antigen detection in enteropathogenic 51. Barassi C, Soprana E, Pastori C, Longhi R,
Escherichia coli using secretory immunoglobulin Buratti E, Lillo F, Marenzi C, Lazzarin A,
A antibodies isolated from human breast milk. Siccardi AG, Lopalco L. 2005. Induction of
Infect Immun 68:50305036. murine mucosal CCR5-reactive antibodies as
40. Schlesinger LS, Hull SR, Kaufman TM. 1994. an anti-human immunodeficiency virus strat-
Binding of the terminal mannosyl units of egy. J Virol 79:68486858.
lipoarabinomannan from a virulent strain of 52. Wintachai P, Wikan N, Kuadkitkan A,
Mycobacterium tuberculosis to human macro- Jaimipuk T, Ubol S, Pulmanausahakul R,
phages. J Immunol 152:40704079. Auewarakul P, Kasinrerk W, Weng WY,
41. Kornspan JD, Tarshis M, Rottem S. 2011. Panyasrivanit M, Paemanee A, Kittisenachai
Adhesion and biofilm formation of Mycoplasma S, Roytrakul S, Smith DR. 2012. Identification
pneumoniae on an abiotic surface. Arch Microbiol of prohibitin as a Chikungunya virus receptor
193:833836. protein. J Med Virol 84:17571770.
42. Holder AA, Freeman RR. 1981. Immunization 53. Kondratowicz AS, Lennemann NJ, Sinn PL,
against blood-stage rodent malaria using puri- Davey RA, Hunt CL, Moller-Tank S,
fied parasite antigens. Nature 294:361364. Meyerholz DK, Rennert P, Mullins RF,
43. Perkins ME, Rocco LJ. 1988. Sialic acid- Brindley M, Sandersfeld LM, Quinn K,
dependent binding of Plasmodium falciparum Weller M, McCray PB Jr, Chiorini J,
merozoite surface antigen, Pf200, to human Maury W. 2011. T-cell immunoglobulin and
erythrocytes. J Immunol 141:31903196. mucin domain 1 (TIM-1) is a receptor for Zaire
44. Bolad A, Berzins K. 2000. Antigenic diversity Ebolavirus and Lake Victoria Marburgvirus.
of Plasmodium falciparum and antibody-medi- Proc Natl Acad Sci USA 108:84268431.
ated parasite neutralization. Scand J Immunol 54. Bruno CJ, Jacobson JM. 2010. Ibalizumab: an
52:233239. anti-CD4 monoclonal antibody for the treatment of
45. Clausen TM, Christoffersen S, Dahlback M, HIV-1 infection. J Antimicrob Chemother 65:1839
Langkilde AE, Jensen KE, Resende M, Agerbaek 1841.
MO, Andersen D, Berisha B, Ditlev SB, Pinto 55. Meuleman P, Hesselgesser J, Paulson M,
VV, Nielsen MA, Theander TG, Larsen S, Vanwolleghem T, Desombere I, Reiser H,
Salanti A. 2012. Structural and functional insight Leroux-Roels G. 2008. Anti-CD81 antibodies
into how the Plasmodium falciparum VAR2CSA can prevent a hepatitis C virus infection in
protein mediates binding to chondroitin sulfate A vivo. Hepatology 48:17611768.
in placental malaria. J Biol Chem 287:23332 56. Silvie O, Rubinstein E, Franetich JF, Prenant M,
23345. Belnoue E, Renia L, Hannoun L, Eling W, Levy
CHAPTER 2 Functions of Antibodies 39

S, Boucheix C, Mazier D. 2003. Hepatocyte CD81 67. Stewart PL, Nemerow GR. 1997. Recent
is required for Plasmodium falciparum and Plas- structural solutions for antibody neutralization
modium yoelii sporozoite infectivity. Nat Med of viruses. Trends Microbiol 5:229233.
9:9396. 68. Wien MW, Filman DJ, Stura EA, Guillot S,
57. Schubert A, Zakikhany K, Pietrocola G, Meinke Delpeyroux F, Crainic R, Hogle JM. 1995.
A, Speziale P, Eikmanns BJ, Reinscheid DJ. Structure of the complex between the Fab
2004. The fibrinogen receptor FbsA promotes fragment of a neutralizing antibody for type 1
adherence of Streptococcus agalactiae to human poliovirus and its viral epitope. Nat Struct Biol
epithelial cells. Infect Immun 72:61976205. 2:232243.
58. Edwards MJ, Dimmock NJ. 2001. Hemagglu- 69. Maciejewski JP, Weichold FF, Young NS,
tinin 1-specific immunoglobulin G and Fab Cara A, Zella D, Reitz MS Jr, Gallo RC. 1995.
molecules mediate postattachment neutraliza- Intracellular expression of antibody fragments
tion of influenza A virus by inhibition of an directed against HIV reverse transcriptase
early fusion event. J Virol 75:1020810218. prevents HIV infection in vitro. Nat Med 1:
59. de Rosny E, Vassell R, Jiang S, Kunert R, 667673.
Weiss CD. 2004. Binding of the 2F5 monoclo- 70. Varghese R, Mikyas Y, Stewart PL, Ralston
nal antibody to native and fusion-intermediate R. 2004. Postentry neutralization of adenovi-
forms of human immunodeficiency virus type 1 rus type 5 by an antihexon antibody. J Virol
gp41: implications for fusion-inducing confor- 78:1232012332.
mational changes. J Virol 78:26272631. 71. Ishii Y, Tanaka K, Kondo K, Takeuchi T,
60. Lorizate M, Cruz A, Huarte N, Kunert R, Mori S, Kanda T. 2010. Inhibition of nuclear
Perez-Gil J, Nieva JL. 2006. Recognition and entry of HPV16 pseudovirus-packaged DNA
blocking of HIV-1 gp41 pre-transmembrane by an anti-HPV16 L2 neutralizing antibody.
sequence by monoclonal 4E10 antibody in a Virology 406:181188.
Raft-like membrane environment. J Biol Chem 72. Mazanec MB, Kaetzel CS, Lamm ME,
281:3959839606. Fletcher D, Nedrud JG. 1992. Intracellular
61. Kaufmann B, Nybakken GE, Chipman PR, neutralization of virus by immunoglobulin A
Zhang W, Diamond MS, Fremont DH, Kuhn antibodies. Proc Natl Acad Sci USA 89:6901
RJ, Rossmann MG. 2006. West Nile virus in 6905.
complex with the Fab fragment of a neutraliz- 73. Yan H, Lamm ME, Bjorling E, Huang YT.
ing monoclonal antibody. Proc Natl Acad Sci 2002. Multiple functions of immunoglobulin A
USA 103:1240012404. in mucosal defense against viruses: an in vitro
62. Pierson TC, Diamond MS. 2008. Molecular measles virus model. J Virol 76:1097210979.
mechanisms of antibody-mediated neutralisation 74. Mazanec MB, Coudret CL, Fletcher DR. 1995.
of flavivirus infection. Expert Rev Mol Med 10:e12. Intracellular neutralization of influenza virus
doi:10.1017/S1462399408000665. by immunoglobulin A anti-hemagglutinin mono-
63. Barbey-Martin C, Gigant B, Bizebard T, Calder clonal antibodies. J Virol 69:13391343.
LJ, Wharton SA, Skehel JJ, Knossow M. 2002. 75. Zhou D, Zhang Y, Li Q, Chen Y, He B, Yang J,
An antibody that prevents the hemagglutinin low Tu H, Lei L, Yan H. 2011. Matrix protein-specific
pH fusogenic transition. Virology 294:7074. IgA antibody inhibits measles virus replication by
64. Ekiert DC, Bhabha G, Elsliger MA, Friesen intracellular neutralization. J Virol 85:11090
RH, Jongeneelen M, Throsby M, Goudsmit 11097.
J, Wilson IA. 2009. Antibody recognition of 76. Bomsel M, Heyman M, Hocini H, Lagaye
a highly conserved influenza virus epitope. S, Belec L, Dupont C, Desgranges C. 1998.
Science 324:246251. Intracellular neutralization of HIV transcytosis
65. Cao Z, Meng J, Li X, Wu R, Huang Y, He Y. across tight epithelial barriers by anti-HIV
2012. The epitope and neutralization mecha- envelope protein dIgA or IgM. Immunity 9:
nism of AVFluIgG01, a broad-reactive human 277287.
monoclonal antibody against H5N1 influenza 77. Corthesy B, Benureau Y, Perrier C, Fourgeux
virus. PLoS One 7:e38126. doi:10.1371/journal. C, Parez N, Greenberg H, Schwartz-Cornil I.
pone.0038126. 2006. Rotavirus anti-VP6 secretory immuno-
66. Bai Y, Ye L, Tesar DB, Song H, Zhao D, globulin A contributes to protection via intra-
Bjorkman PJ, Roopenian DC, Zhu X. 2011. cellular neutralization but not via immune
Intracellular neutralization of viral infection in exclusion. J Virol 80:1069210699.
polarized epithelial cells by neonatal Fc recep- 78. Feng N, Lawton JA, Gilbert J, Kuklin N, Vo
tor (FcRn)-mediated IgG transport. Proc Natl P, Prasad BV, Greenberg HB. 2002. Inhibition
Acad Sci USA 108:1840618411. of rotavirus replication by a non-neutralizing,
40 FORTHAL

rotavirus VP6-specific IgA mAb. J Clin Invest 92. Diamond MS, Shrestha B, Mehlhop E, Sitati
109:12031213. E, Engle M. 2003. Innate and adaptive immune
79. Thouvenin E, Schoehn G, Rey F, Petitpas I, responses determine protection against dis-
Mathieu M, Vaney MC, Cohen J, Kohli E, seminated infection by West Nile encephalitis
Pothier P, Hewat E. 2001. Antibody inhibi- virus. Viral Immunol 16:259278.
tion of the transcriptase activity of the rota- 93. Vogt MR, Dowd KA, Engle M, Tesh RB,
virus DLP: a structural view. J Mol Biol 307:161 Johnson S, Pierson TC, Diamond MS. 2011.
172. Poorly neutralizing cross-reactive antibodies
80. Mallery DL, McEwan WA, Bidgood SR, Towers against the fusion loop of West Nile virus
GJ, Johnson CM, James LC. 2010. Antibodies envelope protein protect in vivo via Fcgamma
mediate intracellular immunity through tripartite receptor and complement-dependent effector
motif-containing 21 (TRIM21). Proc Natl Acad Sci mechanisms. J Virol 85:1156711580.
USA 107:1998519990. 94. Kruijsen D, Bakkers MJ, van Uden NO,
81. Wang X, Kikuchi T, Rikihisa Y. 2006. Two Viveen MC, van der Sluis TC, Kimpen JL,
monoclonal antibodies with defined epitopes Leusen JH, Coenjaerts FE, van Bleek GM.
of P44 major surface proteins neutralize Ana- 2010. Serum antibodies critically affect virus-
plasma phagocytophilum by distinct mecha- specific CD4+/CD8+ T cell balance during res-
nisms. Infect Immun 74:18731882. piratory syncytial virus infections. J Immunol
82. Edelson BT, Unanue ER. 2001. Intracellular 185:64896498.
antibody neutralizes Listeria growth. Immunity 95. Terajima M, Cruz J, Co MD, Lee JH, Kaur K,
14:503512. Wrammert J, Wilson PC, Ennis FA. 2011.
83. Bout D, Moretto M, Dimier-Poisson I, Gatel Complement-dependent lysis of influenza a
DB. 1999. Interaction between Toxoplasma virus-infected cells by broadly cross-reactive
gondii and enterocyte. Immunobiology 201: human monoclonal antibodies. J Virol 85:
225228. 1346313467.
84. Mineo JR, Khan IA, Kasper LH. 1994. Toxo- 96. Frank AL, Puck J, Hughes BJ, Cate TR. 1980.
plasma gondii: a monoclonal antibody that Microneutralization test for influenza A and B
inhibits intracellular replication. Exp Parasitol and parainfluenza 1 and 2 viruses that uses
79:351361. continuous cell lines and fresh serum enhance-
85. Webster RG, Laver WG. 1967. Preparation ment. J Clin Microbiol 12:426432.
and properties of antibody directed specifically 97. Linscott WD, Levinson WE. 1969. Comple-
against the neuraminidase of influenza virus. ment components required for virus neutrali-
J Immunol 99:4955. zation by early immunoglobulin antibody. Proc
86. Hughey PG, Roberts PC, Holsinger LJ, Natl Acad Sci USA 64:520527.
Zebedee SL, Lamb RA, Compans RW. 1995. 98. Yoshino K, Taniguchi S. 1965. Studies on the
Effects of antibody to the influenza A virus M2 neutralization of herpes simplex virus. I.
protein on M2 surface expression and virus Appearance of neutralizing antibodies having
assembly. Virology 212:411421. different grades of complement requirement.
87. Corboba P, Grutadauria S, Cuffini C, Zapata Virology 26:4453.
M. 2000. Neutralizing monoclonal antibody to 99. Ozaki Y, Tabeyi K. 1967. Studies on the
the E1 glycoprotein epitope of rubella virus neutralization of Japanese encephalitis virus.
mediates virus arrest in VERO cells. Viral I. Application of kinetic neutralization to the
Immunol 13:8392. measurement of the neutralizing potency of
88. Murphy K, Travers P, Walport M. 2008. antiserum. J Immunol 98:12181223.
The complement system and innate immunity, 100. Johnson JB, Capraro GA, Parks GD. 2008.
p 6180. Janeways Immunobiology, 7th ed. Differential mechanisms of complement-
Garland Science, New York, NY. mediated neutralization of the closely related
89. DiScipio RG, Schraufstatter IU. 2007. The paramyxoviruses simian virus 5 and mumps
role of the complement anaphylatoxins in virus. Virology 376:112123.
the recruitment of eosinophils. Int Immuno- 101. Corbeil S, Seguin C, Trudel M. 1996. Involve-
pharmacol 7:19091923. ment of the complement system in the protec-
90. Walport MJ. 2001. Complement. First of two tion of mice from challenge with respiratory
parts. N Engl J Med 344:10581066. syncytial virus Long strain following passive
91. Prodinger WM, Wurzner R, Stoiber H, Dierich immunization with monoclonal antibody 18A2B2.
MP. 2003. Complement, p 10771103. In Paul W Vaccine 14:521525.
(ed), Fundamental Immunology, 5th ed. 102. Boere WA, Benaissa-Trouw BJ, Harmsen T,
Lippincott Williams & Wilkins, Philadelphia, PA. Erich T, Kraaijeveld CA, Snippe H. 1986. The
CHAPTER 2 Functions of Antibodies 41

role of complement in monoclonal antibody- 114. Ram S, Lewis LA, Rice PA. 2010. Infections of
mediated protection against virulent Semliki people with complement deficiencies and
Forest virus. Immunology 58:553559. patients who have undergone splenectomy.
103. Saifuddin M, Parker CJ, Peeples ME, Gorny Clin Microbiol Rev 23:740780.
MK, Zolla-Pazner S, Ghassemi M, Rooney IA, 115. Amir J, Scott MG, Nahm MH, Granoff DM.
Atkinson JP, Spear GT. 1995. Role of virion- 1990. Bactericidal and opsonic activity of IgG1
associated glycosylphosphatidylinositol-linked and IgG2 anticapsular antibodies to Haemo-
proteins CD55 and CD59 in complement resis- philus influenzae type b. J Infect Dis 162:163
tance of cell line-derived and primary isolates 171.
of HIV-1. J Exp Med 182:501509. 116. Frasch CE, Borrow R, Donnelly J. 2009.
104. Schmitz J, Zimmer JP, Kluxen B, Aries S, Bactericidal antibody is the immunologic
Bogel M, Gigli I, Schmitz H. 1995. Antibody- surrogate of protection against meningococcal
dependent complement-mediated cytotoxicity disease. Vaccine 27(Suppl 2):B112B116.
in sera from patients with HIV-1 infection is 117. Welsch JA, Moe GR, Rossi R, Adu-Bobie J,
controlled by CD55 and CD59. J Clin Invest Rappuoli R, Granoff DM. 2003. Antibody to
96:15201526. genome-derived neisserial antigen 2132, a Neis-
105. Zhou ZH, Wild T, Xiong Y, Sylvers P, Zhang seria meningitidis candidate vaccine, confers
Y, Zhang L, Wahl L, Wahl SM, Kozlowski S, protection against bacteremia in the absence
Notkins AL. 2013. Polyreactive Antibodies of complement-mediated bactericidal activity.
Plus Complement Enhance the Phagocytosis J Infect Dis 188:17301740.
of Cells Made Apoptotic by UV-Light or HIV. 118. Plested JS, Welsch JA, Granoff DM. 2009. Ex
Sci Rep 3:2271. doi:10.1038/srep02271. vivo model of meningococcal bacteremia using
106. Willey S, Aasa-Chapman MM, OFarrell S, human blood for measuring vaccine-induced
Pellegrino P, Williams I, Weiss RA, Neil SJ. serum passive protective activity. Clin Vaccine
2011. Extensive complement-dependent enhance- Immunol 16:785791.
ment of HIV-1 by autologous non-neutralising 119. Granoff DM. 2009. Relative importance of
antibodies at early stages of infection. Retro- complement-mediated bactericidal and opsonic
virology 8:16. doi:10.1186/1742-4690-8-16. activity for protection against meningococcal
107. Stoiber H, Banki Z, Wilflingseder D, Dierich disease. Vaccine 27(Suppl 2):B117B125.
MP. 2008. Complement-HIV interactions during 120. Horwitz MA, Silverstein SC. 1981. Interaction of
all steps of viral pathogenesis. Vaccine 26:3046 the Legionnaires disease bacterium (Legionella
3054. pneumophila) with human phagocytes. I. L.
108. Huber G, Banki Z, Lengauer S, Stoiber H. pneumophila resists killing by polymorphonuclear
2011. Emerging role for complement in HIV leukocytes, antibody, and complement. J Exp Med
infection. Curr Opin HIV AIDS 6:419426. 153:386397.
109. Jayasekera JP, Moseman EA, Carroll MC. 2007. 121. Lindow JC, Fimlaid KA, Bunn JY, Kirkpatrick
Natural antibody and complement mediate neu- BD. 2011. Antibodies in action: role of human
tralization of influenza virus in the absence of prior opsonins in killing Salmonella enterica serovar
immunity. J Virol 81:34873494. Typhi. Infect Immun 79:31883194.
110. Carroll MC. 2004. The complement system in 122. Cheng SC, Sprong T, Joosten LA, van der
regulation of adaptive immunity. Nat Immunol Meer JW, Kullberg BJ, Hube B, Schejbel L,
5:981986. Garred P, van Deuren M, Netea MG. 2012.
111. Stager S, Alexander J, Kirby AC, Botto M, Complement plays a central role in Candida
Rooijen NV, Smith DF, Brombacher F, Kaye albicans-induced cytokine production by
PM. 2003. Natural antibodies and complement human PBMCs. Eur J Immunol 42:9931004.
are endogenous adjuvants for vaccine-induced 123. Han Y, Kozel TR, Zhang MX, MacGill RS,
CD8+ T-cell responses. Nat Med 9:12871292. Carroll MC, Cutler JE. 2001. Complement is
112. Lindorfer MA, Hahn CS, Foley PL, Taylor essential for protection by an IgM and an IgG3
RP. 2001. Heteropolymer-mediated clearance monoclonal antibody against experimental,
of immune complexes via erythrocyte CR1: hematogenously disseminated candidiasis. J
mechanisms and applications. Immunol Rev Immunol 167:15501557.
183:1024. 124. Zaragoza O, Casadevall A. 2006. Monoclonal
113. Nelson RA Jr. 1953. The immune-adherence antibodies can affect complement deposition
phenomenon; an immunologically specific re- on the capsule of the pathogenic fungus
action between microorganisms and eryth- Cryptococcus neoformans by both classical
rocytes leading to enhanced phagocytosis. pathway activation and steric hindrance. Cell
Science 118:733737. Microbiol 8:18621876.
42 FORTHAL

125. Zhong Z, Pirofski LA. 1998. Antifungal activity bodies against multiple, non-variant target
of a human antiglucuronoxylomannan anti- epitopes of the Plasmodium falciparum gamete
body. Clin Diagn Lab Immunol 5:5864. surface antigen Pfs230 are all complement-
126. Taborda CP, Casadevall A. 2002. CR3 fixing. Parasite Immunol 16:511519.
(CD11b/CD18) and CR4 (CD11c/CD18) are 136. Rener J, Graves PM, Carter R, Williams JL,
involved in complement-independent anti- Burkot TR. 1983. Target antigens of transmis-
body-mediated phagocytosis of Cryptococcus sion-blocking immunity on gametes of Plas-
neoformans. Immunity 16:791802. modium falciparum. J Exp Med 158:976981.
127. Ayi K, Turrini F, Piga A, Arese P. 2004. 137. Roeffen W, Geeraedts F, Eling W, Beckers P,
Enhanced phagocytosis of ring-parasitized Wizel B, Kumar N, Lensen T, Sauerwein R.
mutant erythrocytes: a common mechanism 1995. Transmission blockade of Plasmodium
that may explain protection against falciparum falciparum malaria by anti-Pfs230-specific
malaria in sickle trait and beta-thalassemia antibodies is isotype dependent. Infect Immun
trait. Blood 104:33643371. 63:467471.
128. Luzzi GA, Merry AH, Newbold CI, Marsh K, 138. Macaskill JA, Holmes PH, Whitelaw DD,
Pasvol G. 1991. Protection by alpha-thalassae- McConnell I, Jennings FW, Urquhart
mia against Plasmodium falciparum malaria: GM. 1980. Immunological clearance of 75Se-
modified surface antigen expression rather labelled Trypanosoma brucei in mice. II.
than impaired growth or cytoadherence. Mechanisms in immune animals. Immunology
Immunol Lett 30:233240. 40:629635.
129. Yuthavong Y, Bunyaratvej A, Kamchon- 139. Pan W, Ogunremi O, Wei G, Shi M, Tabel H.
wongpaisan S. 1990. Increased susceptibility of 2006. CR3 (CD11b/CD18) is the major macro-
malaria-infected variant erythrocytes to the phage receptor for IgM antibody-mediated
mononuclear phagocyte system. Blood Cells phagocytosis of African trypanosomes: diverse
16:591597. effect on subsequent synthesis of tumor ne-
130. Cappadoro M, Giribaldi G, OBrien E, crosis factor alpha and nitric oxide. Microbes
Turrini F, Mannu F, Ulliers D, Simula G, Infect 8:12091218.
Luzzatto L, Arese P. 1998. Early phagocy- 140. Owuor BO, Odhiambo CO, Otieno WO,
tosis of glucose-6-phosphate dehydrogenase Adhiambo C, Makawiti DW, Stoute JA. 2008.
(G6PD)-deficient erythrocytes parasitized by Reduced immune complex binding capacity and
Plasmodium falciparum may explain malaria increased complement susceptibility of red cells
protection in G6PD deficiency. Blood 92:2527 from children with severe malaria-associated
2534. anemia. Mol Med 14:8997.
131. Kumaratilake LM, Ferrante A, Jaeger T, 141. Patel SN, Berghout J, Lovegrove FE, Ayi K,
Morris-Jones SD. 1997. The role of comple- Conroy A, Serghides L, Min-oo G, Gowda
ment, antibody, and tumor necrosis factor alpha DC, Sarma JV, Rittirsch D, Ward PA, Liles
in the killing of Plasmodium falciparum by the WC, Gros P, Kain KC. 2008. C5 deficiency and
monocytic cell line THP-1. Infect Immun 65: C5a or C5aR blockade protects against cerebral
53425345. malaria. J Exp Med 205:11331143.
132. Salmon D, Vilde JL, Andrieu B, Simonovic R, 142. Farrell HE, Shellam GR. 1991. Protection
Lebras J. 1986. Role of immune serum and against murine cytomegalovirus infection by
complement in stimulation of the metabolic passive transfer of neutralizing and non-
burst of human neutrophils by Plasmodium neutralizing monoclonal antibodies. J Gen Virol
falciparum. Infect Immun 51:801806. 72(Pt 1):149156.
133. Pang XL, Horii T. 1998. Complement-mediated 143. Horton RE, Vidarsson G. 2013. Antibodies and
killing of Plasmodium falciparum erythrocytic their receptors: different potential roles in
schizont with antibodies to the recombinant mucosal defense. Front Immunol 4:200.
serine repeat antigen (SERA). Vaccine 16:1299 doi:10.3389/fimmu.2013.00200.
1305. 144. Takai T. 2002. Roles of Fc receptors in auto-
134. Healer J, McGuinness D, Hopcroft P, Haley S, immunity. Nat Rev Immunol 2:580592.
Carter R, Riley E. 1997. Complement-mediated 145. Clark MR, Clarkson SB, Ory PA, Stollman N,
lysis of Plasmodium falciparum gametes by Goldstein IM. 1989. Molecular basis for a poly-
malaria-immune human sera is associated with morphism involving Fc receptor II on human
antibodies to the gamete surface antigen Pfs230. monocytes. J Immunol 143:17311734.
Infect Immun 65:30173023. 146. Warmerdam PA, van de Winkel JG, Vlug A,
135. Read D, Lensen AH, Begarnie S, Haley S, Raza Westerdaal NA, Capel PJ. 1991. A single amino
A, Carter R. 1994. Transmission-blocking anti- acid in the second Ig-like domain of the human
CHAPTER 2 Functions of Antibodies 43

Fc gamma receptor II is critical for human IgG2 157. Lyerly HK, Reed DL, Matthews TJ, Langlois
binding. J Immunol 147:13381343. AJ, Ahearne PA, Petteway SR Jr, Weinhold
147. Ravetch JV, Perussia B. 1989. Alternative KJ. 1987. Anti-GP 120 antibodies from HIV
membrane forms of Fc gamma RIII(CD16) on seropositive individuals mediate broadly reac-
human natural killer cells and neutrophils. Cell tive anti-HIV ADCC. AIDS Res Hum Retro-
type-specific expression of two genes that viruses 3:409422.
differ in single nucleotide substitutions. J Exp 158. Torben W, Ahmad G, Zhang W, Nash S, Le L,
Med 170:481497. Karmakar S, Siddiqui AA. 2012. Role of anti-
148. Ory PA, Goldstein IM, Kwoh EE, Clarkson body dependent cell mediated cytotoxicity
SB. 1989. Characterization of polymorphic (ADCC) in Sm-p80-mediated protection against
forms of Fc receptor III on human neutrophils. Schistosoma mansoni. Vaccine 30:67536758.
J Clin Invest 83:16761681. 159. Bouharoun-Tayoun H, Oeuvray C, Lunel F,
149. Ory PA, Clark MR, Kwoh EE, Clarkson SB, Druilhe P. 1995. Mechanisms underlying the
Goldstein IM. 1989. Sequences of complemen- monocyte-mediated antibody-dependent kill-
tary DNAs that encode the NA1 and NA2 forms ing of Plasmodium falciparum asexual blood
of Fc receptor III on human neutrophils. J Clin stages. J Exp Med 182:409418.
Invest 84:16881691. 160. Forthal DN, Landucci G, Daar ES. 2001.
150. Bruhns P, Iannascoli B, England P, Mancardi Antibody from patients with acute human
DA, Fernandez N, Jorieux S, Daeron M. 2009. immunodeficiency virus (HIV) infection inhib-
Specificity and affinity of human Fcgamma recep- its primary strains of HIV type 1 in the presence
tors and their polymorphic variants for human IgG of natural-killer effector cells. J Virol 75:6953
subclasses. Blood 113:37163725. 6961.
151. Wiener E, Jolliffe VM, Scott HC, Kumpel BM, 161. Weber S, Tian H, van Rooijen N, Pirofski LA.
Thompson KM, Melamed MD, Hughes-Jones 2012. A serotype 3 pneumococcal capsular
NC. 1988. Differences between the activities of polysaccharide-specific monoclonal antibody
human monoclonal IgG1 and IgG3 anti-D requires Fcgamma receptor III and macro-
antibodies of the Rh blood group system in phages to mediate protection against pneumo-
their abilities to mediate effector functions of coccal pneumonia in mice. Infect Immun 80:
monocytes. Immunology 65:159163. 13141322.
152. Shields RL, Lai J, Keck R, OConnell LY, 162. Sun D, Raisley B, Langer M, Iyer JK,
Hong K, Meng YG, Weikert SH, Presta LG. Vedham V, Ballard JL, James JA, Metcalf
2002. Lack of fucose on human IgG1 N-linked J, Coggeshall KM. 2012. Anti-peptidoglycan
oligosaccharide improves binding to human antibodies and Fcgamma receptors are the key
Fcgamma RIII and antibody-dependent cellu- mediators of inflammation in Gram-positive
lar toxicity. J Biol Chem 277:2673326740. sepsis. J Immunol 189:24232431.
153. Forthal DN, Gach JS, Landucci G, Jez J, Strasser 163. Song X, Tanaka S, Cox D, Lee SC. 2004. Fcgamma
R, Kunert R, Steinkellner H. 2010. Fc-glycosyla- receptor signaling in primary human microglia:
tion influences Fcgamma receptor binding and differential roles of PI-3K and Ras/ERK MAPK
cell-mediated anti-HIV activity of monoclonal pathways in phagocytosis and chemokine induc-
antibody 2G12. J Immunol 185:68766882. tion. J Leukoc Biol 75:11471155.
154. Lux A, Nimmerjahn F. 2011. Impact of differ- 164. Porcherie A, Mathieu C, Peronet R, Schneider
ential glycosylation on IgG activity. Adv Exp E, Claver J, Commere PH, Kiefer-Biasizzo H,
Med Biol 780:113124. Karasuyama H, Milon G, Dy M, Kinet JP,
155. Moldt B, Shibata-Koyama M, Rakasz EG, Louis J, Blank U, Mecheri S. 2011. Critical role
Schultz N, Kanda Y, Dunlop DC, Finstad SL, of the neutrophil-associated high-affinity re-
Jin C, Landucci G, Alpert MD, Dugast AS, ceptor for IgE in the pathogenesis of experi-
Parren PW, Nimmerjahn F, Evans DT, Alter G, mental cerebral malaria. J Exp Med 208:2225
Forthal DN, Schmitz JE, Iida S, Poignard P, 2236.
Watkins DI, Hessell AJ, Burton DR. 2012. A 165. Forthal DN, Landucci G, Phan TB, Becerra J.
nonfucosylated variant of the anti-HIV-1 mono- 2005. Interactions between natural killer cells
clonal antibody b12 has enhanced FcgammaRIIIa- and antibody Fc result in enhanced antibody
mediated antiviral activity in vitro but does not neutralization of human immunodeficiency
improve protection against mucosal SHIV chal- virus type 1. J Virol 79:20422049.
lenge in macaques. J Virol 86:61896196. 166. Brown BK, Wieczorek L, Kijak G, Lombardi
156. Forthal DN, Moog C. 2009. Fc receptor- K, Currier J, Wesberry M, Kappes JC, Ngauy
mediated antiviral antibodies. Curr Opin HIV V, Marovich M, Michael N, Ochsenbauer C,
AIDS 4:388393. Montefiori DC, Polonis VR. 2012. The role of
44 FORTHAL

natural killer (NK) cells and NK cell receptor 177. Forthal D, Hope TJ, Alter G. 2013. New
polymorphisms in the assessment of HIV-1 paradigms for functional HIV-specific non-
neutralization. PLoS One 7:e29454. doi:10.1371/ neutralizing antibodies. Curr Opin HIV AIDS
journal.pone.0029454. 8:392400.
167. Holl V, Peressin M, Decoville T, Schmidt S, 178. Lowell GH, Smith LF, Artenstein MS, Nash
Zolla-Pazner S, Aubertin AM, Moog C. 2006. GS, MacDermott RP Jr. 1979. Antibody-
Nonneutralizing antibodies are able to inhibit dependent cell-mediated antibacterial activity
human immunodeficiency virus type 1 replica- of human mononuclear cells. I. K lymphocytes
tion in macrophages and immature dendritic and monocytes are effective against menin-
cells. J Virol 80:61776181. gococi in cooperation with human imune sera.
168. Holl V, Hemmerter S, Burrer R, Schmidt S, J Exp Med 150:127137.
Bohbot A, Aubertin AM, Moog C. 2004. 179. Lowell GH, MacDermott RP, Summers PL,
Involvement of Fc gamma RI (CD64) in the Reeder AA, Bertovich MJ, Formal SB. 1980.
mechanism of HIV-1 inhibition by polyclonal Antibody-dependent cell-mediated antibacte-
IgG purified from infected patients in cultured rial activity: K lymphocytes, monocytes, and
monocyte-derived macrophages. J Immunol 173: granulocytes are effective against shigella. J
62746283. Immunol 125:27782784.
169. Anderson DR, Grillo-Lopez A, Varns C, 180. Tagliabue A, Nencioni L, Villa L, Keren DF,
Chambers KS, Hanna N. 1997. Targeted anti- Lowell GH, Boraschi D. 1983. Antibody-
cancer therapy using rituximab, a chimaeric anti- dependent cell-mediated antibacterial activity
CD20 antibody (IDEC-C2B8) in the treatment of of intestinal lymphocytes with secretory IgA.
non-Hodgkins B-cell lymphoma. Biochem Soc Nature 306:184186.
Trans 25:705708. 181. Tagliabue A, Boraschi D, Villa L, Keren DF,
170. Sliwkowski MX, Lofgren JA, Lewis GD, Lowell GH, Rappuoli R, Nencioni L. 1984.
Hotaling TE, Fendly BM, Fox JA. 1999. IgA-dependent cell-mediated activity against
Nonclinical studies addressing the mechanism enteropathogenic bacteria: distribution, speci-
of action of trastuzumab (Herceptin). Semin ficity, and characterization of the effector cells.
Oncol 26:6070. J Immunol 133:988992.
171. Clynes RA, Towers TL, Presta LG, Ravetch 182. Sestini P, Nencioni L, Villa L, Boraschi D,
JV. 2000. Inhibitory Fc receptors modulate Tagliabue A. 1988. IgA-driven antibacterial
in vivo cytotoxicity against tumor targets. Nat activity against Streptococcus pneumoniae by
Med 6:443446. mouse lung lymphocytes. Am Rev Respir Dis
172. Shore SL, Nahmias AJ, Starr SE, Wood 137:138143.
PA, McFarlin DE. 1974. Detection of cell- 183. Messick JB, Rikihisa Y. 1992. Presence of
dependent cytotoxic antibody to cells infected parasite antigen on the surface of P388D1 cells
with herpes simplex virus. Nature 251:350352. infected with Ehrlichia risticii. Infect Immun
173. Balachandran N, Bacchetti S, Rawls WE. 1982. 60:30793086.
Protection against lethal challenge of BALB/c mice 184. Koster FT, Kirkpatrick TL, Rowatt JD, Baca
by passive transfer of monoclonal antibodies to five OG. 1984. Antibody-dependent cellular cyto-
glycoproteins of herpes simplex virus type 2. Infect toxicity of Coxiella burnetii-infected J774
Immun 37:11321137. macrophage target cells. Infect Immun 43:
174. Gorander S, Harandi AM, Lindqvist M, 253256.
Bergstrom T, Liljeqvist JA. 2012. Glycoprotein 185. Galdiero F, Romano Carratelli C, Nuzzo I,
G of herpes simplex virus 2 as a novel vaccine Folgore A. 1985. Cytotoxic antibody depen-
antigen for immunity to genital and neurolog- dent cells in mice experimentally infected with
ical disease. J Virol 86:75447553. Brucella abortus. Microbiologica 8:217224.
175. Chu CF, Meador MG, Young CG, Strasser JE, 186. Shannon JG, Cockrell DC, Takahashi K,
Bourne N, Milligan GN. 2008. Antibody- Stahl GL, Heinzen RA. 2009. Antibody-
mediated protection against genital herpes mediated immunity to the obligate intracellular
simplex virus type 2 disease in mice by Fc bacterial pathogen Coxiella burnetii is Fc
gamma receptor-dependent and -independent receptor- and complement-independent. BMC
mechanisms. J Reprod Immunol 78:5867. Immunol 10:26. doi:10.1186/1471-2172-10-26.
176. Jegerlehner A, Schmitz N, Storni T, 187. Kazura JW. 1981. Host defense mechanisms
Bachmann MF. 2004. Influenza A vaccine against nematode parasites: destruction of
based on the extracellular domain of M2: weak newborn Trichinella spiralis larvae by human
protection mediated via antibody-dependent NK antibodies and granulocytes. J Infect Dis 143:
cell activity. J Immunol 172:55985605. 712718.
CHAPTER 2 Functions of Antibodies 45

188. Venturiello SM, Giambartolomei GH, Costantino serum dependent cell-mediated destruction of
SN. 1993. Immune killing of newborn Trichinella infective larvae in vitro. Trans R Soc Trop Med
larvae by human leucocytes. Parasite Immunol Hyg 76:362370.
15:559564. 202. Parab PB, Rajasekariah GR, Chandrashekar
189. Gounni AS, Lamkhioued B, Ochiai K, Tanaka Y, R, Alkan SS, Braun DG, Subrahmanyam D.
Delaporte E, Capron A, Kinet JP, Capron M. 1988. Characterization of a monoclonal anti-
1994. High-affinity IgE receptor on eosinophils is body against infective larvae of Brugia malayi.
involved in defence against parasites. Nature Immunology 64:169174.
367:183186. 203. Gray CA, Lawrence RA. 2002. A role for antibody
190. Capron M, Capron A. 1994. Immunoglobulin and Fc receptor in the clearance of Brugia malayi
E and effector cells in schistosomiasis. Science microfilariae. Eur J Immunol 32:11141120.
264:18761877. 204. Albright JW, Stewart MJ, Latham PS,
191. Zhou S, Liu S, Song G, Xu Y, Sun W. 2000. Albright JF. 1994. Antibody-facilitated mac-
Protective immunity induced by the full-length rophage killing of Trypanosoma musculi is
cDNA encoding paramyosin of Chinese Schisto- an extracellular process as studied in several
soma japonicum. Vaccine 18:31963204. variations of an in vitro analytical system.
192. Capron A. 1998. Schistosomiasis: forty years J Leukoc Biol 56:636643.
war on the worm. Parasitol Today 14:379384. 205. Townsend J, Duffus WP. 1982. Trypanosoma
193. Joseph M, Auriault C, Capron A, Vorng H, theileri: antibody-dependent killing by purified
Viens P. 1983. A new function for platelets: populations of bovine leucocytes. Clin Exp
IgE-dependent killing of schistosomes. Nature Immunol 48:289299.
303:810812. 206. Kierszenbaum F, Hayes MM. 1980. Mecha-
194. Khalife J, Capron M, Capron A, Grzych JM, nisms of resistance against experimental Trypano-
Butterworth AE, Dunne DW, Ouma JH. 1986. soma cruzi infection. Requirements for cellular
Immunity in human schistosomiasis mansoni. destruction of circulating forms of T. cruzi in
Regulation of protective immune mechanisms by human and murine in vitro systems. Immunology
IgM blocking antibodies. J Exp Med 164:16261640. 40:6166.
195. Auriault C, Gras-Masse H, Pierce RJ, 207. Piedrafita D, Parsons JC, Sandeman RM,
Butterworth AE, Wolowczuk I, Capron M, Wood PR, Estuningsih SE, Partoutomo S,
Ouma JH, Balloul JM, Khalife J, Neyrinck JL. Spithill TW. 2001. Antibody-dependent cell-
1990. Antibody response of Schistosoma mansoni- mediated cytotoxicity to newly excysted juve-
infected human subjects to the recombinant P28 nile Fasciola hepatica in vitro is mediated by
glutathione-S-transferase and to synthetic peptides. reactive nitrogen intermediates. Parasite Immunol
J Clin Microbiol 28:19181924. 23:473482.
196. Demeure CE, Rihet P, Abel L, Ouattara M, 208. Nolan TJ, Rotman HL, Bhopale VM, Schad
Bourgois A, Dessein AJ. 1993. Resistance to GA, Abraham D. 1995. Immunity to a chal-
Schistosoma mansoni in humans: influence of lenge infection of Strongyloides stercoralis
the IgE/IgG4 balance and IgG2 in immunity third-stage larvae in the jird. Parasite Immunol
to reinfection after chemotherapy. J Infect Dis 17:599604.
168:10001008. 209. Bekhti K, Kazanji M, Pery P. 1992. In vitro
197. Lawrence RA. 2001. Immunity to filarial nema- interactions between murine neutrophils and
todes. Vet Parasitol 100:3344. Eimeria falciformis sporozoites. Res Immunol
198. Haque A, Joseph M, Ouaissi MA, Capron 143:909917.
M, Capron A. 1980. IgE antibody-mediated 210. Smith PD, Keister DB, Elson CO. 1983. Human
cytotoxicity of rat macrophages against micro- host response to Giardia lamblia. II. Antibody-
filaria of Dipetalonema citeae in vitro. Clin Exp dependent killing in vitro. Cell Immunol 82:308
Immunol 40:487495. 315.
199. Weiss N, Tanner M. 1979. Studies on Dipetalo- 211. Khusmith S, Druilhe P. 1983. Cooperation
nema viteae (Filarioidea) 3. Antibody-dependent between antibodies and monocytes that inhibit
cell-mediated destruction of microfilariae in vivo. in vitro proliferation of Plasmodium falciparum.
Tropenmed Parasitol 30:7380. Infect Immun 41:219223.
200. Mehta K, Sindhu RK, Subrahmanyam D, 212. Jafarshad A, Dziegiel MH, Lundquist R,
Hopper K, Nelson DS, Rao CK. 1981. Antibody- Nielsen LK, Singh S, Druilhe PL. 2007. A novel
dependent cell-mediated effects in bancroftian antibody-dependent cellular cytotoxicity mecha-
filariasis. Immunology 43:117123. nism involved in defense against malaria requires
201. Sim BK, Kwa BH, Mak JW. 1982. Immune costimulation of monocytes FcgammaRII and
responses in human Brugia malayi infections: FcgammaRIII. J Immunol 178:30993106.
46 FORTHAL

213. Forthal DN, Landucci G. 1998. In vitro reduc- immune complexes formed with influenza virus.
tion of virus infectivity by antibody-dependent Blood 82:16391646.
cell-mediated immunity. J Immunol Methods 225. Scott CB, Ratcliffe DR, Cramer EB. 1996.
220:129138. Human monocytes are unable to bind to or
214. Forthal DN, Landucci G, Cole KS, Marthas phagocytize IgA and IgG immune complexes
M, Becerra JC, Van Rompay K. 2006. Rhesus formed with influenza virus in vitro. J Immunol
macaque polyclonal and monoclonal anti- 157:351359.
bodies inhibit simian immunodeficiency virus 226. Hellwig SM, van Oirschot HF, Hazenbos
in the presence of human or autologous rhesus WL, van Spriel AB, Mooi FR, van De Winkel
effector cells. J Virol 80:92179225. JG. 2001. Targeting to Fcgamma receptors,
215. Brunner KT, Hurez D, Mc CR, Benacerraf B. but not CR3 (CD11b/CD18), increases clear-
1960. Blood clearance of P32-labeled vesicular ance of Bordetella pertussis. J Infect Dis 183:871
stomatitis and Newcastle disease viruses by the 879.
reticuloendothelial system in mice. J Immunol 227. Clatworthy MR, Smith KG. 2004. FcgammaRIIb
85:99105. balances efficient pathogen clearance and the
216. Igarashi T, Brown C, Azadegan A, Haigwood cytokine-mediated consequences of sepsis. J Exp
N, Dimitrov D, Martin MA, Shibata R. 1999. Med 199:717723.
Human immunodeficiency virus type 1 neu- 228. Mold C, Rodic-Polic B, Du Clos TW. 2002.
tralizing antibodies accelerate clearance of cell- Protection from Streptococcus pneumoniae
free virions from blood plasma. Nat Med 5:211 infection by C-reactive protein and natural anti-
216. body requires complement but not Fc gamma
217. Kim YB, Bradley SG, Watson DW. 1967. receptors. J Immunol 168:63756381.
Ontogeny of the immune response. IV. The 229. Yee AM, Phan HM, Zuniga R, Salmon JE,
role of antigen elimination in the true primary Musher DM. 2000. Association between
immune response in germ-free, colostrum- FcgammaRIIa-R131 allotype and bacteremic pneu-
deprived piglets. J Immunol 99:320326. mococcal pneumonia. Clin Infect Dis 30:2528.
218. Glenny AT, Hopkins BE. 1923. Duration of 230. Yuan FF, Wong M, Pererva N, Keating J, Davis
passive immunity. J Hyg (Lond) 22:208221. AR, Bryant JA, Sullivan JS. 2003. FcgammaRIIA
219. Fujisawa H. 2008. Neutrophils play an essen- polymorphisms in Streptococcus pneumoniae
tial role in cooperation with antibody in both infection. Immunol Cell Biol 81:192195.
protection against and recovery from pulmo- 231. Rodriguez ME, van der Pol WL, Sanders
nary infection with influenza virus in mice. J LA, van de Winkel JG. 1999. Crucial role of
Virol 82:27722783. FcgammaRIIa (CD32) in assessment of func-
220. Huber VC, Lynch JM, Bucher DJ, Le J, tional anti-Streptococcus pneumoniae anti-
Metzger DW. 2001. Fc receptor-mediated phago- body activity in human sera. J Infect Dis 179:
cytosis makes a significant contribution to clear- 423433.
ance of influenza virus infections. J Immunol 232. Bredius RG, Fijen CA, De Haas M, Kuijper EJ,
166:73817388. Weening RS, Van de Winkel JG, Out TA. 1994.
221. Chan KR, Zhang SL, Tan HC, Chan YK, Chow Role of neutrophil Fc gamma RIIa (CD32) and Fc
A, Lim AP, Vasudevan SG, Hanson BJ, Ooi gamma RIIIb (CD16) polymorphic forms in
EE. 2011. Ligation of Fc gamma receptor IIB phagocytosis of human IgG1- and IgG3-opsonized
inhibits antibody-dependent enhancement of bacteria and erythrocytes. Immunology 83:624
dengue virus infection. Proc Natl Acad Sci USA 630.
108:1247912484. 233. Fijen CA, Bredius RG, Kuijper EJ, Out TA,
222. Chung KM, Thompson BS, Fremont DH, De Haas M, De Wit AP, Daha MR, De Winkel
Diamond MS. 2007. Antibody recognition of JG. 2000. The role of Fcgamma receptor
cell surface-associated NS1 triggers Fc-gamma polymorphisms and C3 in the immune defence
receptor-mediated phagocytosis and clearance against Neisseria meningitidis in complement-
of West Nile Virus-infected cells. J Virol 81: deficient individuals. Clin Exp Immunol 120:
95519555. 338345.
223. Hashimoto Y, Moki T, Takizawa T, Shiratsuchi 234. Bredius RG, Derkx BH, Fijen CA, de Wit TP, de
A, Nakanishi Y. 2007. Evidence for phagocytosis of Haas M, Weening RS, van de Winkel JG, Out
influenza virus-infected, apoptotic cells by neutro- TA. 1994. Fc gamma receptor IIa (CD32) poly-
phils and macrophages in mice. J Immunol morphism in fulminant meningococcal septic
178:24482457. shock in children. J Infect Dis 170:848853.
224. Ratcliffe DR, Michl J, Cramer EB. 1993. 235. Platonov AE, Shipulin GA, Vershinina IV,
Neutrophils do not bind to or phagocytize human Dankert J, van de Winkel JG, Kuijper EJ.
CHAPTER 2 Functions of Antibodies 47

1998. Association of human Fc gamma RIIa JM, Langer C, Warimwe GM, Duffy MF,
(CD32) polymorphism with susceptibility to Rogerson SJ, Bull PC, Cowman AF, Marsh K,
and severity of meningococcal disease. Clin Beeson JG. 2012. Targets of antibodies against
Infect Dis 27:746750. Plasmodium falciparum-infected erythrocytes in
236. Domingo P, Muniz-Diaz E, Baraldes MA, Arilla malaria immunity. J Clin Invest 122:32273238.
M, Barquet N, Pericas R, Juarez C, Madoz P, 247. Tsuboi N, Asano K, Lauterbach M, Mayadas
Vazquez G. 2002. Associations between Fc gamma TN. 2008. Human neutrophil Fcgamma recep-
receptor IIA polymorphisms and the risk and tors initiate and play specialized nonredundant
prognosis of meningococcal disease. Am J Med roles in antibody-mediated inflammatory dis-
112:1925. eases. Immunity 28:833846.
237. Domingo P, Muniz-Diaz E, Baraldes MA, 248. Lendvai N, Qu XW, Hsueh W, Casadevall A.
Arilla M, Barquet N, Pericas R, Juarez C, 2000. Mechanism for the isotype dependence
Madoz P, Vazquez G. 2004. Relevance of gene- of antibody-mediated toxicity in Cryptococcus
tically determined host factors to the prognosis of neoformans-infected mice. J Immunol 164:
meningococcal disease. Eur J Clin Microbiol Infect 43674374.
Dis 23:634637. 249. Alonso A, Bayon Y, Crespo MS. 1996. The
238. Smith I, Vedeler C, Halstensen A. 2003. expression of cytokine-induced neutrophil
FcgammaRIIa and FcgammaRIIIb polymor- chemoattractants (CINC-1 and CINC-2) in rat
phisms were not associated with meningococcal peritoneal macrophages is triggered by Fc
disease in Western Norway. Epidemiol Infect gamma receptor activation: study of the signal-
130:193199. ing mechanism. Eur J Immunol 26:21652171.
239. Wu Y, Wu W, Wong WM, Ward E, Thrasher AJ, 250. Fernandez N, Renedo M, Sanchez Crespo M.
Goldblatt D, Osman M, Digard P, Canaday DH, 2002. FcgammaR receptors activate MAP ki-
Gustafsson K. 2009. Human gamma delta T cells: a nase and up-regulate the cyclooxygenase path-
lymphoid lineage cell capable of professional way without increasing arachidonic acid release in
phagocytosis. J Immunol 183:56225629. monocytic cells. Eur J Immunol 32:383392.
240. Schlageter AM, Kozel TR. 1990. Opsonization 251. Abrahams VM, Cambridge G, Lydyard PM,
of Cryptococcus neoformans by a family of isotype- Edwards JC. 2000. Induction of tumor necro-
switch variant antibodies specific for the capsular sis factor alpha production by adhered human
polysaccharide. Infect Immun 58:19141918. monocytes: a key role for Fcgamma receptor
241. Sanford JE, Lupan DM, Schlageter AM, type IIIa in rheumatoid arthritis. Arthritis
Kozel TR. 1990. Passive immunization against Rheum 43:608616.
Cryptococcus neoformans with an isotype- 252. Fernandez N, Renedo M, Garcia-Rodriguez
switch family of monoclonal antibodies reac- C, Sanchez Crespo M. 2002. Activation of
tive with cryptococcal polysaccharide. Infect monocytic cells through Fc gamma receptors
Immun 58:19191923. induces the expression of macrophage-inflam-
242. Saylor CA, Dadachova E, Casadevall A. 2010. matory protein (MIP)-1 alpha, MIP-1 beta, and
Murine IgG1 and IgG3 isotype switch variants RANTES. J Immunol 169:33213328.
promote phagocytosis of Cryptococcus neoformans 253. Zhang Y, Zhou Y, Yang Q, Mu C, Duan E,
through different receptors. J Immunol 184:336 Chen J, Yang M, Xia P, Cui B. 2012. Ligation
343. of Fc gamma receptor IIB enhances levels of
243. Szymczak WA, Davis MJ, Lundy SK, Dufaud antiviral cytokine in response to PRRSV infec-
C, Olszewski M, Pirofski LA. 2013. X-linked tion in vitro. Vet Microbiol 160:473480.
immunodeficient mice exhibit enhanced sus- 254. Gallo P, Goncalves R, Mosser DM. 2010. The
ceptibility to Cryptococcus neoformans Infec- influence of IgG density and macrophage Fc
tion. MBio 4. doi:10.1128/mBio.00265-13. (gamma) receptor cross-linking on phagocyto-
244. Murphy K, Travers P, Walport M. 2008. The sis and IL-10 production. Immunol Lett
destruction of antibody-coated pathogens via 133:7077.
Fc receptors. In Janeways Immunobiology, 7th 255. Parcina M, Wendt C, Goet F, Zawatzky R,
ed. Garland Science, New York Zahringer U, Heeg K, Bekeredjian-Ding I.
245. Celada A, Cruchaud A, Perrin LH. 1982. 2008. Staphylococcus aureus-induced plasma-
Opsonic activity of human immune serum on cytoid dendritic cell activation is based on an
in vitro phagocytosis of Plasmodium falciparum IgG-mediated memory response. J Immunol
infected red blood cells by monocytes. Clin Exp 181:38233833.
Immunol 47:635644. 256. Jancar S, Sanchez Crespo M. 2005. Immune
246. Chan JA, Howell KB, Reiling L, Ataide R, complex-mediated tissue injury: a multistep
Mackintosh CL, Fowkes FJ, Petter M, Chesson paradigm. Trends Immunol 26:4855.
48 FORTHAL

257. Bunk S, Sigel S, Metzdorf D, Sharif O, 265. Pearse RN, Kawabe T, Bolland S, Guinamard
Triantafilou K, Triantafilou M, Hartung T, R, Kurosaki T, Ravetch JV. 1999. SHIP recruit-
Knapp S, von Aulock S. 2010. Internalization ment attenuates Fc gamma RIIB-induced B cell
and coreceptor expression are critical for TLR2- apoptosis. Immunity 10:753760.
mediated recognition of lipoteichoic acid in human 266. Nimmerjahn F, Ravetch JV. 2008. Fcgamma
peripheral blood. J Immunol 185:37083717. receptors as regulators of immune responses.
258. Lovgren T, Eloranta ML, Kastner B, Nat Rev Immunol 8:3447.
Wahren-Herlenius M, Alm GV, Ronnblom 267. Regnault A, Lankar D, Lacabanne V,
L. 2006. Induction of interferon-alpha by Rodriguez A, Thery C, Rescigno M, Saito T,
immune complexes or liposomes containing Verbeek S, Bonnerot C, Ricciardi-Castagnoli
systemic lupus erythematosus autoantigen- P, Amigorena S. 1999. Fcgamma receptor-
and Sjogrens syndrome autoantigen-associat- mediated induction of dendritic cell matura-
ed RNA. Arthritis Rheum 54:19171927. tion and major histocompatibility complex
259. Boule MW, Broughton C, Mackay F, Akira S, class I-restricted antigen presentation after im-
Marshak-Rothstein A, Rifkin IR. 2004. Toll- mune complex internalization. J Exp Med 189:371
like receptor 9-dependent and -independent 380.
dendritic cell activation by chromatin-immu- 268. DiScipio RG, Daffern PJ, Jagels MA, Broide
noglobulin G complexes. J Exp Med 199:1631 DH, Sriramarao P. 1999. A comparison of C3a
1640. and C5a-mediated stable adhesion of rolling
260. Means TK, Latz E, Hayashi F, Murali MR, eosinophils in postcapillary venules and tran-
Golenbock DT, Luster AD. 2005. Human sendothelial migration in vitro and in vivo.
lupus autoantibody-DNA complexes activate J Immunol 162:11271136.
DCs through cooperation of CD32 and TLR9. J 269. Godau J, Heller T, Hawlisch H, Trappe M,
Clin Invest 115:407417. Howells E, Best J, Zwirner J, Verbeek JS,
261. Ierino FL, Powell MS, McKenzie IF, Hogarth Hogarth PM, Gerard C, Van Rooijen N, Klos
PM. 1993. Recombinant soluble human Fc A, Gessner JE, Kohl J. 2004. C5a initiates
gamma RII: production, characterization, and the inflammatory cascade in immune complex
inhibition of the Arthus reaction. J Exp Med peritonitis. J Immunol 173:34373445.
178:16171628. 270. Fernandez N, Renedo M, Alonso S, Crespo
262. Sylvestre DL, Ravetch JV. 1994. Fc receptors MS. 2003. Release of arachidonic acid by
initiate the Arthus reaction: redefining the stimulation of opsonic receptors in human
inflammatory cascade. Science 265:10951098. monocytes: the FcgammaR and the comple-
263. Hogarth PM, Pietersz GA. 2012. Fc receptor- ment receptor 3 pathways. J Biol Chem 278:
targeted therapies for the treatment of inflam- 5217952187.
mation, cancer and beyond. Nat Rev Drug 271. Casadevall A, Scharff MD. 1994. Serum ther-
Discov 11:311331. apy revisited: animal models of infection and
264. Ravetch JV, Lanier LL. 2000. Immune inhib- development of passive antibody therapy.
itory receptors. Science 290:8489. Antimicrob Agents Chemother 38:16951702.
Antibody Structure

ROBYN L. STANFIELD1 and IAN A. WILSON1,2


3
Currently, well over 1,000 antibody Fab or Fab variable (Fv) structures have
been determined and deposited in the Protein Data Bank (www.rcsb.org).
Many of these antibodies target proteins found on or secreted by infectious
agents, such as viruses or bacteria. At first glance, one antibody Fab fragment
may look just like another, but closer inspection shows that these workhorses
of the adaptive immune system can capitalize on novel structural features
to tailor their binding sites to accommodate targets of diverse shapes, sizes,
and properties. Recent crystallographic and electron microscopy studies of
human antibodies against human immunodeficiency virus type 1 (HIV-1) and
influenza viruses have revealed some unusual structural features and modes
of antigen recognition that enable these antibodies to effectively and broadly
neutralize their rapidly evolving targets. We will focus here on immunoglob-
ulin G (IgG) antibodies, as these are the best characterized from a struct-
ural perspective.

1
Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla,
CA 92037; 2Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0012-2013

49
50 STANFIELD AND WILSON

BASICS OF ANTIBODY STRUCTURE studies have shown that a single adult has
anywhere from 3 to 9 million unique CDR
A typical mammalian IgG antibody has two H3 sequences in circulation (1).
copies each of two protein chains: the light Since the first X-ray crystal structures of
chain (24 kDa) and the heavy chain (55 Fab fragments were determined in the early
kDa). These chains pair in such a way as to 1970s (2, 3), over a thousand structures of
create three structural domains, two Fabs intact IgGs, Fabs, and Fvs have been deter-
and one Fc, that are linked by a flexible mined and deposited in the Protein Data
hinge region and can be readily cleaved Bank (4). Early studies of the different
into Fab and Fc fragments by proteases. The antibody structures noted many common
Fc fragment is a heavy chain dimer of the features. For example, CDR loops L1, L2,
constant heavy 2 (CH2) and constant heavy 3 L3, H1, and H2 usually are found in a
(CH3) segments, and the Fab fragment is a canonical conformation, which can be
mixed light-heavy chain dimer of variable predicted from conserved sequence motifs
light (VL)-constant light (CL) paired with (5, 6, 7, 8). Other analyses of antibody-
variable heavy (VH)-constant heavy 1 (CH1) antigen complexes showed that, while
segments. Each of the eight component units some antibodies use all six CDR loops to
of an IgG adopts an immunoglobulin protein recognize antigen, CDRs H3, H2, and L3
fold. The variable regions of the heavy and usually make the largest contributions to the
light chains are created by a genetic recom- interface with antigen and CDR L2 usually
bination event called V(D)J (heavy chain) or makes the smallest (9). In fact, CDR L2 often
VJ (light chain) recombination, where all makes no contact with small ligands, al-
possible combinations of numerous different though there are of course a few exceptions
V, D, and J gene segments can theoretically to these generalizations. As would be
lead to around one million different antibody expected, Fabs recognizing large protein
sequence combinations. In fact, this number antigens normally have the largest combin-
is much, much larger, as nucleotide addi- ing sites (around 700 to 900 2 of buried
tions and deletions at the V-D, D-J, and V-J surface on the Fab) and often, but not always,
junctions can create extensive diversity in use all 6 CDR loops, while Fabs interacting
length and sequence, and somatic mutations with small haptens or peptides have smaller
in response to antigen challenge can fur- combining sites (160 to 300 2 for haptens
ther alter the sequence and structure of each and 470 to 560 2 for peptides) and are more
antibody. Further diversity can arise from likely to use a smaller number of CDR loops
posttranslational modifications, such as gly- (10). Antigen size also correlates with trends
cosylation and tyrosine sulfation. The Fab in the shape of the combining site. For
fragment recognizes antigen using some or example, large antigens, such as proteins,
all of its six complementarity-determining often interact with a largely flat, undulating
region (CDR) loops (three from each light combining site, while peptides and small
and heavy chain), which extend out from the antigens usually bind into grooves or pocket-
structurally conserved framework region shaped combining sites (10, 11). Analyses of
and make the vast majority of contacts to Fab and Fv fragments in their unliganded
antigen. Of these six CDRs (L1, L2, L3, H1, and antigen-bound forms have shown that,
H2, and H3), CDR H3 is the most variable in while some antibody combining sites are
length, sequence, and structure. This loop is preformed and do not undergo conforma-
formed at the V-D-J junction, and extra tional changes upon binding antigen, others
nucleotides inserted or deleted between V-D show varying degrees of flexibility, ranging
and D-J account for its extremely high level from small shifts in CDR position, to large
of variability. Recent 454 deep sequencing conformational rearrangements of the CDR
CHAPTER 3 Antibody Structure 51

loops, to shifts in the relative positions and subdomain termed a hammerhead that has a
orientations of the VL and VH domains, and two-headed, bifurcated loop structure (20, 21)
sometimes combinations of all of these (Fig. 1). The loop conformation is maintained
(12, 13). However, as phage-display tech- when PG9 is bound to a scaffold-supported
nology (14, 15), interrogation of memory B gp120 V1/V2 domain (22) (Fig. 1), where the
cells (16, 17), and improved hybridoma large CDR H3 binds to a V2 -strand at the base
techniques (18) have been developed to of a crevice formed by two glycan moieties from
allow for the isolation and structure deter- gp120. The glycan linked to Asn160 is critical
mination of a large number of interesting for the interaction, and a second glycan at
antibodies, some unusual structural features either Asn156 or Asn173 contributes to the
have been revealed, especially in human binding interface. Another potent and broadly
antibodies against viruses, such as HIV-1 neutralizing HIV-1 antibody that also targets
and influenza. the V1/V2 region is PGT145. This Fab has been
structurally characterized in its unliganded
form, and its 31/33-residue CDR H3 loop
CDR H3: SIZE AND SUBTERFUGE adopts an extremely long -hairpin finger
capped by a type I -turn that extends about
It has long been known that CDR H3 is the most 25 above the platform formed by the tips of
variable of all the CDR loops in length, the other CDR loops (Fig. 1). Keeping in mind
sequence, and structure. However, for many that a typical Fab measures about 65 along its
years, the majority of antibody structures long axis, this CDR H3 is almost 40% as long as
determined were from mice, whose antibodies the main body of the Fab itself. It has been
have inherently shorter CDR H3 regions than noted that long CDR H3 loops are often found
those of humans (19), with an average H3 in antiviral antibodies (23) and perhaps are
length (H3 defined with IMGT [international necessary to access recessed regions on the
ImMunoGeneTics information system, http:// viral proteins. This perceived requirement
www.imgt.org] boundaries and Kabat number- for long CDR H3 regions has long concerned
ing H93 to H102; insertions can occur after antiviral vaccine developers, but recently, a 454
H100) of around 11 to 12 and observed lengths deep-sequencing study (24) has shown that
of 1 to 21. In contrast, human CDR H3 regions antibodies with these extremely long H3 CDR
have an average length of about 15 residues, and loops are found in the naive repertoire, albeit
range from 1 to 35 amino acids in length (19). infrequently, with long ( 22/24) and very long
Recent structures of anti-HIV-1 antibodies ( 26/28) H3 loops found in 3.5% and 0.43%
derived from infected donors have revealed of the naive population, respectively. Thus,
several antibodies with extremely long CDR long CDR H3s are generated during the original
H3 regions. Two such antibodies are the V(D)J recombination event, often using the
clonally related PG16 and PG9, which are longest D and J genes, in combination with
potent and broadly neutralizing anti-HIV-1 nucleotide additions at the joining junctions
antibodies that recognize the V1/V2 region in (24). Thus, although long CDR H3s could also
gp120 of the trimeric viral envelope protein. be created through extensive somatic muta-
PG16 and PG9 differ at just 9 amino acid tion, antibodies with these loops are likely
positions within their 28/30-residue CDR H3 available for activation in the naive repertoire.
(in the text and in Table 1, we list the H3 length PG9, PG16, and PGT145 also share the
as defined by both Kabat and IMGT boundaries unusual posttranslational modification of ty-
[Kabat/IMGT], with the IMGT length always 2 rosine sulfation in their CDR H3 regions (22),
amino acids longer). In the unliganded PG16 with up to 2, 1, and 2 tyrosine residues sulfated
structure, the large CDR H3 hovers above the in PG9, PG16, and PGT145, respectively. The
other CDR loops in a hydrogen-bond-stabilized CDRs are all highly negatively charged, and, in
52
STANFIELD AND WILSON
TABLE 1 CDR lengths and posttranslational modications of unusual human antibodies
Length of CDR (residues)a:
L1 L2 L3 H1 H2 H3 H3
Antibody Epitope (2434) (5056) (8997) (3135x) (5065) (95102; Kabat) (93102; IMGT) Modication(s)

PG9 HIV-1 V1/V2 14 7 10 5 17 28 30 Long H3, tyrosine sulfation


PG16 HIV-1 V1/V2 14 7 10 5 17 28 30 Long H3, tyrosine sulfation,
N-linked glycan
PGT128 HIV-1 V3 glycans 9 7 10 7 22 19 21 Long H2, short L1
PGT145 HIV-1 V1/V2 16 7 9 5 19 31 33 Long H3, tyrosine sulfation
2G12 HIV-1 glycan shield 11 7 9 5 17 14 16 Domain-swapped Fab
2D1 Inuenza head 13 7 12 7 18 14 16 Unusual 3-residue insert after
residue H62; only physically
lengthens CDR by 1 residue
CR6261 Inuenza HAb stem 13 7 12 5 17 12 14 Heavy-chain-only binding
C05 Inuenza HA receptor 11 7 9 10 17 24 26 Long H3 dominates binding, long
binding site H1 due to unusual 5-residue insert
at H27 not accounted for
in Kabat CDR denition
a
CDR lengths are dened using the Kabat boundaries, except for H3, which is dened using both Kabat and IMGT boundaries. Note that C05 has a long H1 due to an unusual insert at H27; the
length in the table reects that insertion.
b
HA, hemagglutinin.
CHAPTER 3 Antibody Structure 53

FIGURE 1 Extremely long CDR H3 loops in human anti-HIV-1 antibodies targeting the V1/V2 region of
gp120 in the Env trimer. Fab PG16 (left, PDB 3mug) has a large (28/30 residue), structured hammerhead
CDR H3 (red) with posttranslational modications of one sulfated tyrosine and one N-linked glycan. PG9
(middle, PDB 3u4e) in complex with the V1/V2 domain (yellow) from HIV-1 gp120 (CAP45 isolate) uses
its large CDR H3 to bisect two glycans (shown in yellow ball-and-stick representation) and form a parallel
-sheet interaction with one strand from V1/V2. PG9 has two sulfated tyrosine residues at its tip. Fab
PGT145 (right, PDB 3u1s) has the longest CDR H3 (31/33 residues) yet seen in human antibodies. This CDR
also has two sulfated tyrosine residues at its tip. For the C trace of both Fabs, the CDR loops are colored
orange (L1), magenta (L2), green (L3), blue (H1), pink (H2), and red (H3), with light chains in light gray and
heavy chains in dark gray. All gures were made with MOLSCRIPT (56) and rendered with Raster3D (57).
doi:10.1128/microbiolspec.AID-0012-2013.f1

PG9, the sulfated tyrosines have been shown residues that are thought to interact with
to specifically interact with residues in its gp120, so antibodies with this same posttrans-
cationic b-strand binding site located in the lational modification are thought to bind to
gp120 V2 region (22). Two other antibodies regions on gp120 that interact with CCR5.
that target the V1/V2 region, 2909 and CHO4, Some antibodies to other viral and nonviral
also have sulfated tyrosines within their targets also contain sulfated tyrosines that
highly anionic CDR H3 loops (22, 25, 26). arise from cellular processing of tyrosines
Sulfated tyrosines have also been found in embedded in a sea of acidic residues (27).
several other HIV-1 neutralizing antibodies
(27, 28) against a CD4 receptor-induced
epitope, which overlaps the coreceptor bind- HEAVY CHAIN DOMINATION AND
ing site. When HIV-1 enters cells, it does so by FRAMEWORK CONTACTS
interacting with two receptors, first CD4 and
then a coreceptor that is usually the G protein- We have discussed the important role CDR
coupled receptor CCR5. The CCR5 N termi- H3 can play in antibody-antigen interactions,
nus contains a number of sulfated tyrosine and it is interesting that jawless vertebrates
54 STANFIELD AND WILSON

(the most primitive animals to possess immu- insertion at residue H31) (Fig. 2). By using
noglobulin-based antibodies) have as part of mainly one loop for recognition, the large
their repertoire heavy-chain-only antibodies Fab mimics the mechanism, but not the
called new antigen receptors that bind specific interaction, of the small sialic acid
antigen with only a single VH domain that receptors of influenza virus. Two other
contains only two CDR loops (H1 and H3) (29, interesting anti-influenza antibodies, Fab
30). Camels, llamas, and alpacas also have CR6261 (38) and scFv F10 (39), also use
functional heavy-chain-only antibodies that only their heavy chain and bind to closely
originated from a mutation resulting in loss of overlapping epitopes on the conserved stalk
the CH1 domain. The resulting VH-CH2-CH3 region of the trimeric hemagglutinin (Fig. 2)
heavy chain dimerizes, forming a normal Fc in nearly identical fashions, using primarily
region, but does not associate with the light the H2 CDR loop and contributions from
chain (31, 32, 33). The camelid and shark heavy CDR H3, as well as residues from the
chain variable regions have structurally con- framework V H7275 loop (HV4), in the
verged to have very long CDR H3 regions that case of CR6261. These antibodies neutralize
often contain noncanonical disulfide bonds virus by preventing a conformational rear-
that can link H3 to other CDRs or to different rangement necessary for viral fusion. Inter-
parts of the VH framework region, and several estingly, CR6261, F10, and some other
structures have shown that the loop (H72 to related anti-influenza antibodies (40) all
H75) analogous to HV4 in a T-cell receptor use the VH169 germ line gene (41). This
can also be used to contact antigen (34, 35). germ line gene codes for a CDR H2 with a
These camelid and shark heavy-chain-only hydrophobic tip that is used by these anti-
antibodies have inspired extensive research bodies to bind into hydrophobic pockets.
into single domain (VHH) antibody fragments Other anti-HIV and anti-influenza anti-
that are currently in use as drugs and reagents bodies derived from VH169 include at least
(36). 9 different CD4-induced anti-HIV-1 anti-
Since these shark and camel antibodies bodies (42) that all have protruding hydro-
work perfectly well with only a single VH phobic residues at the tip of CDR H2 and
domain, it is perhaps not surprising that the anti-HIV antibody D5 that targets a gp41
epitopes of some conventional light-heavy fusion intermediate (43) and also contacts a
chain Fabs have a larger contribution from hydrophobic pocket on gp41 with its hydro-
VH than from VL. Heavy chain domination of phobic H2 residues.
the antibody-antigen interaction is not un-
common with small antigens or peptides;
however, it is more unusual with protein DISULFIDES IN THE CDRS
antigens, where all six CDR loops are usually
used to contact antigen. Recently, some Fabs While noncanonical disulfide bonds in the
have been found that use only their VH CDRs of camels and sharks are common,
domain to contact large, protein antigens. disulfide bonds within or between human
Antibody C05 (37) is a broadly neutralizing or murine antibody CDR loops are not.
influenza antibody that uses its long CDR H3 Cysteine is coded in one human germ line
(24/26 residues) to access the very small, but DH gene, and 1.2% of all human CDR H3
highly conserved, receptor binding site on residues have been found to be cysteine
the hemagglutinin surface glycoprotein, (19). With no homologous germ line gene in
with minor additional contacts from an mice, cysteines in H3 are rarer in that
unusually long CDR H1, which has a 5- system. Within complete human CDR H3
amino-acid insertion at position H27 (rather regions, 3.35% of the CDRs were found to
than the more common 0- or 2-residue have one cysteine and 5.98% contained two
CHAPTER 3 Antibody Structure 55

FIGURE 2 Broadly neutralizing Fab CR6261 (left) uses only its heavy chain (dark gray, with blue, pink, and
red CDR loops) to recognize the stem region of a hemagglutinin trimer. Fab C05 (right) binds to a
conserved region near the receptor binding site, using mainly its large CDR H3 (red), with minor contact
from CDR H1 (blue). doi:10.1128/microbiolspec.AID-0012-2013.f2

cysteine residues. One of the earliest anti- of the ligand-bound structures, such as
bodies to have its Fab crystal structure PGT128 and F045-092, the Fab contacts
determined, human KOL (2), has a disulfide with the antigen include the disulfide, so
within its CDR H3, between residues H100a these are important for the antigen recog-
and H100f. Crystal structures for other nition process. The disulfides may also be of
antibodies containing H3 disulfides exist importance in stabilizing a particular CDR
for human Fab 8F9 (44) to human cyto- conformation or position.
megalovirus (disulfide between H98 and
H100c), human Fab CB3S (45) that recog-
nizes BLyS receptor 3 (disulfide between SELF-CARBOHYDRATE RECOGNITION:
H97 and H100d), and a human anti-influ- PENETRATING THE GLYCAN SHIELD
enza Fab F045-092 (P. Lee, personal com-
munication) (disulfide between H100c and The envelope proteins on viruses, such as HIV-
H100f). Anti-HIV Fab PGT128 (46) has a 1, are extensively glycosylated using host-
disulfide bond linking CDR H1 to CDR H2 derived enzymes, so the viral glycans contain
(residues H32 and H52B), and murine anti- only human carbohydrate moieties. Thus, one
HIV Fab 59.1 against the gp120 V3 region would expect that antibodies would not target
(47) also has a disulfide bond linking these such glycosylated regions in viral proteins. In
two CDRs (residues H35 and H52). In some fact, the dense coat of carbohydrates covering
56 STANFIELD AND WILSON

most of the outside of the gp120 envelope extensively mutated in comparison to its
protein has long been termed the glycan putative germ line sequence, with 38 and 16
shield or silent face (48) because it was somatic mutations in the heavy and light
thought to make the protein invisible to the chains, respectively. However, just 5 to 7 of
immune system. However, several anti-HIV-1 these mutations are enough to cause the germ
antibodies have now been discovered whose line Fab to become significantly domain ex-
primary targets are these same self glycans. It is changed (49), with four being especially im-
thought that the extremely high density of portant: Ala at H14, Glu at H75, Ile at H19, and
high-mannose glycans on gp120 is unusual Pro at H113. Structures of 2G12 in complex
among human proteins and, thus, is responsi- with Man9GlcNAc2 carbohydrates and other,
ble for the surprising immunogenicity. The smaller, components of high-mannose sugars
first such antibody, 2G12, that was found to (50, 51), show that each antigen-combining site
target the HIV-1 glycan shield is among the of the dimeric Fab binds to the D1 arm of one
most unusual antibodies found to date because Man9GlcNAc2 sugar, closely contacting the
the two Fab fragments on the IgG are domain- two terminal mannose sugars. In addition,
swapped through their VH domains so that the sugar groups from a crystallographic symme-
two Fab fragments are intimately interlocked try-related D2 arm bind in the cleft between
in a side-by-side arrangement (Fig. 3). 2G12 is the two closely spaced VH domains, suggesting

FIGURE 3 Self-carbohydrate recognition by HIV-1 antibodies. (Left) Dimeric, domain-swapped 2G12


binds the D1 arms of GlcNAc2Man9 (yellow) via two closely spaced primary combining sites. A third
potential binding site, located between the closely spaced VH and VH domains, binds symmetry-related
sugars (orange). (Middle and right) PGT128 also binds to two sugar moieties and inserts its CDR H3 (red)
and long H2 (magenta) between two high-mannose sugars to contact the protein surface. The Fab-
antigen complex is shown from two different sides to illustrate the interaction. 2G12, PGT128, and PG9
(Fig. 1) bind two high-mannose sugars, either with two separate binding sites, as in 2G12, or by wedging
long CDR loops between two carbohydrates, as in PGT128 and PG9 recognition. doi:10.1128/
microbiolspec.AID-0012-2013.f3
CHAPTER 3 Antibody Structure 57

a possible secondary binding site. Antibody binding sites for carbohydrate to increase the
affinity for carbohydrates is usually weak avidity of an otherwise weak interaction. The
(micro- to millimolar range); however, multi- heavy chain regions of PG9, PG16, PGT128, and
valent binding of the dimeric 2G12 Fab in- PGT145 also give rise to uniquely shaped,
creases avidity to the nanomolar range, which convex paratopes that can penetrate the glycan
increases the effectiveness of 2G12 in viral shield. These paratope topologies differ from
neutralization. the more traditional flat and undulating, or
More recently, several extremely broad and concave pocket or groove shapes (10, 11) (Fig.
potent neutralizing antibodies have been dis- 4). In addition, the 2G12 paratope is very broad,
covered that also bind to high-mannose glycans extending from one Fab binding site to another,
on gp120 without the use of 2G12-like domain in which up to four potential binding pockets
exchange. Already mentioned above are PG9 for carbohydrate are located.
and PG16, which use their large hammer-
head CDR H3 to bind in a cleft between two
sugars on the gp120 V1/V2 region, with one UNUSUAL INDELS
sugar on each side of the large CDR H3, and
backbone-backbone hydrogen bonds from the Some recently isolated influenza and HIV-1
-strand-like top of H3 to a -strand of the V1/ antibodies have highly unusual insertion mu-
V2 region (22). PGT128 is another broad and tations. We have already described the long H1
potent antibody that binds to a carbohydrate- insert in Fab C05 and the long H2 insert in Fab
containing epitope at the base of the gp120 V3 PGT128. In both of these cases, the inserted
loop (Fig. 3). A glycan at gp120 residue Asn332 residues cause the CDR loop to become longer
binds into the primary combining site of but do not alter any of the Fab framework
PGT128, contacting residues in H2, H3, L3, structure (Fig. 5). Antibody 2D1 is an influenza
and framework region 2, while the Asn301 antibody with a 3-amino-acid insertion on the
glycan contacts residues in CDR H1, frame- C-terminal side of CDR H2 at position H62
work region 3, and the disulfide bond linking (52). This insertion causes a register shift,
CDR H1 to CDR H2. Contact is also made from lengthening the CDR H2 by one residue and,
Fab to several backbone atoms of the gp120 in surprisingly, extending the H65-H66 loop
another -strand-like interaction. PGT128 has (which faces toward the Fab Fc region) (Fig.
a moderately long CDR H3 loop (19/21 amino 5). The bulge at the tip of H2 also causes
acids), the longest H2 CDR loop yet seen, with distortion of the neighboring H1 CDR. This is
a 6-amino-acid insert after residue H52 (inser- the first insertion that we are aware of that has
tions here are usually 1 or 3 amino acids in not simply resulted in extension of a CDR loop.
length), and a very short CDR L1 (residues 28 Removal of the inserted residues in 2D1 results
and 29 are missing). These insertions and in canonical structures for the H1 and H2 CDRs
deletions combine to make the heavy chain but reduced affinity for hemagglutinin and
component of the Fab paratope much more reduced viral neutralization.
elevated than the light chain part (Fig. 3), and
most of the interactions between PGT128 and
gp120 are with the heavy chain. MECHANISMS OF ANTIBODY
PGT128, PG9, and PG16 all bind to NEUTRALIZATION
carbohydrates so that they contact multiple
mannose and/or GlcNAc sugar moieties, as Working in concert with the novel antiviral
opposed to merely contacting the distal tips of antibody structural features are some equal-
the Man9GlcNAc2 residues, as with 2G12. ly novel mechanisms of neutralization. The
Thus, 2G12, PG9, PG16, and PGT128 have all classic model for antibody neutralization of
devised ways to create multiple or large virus is the binding of antibody to virus and
58 STANFIELD AND WILSON

FIGURE 4 Diversity in the shape of antibody combining sites. Fab domain swapping and extremely long
CDR H3 loops result in unusually shaped paratopes. (Top) Fv molecular surfaces are shown in gray, with
paratopes (if known) colored in magenta and antigens shown in yellow. Classic antibody binding sites
are usually pockets for small haptens (DB3, progesterone), grooves for peptides (B13I2, myohemerythrin
peptide; 17/9, inuenza peptide), or at, undulating surfaces for proteins (HyHEL-10, lysozyme; HyHEL-5,
lysozyme). (Bottom) In contrast, domain-swapped 2G12 has created a paratope consisting of 4 different
binding pockets, and PGT128 and PG9 have protruding, convex paratopes that can be inserted between
two large carbohydrate moieties. A structure of PGT145 with its antigen has not yet been determined,
but its very long CDR H3 can be clearly visualized. doi:10.1128/microbiolspec.AID-0012-2013.f4

the subsequent blocking by the bulky IgG of potently as an IgG than as an Fab, although the
virus attachment to a host cell receptor. binding affinities for Fab and IgG to isolated
However, some recent work has uncovered gp120 trimers are very similar. Thus, the IgG is
other mechanisms. The anti-influenza anti- postulated to cross-link different gp120
bodies CR6261, F10, and CR9114 (53) all trimers on the viral surface. The PGT128 also
bind to the hemagglutinin trimer stem and reduces the lifetime of viral infectivity more
have been shown to block conformational than comparable antibodies, so the cross-
changes that must take place in the hemag- linking may be causing conformational
glutinin to bring about membrane fusion. changes in the gp120 trimers that interfere
Two other anti-influenza antibodies, CR8033 with their functionality (46).
and CR8071 (53), bind to two different
epitopes on the hemagglutinin head and,
while they do not prevent viral entry or SUMMARY
genomic replication, they do prevent the
virus from propagating. In the presence of Recent advances in the isolation, identifica-
these antibodies, newly created viruses are tion, and production of potent and broadly
not released from the infected cell, similar to neutralizing human, antiviral antibodies
what is seen with the neuraminidase inhibitor have resulted in many exciting new crystal
zanamivir. The anti-HIV antibody PGT128 has structures for these antibodies in complex
been shown to neutralize virus much more with their antigens. While the ultimate goal
CHAPTER 3 Antibody Structure 59

altered the landscape, and human antibodies


have now completely changed our view of
the range of structural features and mecha-
nisms that can be used to target the universe
of potential antigens. The last few years have
been very exhilarating and suggest that the
next 30 years of antibody structural studies
will prove just as exciting, as more novel
human antibodies are discovered, as well as
those from other species such as cows,
whose extravagantly long and cysteine-rich
CDR H3s far exceed anything seen so far (54,
55). More importantly, these studies will no
doubt prove helpful in the design of new
vaccines and antibody therapeutics to pre-
FIGURE 5 Unusual insertions in CDRs H1 and H2
from Fabs C05 (red), 2D1 (yellow), and PGT128
vent human disease.
(blue). (Left) In these H1 CDRs, the colored areas
cover residues H26 to H35b. C05 has an unusually ACKNOWLEDGMENT
long 5-residue insertion in H1, while PGT128 and
2D1 have more common 2-residue insertions in Conflicts of interest: I declare no conflicts.
H1. The 2D1 H1 CDR has shifted away its canonical
conformation due to a clash with its long H2 loop.
(Right) H2 CDRs from the same Fabs, with the CITATION
same coloring from positions H51 to H57. C05 has
a normal H2 CDR with a 1-residue insertion after Stanfield RL, Wilson IA. 2014. Antibody
residue 52. PGT128 has a very rare 6-residue structure. Microbiol Spectrum 2(2):AID-
insertion in this CDR, which is reected in its 0012-2013.
unusually long H2 CDR loop. 2D1 has a 3-residue
insertion in the CDR that extends the physical CDR
length by 1 residue, but the other inserted REFERENCES
residues are accommodated in the loop around
H63H64 that faces the CH1 domain. doi:10.1128/ 1. Arnaout R, Lee W, Cahill P, Honan T, Sparrow
microbiolspec.AID-0012-2013.f5 T, Weiand M, Nusbaum C, Rajewsky K, Koralov
SB. 2011. High-resolution description of antibody
heavy-chain repertoires in humans. PLoS One 6:
of producing effective vaccines to prevent e22365.
viral infection has been advanced by the 2. Palm W. 1974. The physico-chemical character-
identification of novel epitope regions on ization and crystallization of immunoglobulins and
their fragmentation products: a contribution to the
viral antigens, many structural features of elucidation of the three-dimensional structure
the antibodies themselves have proven to of antibodies. II. Human immunoglobulin Kol: a
be surprising. Almost 30 years have passed myeloma protein of the class IgG, gamma2 kappa2.
since the first Fab structure was determined Hoppe Seylers Z Physiol Chem 355:877880.
at atomic resolution, yet new insights into 3. Amzel LM, Poljak RJ, Saul F, Varga JM,
Richards FF. 1974. The three dimensional struc-
how these immune receptors can adapt to ture of a combining region-ligand complex of
any pathogen are constantly being revealed. immunoglobulin NEW at 3.5- resolution. Proc
A plethora of mouse antibody structures, Natl Acad Sci USA 71:14271430.
from the few that trickled out in the 1970s to 4. Berman H, Henrick K, Nakamura H. 2003.
increasing numbers in the 1980s and 1990s, Announcing the worldwide Protein Data Bank.
Nat Struct Biol 10:980.
led one to believe that everything was known 5. Chothia C, Lesk AM. 1987. Canonical structures
about antibody structure and function. for the hypervariable regions of immunoglobulins.
Structures of camel and shark antibodies J Mol Biol 196:901917.
60 STANFIELD AND WILSON

6. Al-Lazikani B, Lesk AM, Chothia C. 1997. 2003. Expressed murine and human CDR-H3
Standard conformations for the canonical struc- intervals of equal length exhibit distinct reper-
tures of immunoglobulins. J Mol Biol 273:927948. toires that differ in their amino acid composition
7. Martin AC, Thornton JM. 1996. Structural fam- and predicted range of structures. J Mol Biol
ilies in loops of homologous proteins: automatic 334:733749.
classification, modelling and application to anti- 20. Pejchal R, Walker LM, Stanfield RL, Phogat SK,
bodies. J Mol Biol 263:800815. Koff WC, Poignard P, Burton DR, Wilson IA.
8. North B, Lehmann A, Dunbrack RL Jr. 2011. 2010. Structure and function of broadly reactive
A new clustering of antibody CDR loop con- antibody PG16 reveal an H3 subdomain that
formations. J Mol Biol 406:228256. mediates potent neutralization of HIV-1. Proc
9. Wilson IA, Stanfield RL. 1994. Antibody- Natl Acad Sci USA 107:1148311488.
antigen interactions: new structures and new 21. Pancera M, McLellan JS, Wu X, Zhu J,
conformational changes. Curr Opin Struct Biol Changela A, Schmidt SD, Yang Y, Zhou T,
4:857867. Phogat S, Mascola JR, Kwong PD. 2010.
10. Wilson IA, Stanfield RL. 1993. Antibody- Crystal structure of PG16 and chimeric dissec-
antigen interactions. Curr Opin Struct Biol tion with somatically related PG9: structure-
3:113118. function analysis of two quaternary-specific
11. MacCallum RM, Martin AC, Thornton JM. antibodies that effectively neutralize HIV-1. J
1996. Antibody-antigen interactions: contact Virol 84:80988110.
analysis and binding site topography. J Mol 22. McLellan JS, Pancera M, Carrico C, Gorman
Biol 262:732745. J, Julien JP, Khayat R, Louder R, Pejchal R,
12. Stanfield RL, Takimoto-Kamimura M, Rini Sastry M, Dai K, ODell S, Patel N, Shahzad-
JM, Profy AT, Wilson IA. 1993. Major anti- ul-Hussan S, Yang Y, Zhang B, Zhou T, Zhu
gen-induced domain rearrangements in an J, Boyington JC, Chuang GY, Diwanji D,
antibody. Structure 1:8393. Georgiev I, Kwon YD, Lee D, Louder MK,
13. Stanfield RL, Wilson IA. 1994. Antigen-in- Moquin S, Schmidt SD, Yang ZY, Bonsignori
duced conformational changes in antibodies: a M, Crump JA, Kapiga SH, Sam NE, Haynes
problem for structural prediction and design. BF, Burton DR, Koff WC, Walker LM,
Trends Biotechnol 12:275279. Phogat S, Wyatt R, Orwenyo J, Wang LX,
14. Kang AS, Barbas CF, Janda KD, Benkovic SJ, Arthos J, Bewley CA, Mascola JR, Nabel GJ,
Lerner RA. 1991. Linkage of recognition and Schief WR, Ward AB, Wilson IA, Kwong PD.
replication functions by assembling combinatorial 2011. Structure of HIV-1 gp120 V1/V2 domain
antibody Fab libraries along phage surfaces. Proc with broadly neutralizing antibody PG9. Na-
Natl Acad Sci USA 88:43634366. ture 480:336343.
15. Clackson T, Hoogenboom HR, Griffiths AD, 23. Collis AV, Brouwer AP, Martin AC. 2003.
Winter G. 1991. Making antibody fragments Analysis of the antigen combining site: correlations
using phage display libraries. Nature 352:624628. between length and sequence composition of the
16. Scheid JF, Mouquet H, Feldhahn N, Walker BD, hypervariable loops and the nature of the antigen.
Pereyra F, Cutrell E, Seaman MS, Mascola JR, J Mol Biol 325:337354.
Wyatt RT, Wardemann H, Nussenzweig MC. 24. Briney BS, Willis JR, Crowe JE Jr. 2012.
2009. A method for identification of HIV gp140 Human peripheral blood antibodies with long
binding memory B cells in human blood. J HCDR3s are established primarily at original
Immunol Methods 343:6567. recombination using a limited subset of
17. Walker LM, Phogat SK, Chan-Hui PY, germline genes. PLoS One 7:e36750.
Wagner D, Phung P, Goss JL, Wrin T, 25. Changela A, Wu X, Yang Y, Zhang B, Zhu J,
Simek MD, Fling S, Mitcham JL, Lehrman JK, Nardone GA, ODell S, Pancera M, Gorny
Priddy FH, Olsen OA, Frey SM, Hammond PW, MK, Phogat S, Robinson JE, Stamatatos L,
Kaminsky S, Zamb T, Moyle M, Koff WC, Zolla-Pazner S, Mascola JR, Kwong PD. 2011.
Poignard P, Burton DR. 2009. Broad and potent Crystal structure of human antibody 2909
neutralizing antibodies from an African donor reveals conserved features of quaternary struc-
reveal a new HIV-1 vaccine target. Science 326: ture-specific antibodies that potently neutral-
285289. ize HIV-1. J Virol 85:25242535.
18. Zhang C. 2012. Hybridoma technology for the 26. Spurrier B, Sampson JM, Totrov M, Li H,
generation of monoclonal antibodies. Methods ONeal T, Williams C, Robinson J, Gorny
Mol Biol 901:117135. MK, Zolla-Pazner S, Kong XP. 2011. Structural
19. Zemlin M, Klinger M, Link J, Zemlin C, Bauer analysis of human and macaque mAbs 2909 and
K, Engler JA, Schroeder HW Jr, Kirkham PM. 2.5B: implications for the configuration of the
CHAPTER 3 Antibody Structure 61

quaternary neutralizing epitope of HIV-1 gp120. Lerner RA, Bhatt RR, Wilson IA. 2012. Cross-
Structure 19:691699. neutralization of influenza A viruses mediated
27. Choe H, Li W, Wright PL, Vasilieva N, by a single antibody loop. Nature 489:526532.
Venturi M, Huang CC, Grundner C, Dorfman 38. Ekiert DC, Bhabha G, Elsliger MA, Friesen
T, Zwick MB, Wang L, Rosenberg ES, Kwong RH, Jongeneelen M, Throsby M, Goudsmit J,
PD, Burton DR, Robinson JE, Sodroski JG, Wilson IA. 2009. Antibody recognition of a
Farzan M. 2003. Tyrosine sulfation of human highly conserved influenza virus epitope. Sci-
antibodies contributes to recognition of the ence 324:246251.
CCR5 binding region of HIV-1 gp120. Cell 114: 39. Sui J, Hwang WC, Perez S, Wei G, Aird D,
161170. Chen LM, Santelli E, Stec B, Cadwell G, Ali
28. Huang CC, Lam SN, Acharya P, Tang M, M, Wan H, Murakami A, Yammanuru A,
Xiang SH, Hussan SS, Stanfield RL, Robinson Han T, Cox NJ, Bankston LA, Donis RO,
J, Sodroski J, Wilson IA, Wyatt R, Bewley Liddington RC, Marasco WA. 2009. Structural
CA, Kwong PD. 2007. Structures of the CCR5 and functional bases for broad-spectrum neutral-
N terminus and of a tyrosine-sulfated antibody ization of avian and human influenza A viruses.
with HIV-1 gp120 and CD4. Science 317:1930 Nat Struct Mol Biol 16:265273.
1934. 40. Kashyap AK, Steel J, Oner AF, Dillon MA,
29. Stanfield RL, Dooley H, Flajnik MF, Wilson Swale RE, Wall KM, Perry KJ, Faynboym A,
IA. 2004. Crystal structure of a shark single- Ilhan M, Horowitz M, Horowitz L, Palese P,
domain antibody V region in complex with Bhatt RR, Lerner RA. 2008. Combinatorial
lysozyme. Science 305:17701773. antibody libraries from survivors of the Turk-
30. Nuttall SD. 2012. Overview and discovery of ish H5N1 avian influenza outbreak reveal virus
IgNARs and generation of VNARs. Methods neutralization strategies. Proc Natl Acad Sci
Mol Biol 911:2736. USA 105:59865991.
31. Muyldermans S, Atarhouch T, Saldanha J, 41. Throsby M, van den Brink E, Jongeneelen
Barbosa JA, Hamers R. 1994. Sequence and M, Poon LL, Alard P, Cornelissen L, Bakker
structure of VH domain from naturally A, Cox F, van Deventer E, Guan Y, Cinatl J,
occurring camel heavy chain immunoglobulins ter Meulen J, Lasters I, Carsetti R, Peiris
lacking light chains. Protein Eng 7:11291135. M, de Kruif J, Goudsmit J. 2008. Hetero-
32. Spinelli S, Frenken L, Bourgeois D, de Ron L, subtypic neutralizing monoclonal antibodies cross-
Bos W, Verrips T, Anguille C, Cambillau C, protective against H5N1 and H1N1 recovered from
Tegoni M. 1996. The crystal structure of a human IgM+ memory B cells. PLoS One 3:e3942.
llama heavy chain variable domain. Nat Struct 42. Huang CC, Venturi M, Majeed S, Moore MJ,
Biol 3:752757. Phogat S, Zhang MY, Dimitrov DS, Hendrickson
33. Muyldermans S, Cambillau C, Wyns L. 2001. WA, Robinson J, Sodroski J, Wyatt R, Choe H,
Recognition of antigens by single-domain an- Farzan M, Kwong PD. 2004. Structural basis of
tibody fragments: the superfluous luxury of tyrosine sulfation and VH-gene usage in antibodies
paired domains. Trends Biochem Sci 26:230 that recognize the HIV type 1 coreceptor-binding
235. site on gp120. Proc Natl Acad Sci USA 101:2706
34. Stanfield RL, Dooley H, Verdino P, Flajnik 2711.
MF, Wilson IA. 2007. Maturation of shark 43. Luftig MA, Mattu M, Di Giovine P, Geleziunas
single-domain (IgNAR) antibodies: evidence R, Hrin R, Barbato G, Bianchi E, Miller MD,
for induced-fit binding. J Mol Biol 367:358 Pessi A, Carfi A. 2006. Structural basis for HIV-1
372. neutralization by a gp41 fusion intermediate-
35. Fanning SW, Horn JR. 2011. An anti-hapten directed antibody. Nat Struct Mol Biol 13:740747.
camelid antibody reveals a cryptic binding site 44. Thomson CA, Bryson S, McLean GR, Creagh
with significant energetic contributions from a AL, Pai EF, Schrader JW. 2008. Germline
nonhypervariable loop. Protein Sci 20:1196 V-genes sculpt the binding site of a family of
1207. antibodies neutralizing human cytomegalovirus.
36. Vanlandschoot P, Stortelers C, Beirnaert E, EMBO J 27:25922602.
Ibanez LI, Schepens B, Depla E, Saelens X. 45. Lee CV, Hymowitz SG, Wallweber HJ,
2011. Nanobodies(R): new ammunition to bat- Gordon NC, Billeci KL, Tsai SP, Compaan
tle viruses. Antiviral Res 92:389407. DM, Yin J, Gong Q, Kelley RF, DeForge LE,
37. Ekiert DC, Kashyap AK, Steel J, Rubrum A, Martin F, Starovasnik MA, Fuh G. 2006.
Bhabha G, Khayat R, Lee JH, Dillon MA, Synthetic anti-BR3 antibodies that mimic
ONeil RE, Faynboym AM, Horowitz M, BAFF binding and target both human and
Horowitz L, Ward AB, Palese P, Webby R, murine B cells. Blood 108:31033111.
62 STANFIELD AND WILSON

46. Pejchal R, Doores KJ, Walker LM, Khayat R, Burton DR, Wong CH, Wilson IA. 2005.
Huang PS, Wang SK, Stanfield RL, Julien JP, Dissection of the carbohydrate specificity of
Ramos A, Crispin M, Depetris R, Katpally U, the broadly neutralizing anti-HIV-1 antibody
Marozsan A, Cupo A, Maloveste S, Liu Y, 2G12. Proc Natl Acad Sci USA 102:1337213377.
McBride R, Ito Y, Sanders RW, Ogohara C, 52. Krause JC, Ekiert DC, Tumpey TM, Smith
Paulson JC, Feizi T, Scanlan CN, Wong CH, PB, Wilson IA, Crowe JE Jr. 2011. An
Moore JP, Olson WC, Ward AB, Poignard P, insertion mutation that distorts antibody bind-
Schief WR, Burton DR, Wilson IA. 2011. A ing site architecture enhances function of a
potent and broad neutralizing antibody recog- human antibody. MBio 2:e00345e00310.
nizes and penetrates the HIV glycan shield. 53. Dreyfus C, Laursen NS, Kwaks T, Zuijdgeest
Science 334:10971103. D, Khayat R, Ekiert DC, Lee JH, Metlagel Z,
47. Ghiara JB, Stura EA, Stanfield RL, Profy AT, Bujny MV, Jongeneelen M, van der Vlugt R,
Wilson IA. 1994. Crystal structure of the Lamrani M, Korse HJ, Geelen E, Sahin O,
principal neutralization site of HIV-1. Science Sieuwerts M, Brakenhoff JP, Vogels R, Li
264:8285. OT, Poon LL, Peiris M, Koudstaal W, Ward
48. Wyatt R, Kwong PD, Desjardins E, Sweet AB, Wilson IA, Goudsmit J, Friesen RH.
RW, Robinson J, Hendrickson WA, Sodroski 2012. Highly conserved protective epitopes
JG. 1998. The antigenic structure of the HIV on influenza B viruses. Science 337:13431348.
gp120 envelope glycoprotein. Nature 393:705711. 54. Saini SS, Allore B, Jacobs RM, Kaushik A.
49. Huber M, Le KM, Doores KJ, Fulton Z, 1999. Exceptionally long CDR3H region with
Stanfield RL, Wilson IA, Burton DR. 2010. multiple cysteine residues in functional bovine
Very few substitutions in a germ line antibody IgM antibodies. Eur J Immunol 29:24202426.
are required to initiate significant domain 55. Wang F, Ekiert DC, Ahmad I, Yu W, Zhang Y,
exchange. J Virol 84:1070010707. Bazirgan O, Torkamani A, Raudsepp T,
50. Calarese DA, Scanlan CN, Zwick MB, Deechongkit Mwangi W, Criscitiello MF, Wilson IA, Schultz
S, Mimura Y, Kunert R, Zhu P, Wormald MR, PG, Smider VV. 2013. Reshaping antibody diver-
Stanfield RL, Roux KH, Kelly JW, Rudd PM, sity. Cell 153:13791393.
Dwek RA, Katinger H, Burton DR, Wilson IA. 56. Kraulis PJ. 1991. MOLSCRIPT: a program to
2003. Antibody domain exchange is an immuno- produce both detailed and schematic plots of
logical solution to carbohydrate cluster recogni- protein structures. J Appl Crystallogr 24:946950.
tion. Science 300:20652071. 57. Merritt EA, Bacon DJ. 1997. Raster3D: photo-
51. Calarese DA, Lee HK, Huang CY, Best MD, realistic molecular graphics. Methods Enzymol
Astronomo RD, Stanfield RL, Katinger H, 277:505524.
The Role of Complement in Antibody
Therapy for Infectious Diseases

PETER P. WIBROE,1 SHEN Y. HELVIG,1 and S. MOEIN MOGHIMI1


4
INTRODUCTION

The complement system is an integral and evolutionarily ancient component


of the innate immune system, serving as the first line of defense against
common pathogens (Fig. 1) (1). The prime functions of complement in innate
host defense are accomplished through three effector pathways. These
include lysis, inflammation, and opsonization (Fig. 2). The latter is central
to microbial recognition and clearance by phagocytic cells. Complement
further cooperates with Toll-like receptors in response to microbial structure
and infection, in which immune responses are determined through both
synergistic and antagonistic manners (1). Complement is also a functional
bridge between innate and adaptive immunity, orchestrating an integrated
host defense response to pathogenic challenges. For instance, complement
can modulate adaptive immunity by providing signals that reinforce humoral
responses to antigens by priming and regulating T cells and lowering the B-
cell activation threshold (2, 3). It is also an important integral point for cross
talk with other biological cascades to ensure homeostasis is maintained. One
example is the interplay between the complement and coagulation cascades,

1
Centre for Pharmaceutical Nanotechnology and Nanotoxicology, Department of Pharmacy, Faculty of Health
and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen , Denmark.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0015-2014

63
64 WIBROE ET AL.

FIGURE 1 Schematic representation of complement activation by pathogens. The diagram shows the role
of surface-bound antibodies and other complement-sensing molecules in complement triggering. Serum
IgA is monomeric, but IgA in secretions is dimeric. IgM is pentameric. P, properdin; AP, alternative
pathway. doi:10.1128/microbiolspec.AID-0015-2014.f1

whereby the complement system can ampli- In addition to effector functions, comple-
fy coagulation by enhancing local clotting ment also plays a vital role in the maintenance
and, as a result, preventing microbial spread of homeostasis by aiding in the removal of cell
through the systemic circulation. Likewise, debris and aging cells from the host in a
the activated clotting factor XII can activate noninflammatory manner. The safe removal
the classical complement pathway and of endogenous debris is tactful, whereby
thrombin can directly cleave the third and apoptotic cells are opsonized without down-
fifth complement components (C3 and C5, stream amplification that would result in
respectively) (4, 5). inflammation, thus ensuring cell integrity and

FIGURE 2 Antibody-mediated pathogen attack and elimination mechanisms. doi:10.1128/microbiolspec.


AID-0015-2014.f2
CHAPTER 4 The Role of Complement in Antibody Therapy 65

maintenance of homeostasis (6). Complement effector cells to the site of interest. This may
also plays regulatory roles in organ regenera- result in pathogen engulfment via Fc receptors
tion, neuroprotection, and the mobilization of (FcRs), complement receptors (CRs), and/or
hematopoietic stem progenitor cells from the cytokine release. The latter process is known
bone marrow (6). as Ab-dependent cell cytotoxicity (ADCC) and
On account of its potency, selectivity, and may involve natural killer cells, macrophages,
rapid amplifying nature, complement plays a neutrophils, and eosinophils. Finally, bound
central role in the field of antibody (Ab) Abs recruit the complement components that
therapy. A promising future has been forecast kill the cell either directly by complement-
for Abs in the treatment of many pathologies, dependent lysis (complement-dependent
including cancer and infectious diseases, cytotoxicity [CDC]) or through C1q and/or
owing to their targeting properties and effi- C3b/iC3b opsonization processes that activate
cient use of inherent elimination procedures. leukocytes (complement-dependent cell
However, there are only a few drugs on the cytotoxicity [CDCC]).
market and in clinical trials for the treatment The advantage of Ab therapy is in its
of infectious diseases, mainly due to competi- targeting properties. A preparation of MAbs
tion pressure from cheaper broad-spectrum consisting of a single purified Ab with only one
antibiotics (7) and the crucial need for correct epitope may target specific infectious agents
early diagnosis. Consequently, Ab therapy is and leave host resident flora cells undisturbed.
unlikely to outcompete treatments for diseases The response development from Ab binding to
where cheaper and efficient treatments are pathogen killing or elimination is, however,
widely accessible. Nevertheless, emerging an- complex. Accordingly, important consider-
timicrobial drug resistance affirms the need ations on the effector mechanisms must be
for more targeted treatments, as in methicillin- made to achieve the full potential of the
resistant Staphylococcus aureus (8), where Ab formulation, where complement plays a cen-
treatments could have potential. tral role.
Anti-infectious Ab therapy targets either
the pathogen or its toxins. Currently, there is
only one approved anti-infectious monoclonal COMPLEMENT ACTIVATION
antibody (MAb) on the market (palivizumab)
for the treatment and prevention of respirato- The complement system consists of more than
ry syncytial virus infection in infants (9). A 30 circulating and cell-bound proteins (11). This
further few are currently in clinical trials (7). array of proteins is organized into a hierarchy of
The highly complex nature of Ab binding in proteolytic cascades operating through three
affinity and effector functions is another major distinct pathways: the classical, lectin,
challenge in the development of new and and alternative pathways (Fig. 1). These
more-efficient therapeutic Abs. This was the pathways all converge at the step where C3 is
case for motavizumab, an intended new and cleaved by C3 convertases, thereby amplifying
improved version of palivizumab. However, complement response and propagating the
clinical trials were disappointing both in the cascade.
lack of efficacy and in initiating adverse
reactions in some patients (7).
Classical Pathway
Abs mediate pathogen elimination through
different effector mechanisms (Fig. 2). Surface The classical pathway is primarily Ab depen-
neutralization is one approach, where bound dent and is initiated by antigen-Ab complexes.
Abs form a barrier that passivates the invading Only immunoglobulin G (IgG) clusters and IgM
pathogen independent of Fc isotype (10). are capable of initiating the classical pathway
Moreover, bound Abs can directly recruit (4), as discussed below. Complement compo-
66 WIBROE ET AL.

nent C1q can also act as a pattern recognition convertase cleaves C3 to give C3a and C3b.
molecule that can recognize and bind to The latter can attach to pathogen or host cells,
microbial and apoptotic cells without the aid where covalently bound C3b binds factor B,
of Abs, thus activating the classical pathway which in turn is rapidly cleaved by factor D,
directly (12). C1q has a hexameric structure, forming surface-bound C3bBb and aiding de-
which is formed by six globular heads that are position of many molecules of C3b on the
held together by a collagen-like tail. Together surface. Properdin, a positive complement
with two other proteins, C1r and C1s, they regulator, stabilizes the C3bBb convertase,
form the C1 complex. Upon binding of more extending its half-life severalfold (14). On host
than one globular head of the C1q to the cells, complement regulatory proteins such as
constant regions of IgG/IgM or directly to a CR1 and decay-accelerating factor can displace
pathogen surface, C1q undergoes a conforma- Bb from C3b. Factor H is also recruited and
tional change that can activate zymogens C1r accelerates Bb displacement from C3b. In
and C1s, forming an enzymatically active C1 addition to these processes, CR1, membrane
complex that cleaves the C2 and C4 proteins to cofactor of proteolysis, and factor H catalyze
form the classical pathway C3 convertase, the cleavage of bound C3b by recruiting the
C4b2a. plasma protease factor I to produce inactive
C3b.
C3 convertases from all three pathways
Lectin Pathway
can cleave C3 into C3b and C3a compo-
The lectin pathway is triggered when mannose- nents, with the deposits of C3b quickly
binding lectin (MBL) and ficolins bind to a forming new C3 convertases in the presence
surface. These species recognize repeating of factor B and D through the alternative
carbohydrate patterns (e.g., mannose and N- pathway. This positive reinforcement turns
acetylated sugars) on invading pathogens. Host into a large amplification loop, responsible
cells also display carbohydrate units, but these for 80 to 90% of complement response,
are protected by sialic acid, which prevents the independent of the original pathway (6).
binding of MBL (13). The binding of MBL or
ficolin to a surface triggers activation of their
associated serine protease zymogens (man- AB-MEDIATED COMPLEMENT
nose-activating serine protease 1 [MASP-1], ACTIVATION
MASP-2, and MASP-3). Activated MASP-2
cleaves C4, which in turn results in C2 cleavage The three Ab subtypes, IgA, IgG, and IgM,
and formation of a C3 convertase identical to have shown complement-activating proper-
that of the classical pathway. ties (Fig. 1). Collectively, they are able to
activate all three complement pathways,
with IgG-mediated classical pathway activa-
Alternative Pathway
tion standing as the most essential. Common
The alternative pathway is spontaneously acti- to all isotypes is the requirement of a tight
vated at a low but constant rate (tick over). This interaction between the Ab and a surface (e.
is also a mechanism that provides an ongoing g., a cell membrane, allograft, or drug carrier
probing of the surrounding cells. There are surface). This binding induces a conforma-
abundant amounts of circulating C3 in the host, tional change in the Fc/Fab hinge regions or
which is spontaneously hydrolyzed to C3bH2O. associates the Abs into a spatial orientation
This exposes a binding site for factor B. Upon that allows the complement proteins (e.g.,
binding, factor B is cleaved by factor D, forming C1q, C3b) to become both bound and acti-
the alternative pathway fluid-phase C3 vated. The conformational change is vital for
convertase C3bBb. The resulting soluble complement activation. For example, IgG
CHAPTER 4 The Role of Complement in Antibody Therapy 67

can bind to some particulate materials (e.g., IgM


SiO2 particles), but the binding may not be
Another activator of the classical pathway is the
strong enough to accomplish the conforma-
IgM Ab, which possesses an activation mech-
tional change necessary for efficient com-
anism similar to that of IgG. Secreted IgMs
plement activation (15). IgMs also require
circulate in its inactive, planar pentameric
specific conformations for activation, as they
conformation. When efficiently bound to a
have been shown to bind equally well on
target surface, the pentamer adopts a staple
different-sized dextran nanoparticles but
conformation by kinking its Fab regions relative
only induce complement activation when a
to the Fc plane. This change in conformation
distinct curvature criteria is fulfilled (16).
exposes binding sites for C1q and initiates a
complement response. The fact that a single
IgG pentamer is sufficient to initiate complement
further removes the requirement for specific Ab
IgG is the most abundant immunoglobulin
densities. Accordingly, IgM species could make
isotype and is believed to be the most important
promising candidates in MAb therapies, pro-
in complement activation. It is responsible for
vided that the production of stable expression of
the linkage between the diversification of the
the native pentameric form can be achieved on a
adaptive system and the rapid response of
large scale (19).
complement, as IgG can bind to epitopes on
nonself surfaces, resulting in recruitment and
activation of C1q via the classical pathway. The
IgA
active conformation of C1q is achieved by the
binding of more than one C1q head group to The role of IgA in complement activation
Fcg domains of surface-bound IgGs. Multiple is less studied, though IgA can activate
IgGs must therefore be bound to a surface in a complement through different pathways. It
proper intermolecular distance to be able to activates the lectin pathway mainly by acting
initiate a complement response. as a ligand for MBL (20). Furthermore, it is
There are four subtypes of IgG (IgG1 to also believed to increase the alternative
IgG4), each with different biological activities pathway turnover (21, 22). Its complement-
(C1q and FcR affinity) and pharmacokinetic activating properties largely depend on its
profiles (half-life and protease susceptibility) multimeric state and its extent of glycosyl-
(17). IgG1 is the isotype used in the majority of ation, with the most potent activators being
MAbs because of its ability to strongly associate the polymeric and heavily glycosylated spe-
with both C1q and FcgR while possessing good cies. Polymeric IgA is also a ligand for the
circulation half-lives and stability. IgG3 has a FcaR present on phagocytes and has thereby
similar biological activity, only with a reduced shown, in concert with its complement-
half-life, which is most likely due to a longer activating properties, to mediate phagocyto-
hinge region that renders the Ab susceptible to sis of Streptococcus pneumoniae (21). This
proteolysis. Despite the lack of biological activ- elimination mechanism is highly similar to
ity from IgG2 and IgG4, these isotypes have the IgG-mediated activation through FcgR
shown superior bactericidal function against affinity. However, where IgG can induce
Cryptococcus neoformans infections in mice phagocytosis directly through FcgR in the
(18). This finding highlights the complex effec- absence of complement activation, IgA-me-
tor function in Ab therapies in general and diated killing is dependent on phagocyte
suggests specific demands for different infec- preactivation by C5a or tumor necrosis
tions. It further questions the general role of factor alpha (21). Its relatively passive in-
complement in Ab therapies and is discussed flammatory nature is convenient, given the
below. high abundance of IgA at mucosal sites,
68 WIBROE ET AL.

where a continuous exposure to microorga- associates with C6 and C7 components and


nisms and foreign molecules would other- partitions into a lipid bilayer membrane.
wise cause host cell damage. Subsequently, C8 binds to the complex,
After response initiation and coating by which then induces the binding and poly-
active complement complexes on the Fc re- merization of 10 to 16 C9 components,
gion, the IgA1 isotype has been reported to be producing a channel-like structure that
able to detach from the antigenic surface while penetrates the cellular membrane, known
retaining its binding properties toward new as the membrane attack complex (MAC).
antigens. This phenomenon is termed comple- Water, electrolytes, and even enzymes can
ment-coated Ab transfer and is believed to be freely pass the channel, which leads to a
an efficient mechanism for transferring acti- disruption of cell homeostasis and cell lysis
vated complement compounds to nearby anti- (12).
gens without the need to initialize a new MAbs engineered to target specific patho-
response (23). gens can direct a complement activation
Coadministration of IgA and IgG has been leading to the development of the MAC,
shown to induce tumor cell killing by poly- thereby lysing the targeted cells by taking
morphonuclear cells by activating their re- advantage of the bodys own defense system.
spective Fc receptors simultaneously (24). On This action forms the basic mechanism of
the other hand, IgAs affinity to bacterial CDC and is believed to play a major role in
capsular polysaccharides has (paradoxically) Ab-based therapy of certain hematological
been reported to sterically block IgG binding malignancies and solid tumors (27, 28) and a
and the following inflammatory functions (25, less-defined role in elimination of pathogens,
26), thus questioning the robustness of the especially bacterial toxins and viruses (7).
IgA/IgG synergistic effects.
Opsonization
EFFECTOR RESPONSES AND THEIR ROLE The large amounts of opsonins, in particular
IN MAB TREATMENTS C3b produced from the amplification loop,
efficiently coat the surface of the pathogen,
Following the detection of a pathogenic sur- tagging it for recognition by neutrophils, NK
face and initiation of one of the three cells, and monocytes carrying the CRs and C1q
pathways, there are three main effector func- receptors. But as opposed to Ab-mediated cell
tions that contribute to pathogen removal. activation through FcRs, CRs do not internal-
These functions and their effect on MAb ize particles unless they are costimulated by
treatments are outlined below. external factors like tumor necrosis factor
alpha, colony-stimulating factors, or the com-
plement anaphylatoxins C3a and C5a. Further-
MAC and CDC
more, CR-mediated phagocytosis does not
As the complement activation amplifies and release inflammatory mediators like reactive
propagates, the density of C3b increases oxygen intermediates and arachidonic acid
dramatically, which leads to the binding of metabolites (29). This may be a reason why
C3b to already formed C3 convertases, CDCC is believed to have limited influence in
producing the C5 convertase [C4b2a3b, clinically available immunotherapies (30).
(C3b)2Bb]. This permits complement acti- The polysaccharide b-glucan can enhance
vation to enter the terminal phase, where all CR3-dependent cellular cytotoxicity, some-
three pathways converge. C5 convertase times referred to as CR3-DCC (31). It is
cleaves C5 into C5a and C5b. C5a is a potent naturally present on cell walls of yeasts and
soluble anaphylatoxin, whereas C5b quickly fungi, but its coadministration in MAb therapy
CHAPTER 4 The Role of Complement in Antibody Therapy 69

targeting surfaces inherently absent of the complement. However, since most of the
molecule has been shown to induce cyto- applied Abs are of the IgG1 isotype that has
toxicity (32). Activation of complement also the potential to activate complement, and has
facilitates the adaptive immune system via the been shown to do so in vitro, their clinical
binding of opsonized antigens to CR2 on B efficiency may be improved by providing the
cells, which activates specific Ab production proper conditions for a complement response
and the differentiation of B memory cells. development, as suggested before (31). The
Thus, complement does not only become substantial amount of experience within CDC
activated by Abs, but may direct Ab production and complement initiation should provide
against C3b-bound surfaces. Clearly C3b is a approaches to strategically manipulate the
key molecule in translating complement acti- complement system to improve the efficacy
vation into effector responses (14). of immunotherapies, either by improving CDC
or by providing synergistic effects between
CDC and ADCC. Different approaches can
Inammatory Responses
be applied, as briefly outlined below.
The potent anaphylatoxins C3a and C5a are
capable of inducing an inflammatory state
Increasing Complement Response
by degranulation of mast cells and basophil
granulocytes, giving rise to vasodilation and Since Ab density is an important parameter
capillary leakage (14). Furthermore, they for proper complement activation, choosing
are strong chemoattractants that guide the the right conserved epitope or introducing a
migration of circulating neutrophils, mono- cocktail of MAbs with different epitope
cytes, and macrophages to the site of com- specificities may not only avoid escape
plement activation, exerting an important variants of the targeted pathogen but also
role in indirectly facilitating the interaction result in improved complement effector
between opsonins and phagocytes (4). These functions in addition to potential effects of
powerfulbioactivefragmentsarequicklycleaved neutralizing properties (19, 36). In general,
by carboxypeptidases to give C5a-desArg and simulating a controlled pool of polyclonal
C3a-desArg.C5a-desArgretainsupto10%ofthe Abs is believed to have several beneficial
C5ainflammatoryproperties,whileC3a-desArg roles in terms of broader specificity, poten-
isdevoidofproinflammatoryfunctions(33). cy, and robustness and may target groups of
Though not directly responsible for patho- infectious agents that are common for spe-
gen clearance, the complement anaphylatoxins cific diseases or exposures (19, 36). The
with their effector cell-activating and chemo- prevalence of infectious diseases is com-
attractive properties are important mediators monly increased in the case of a dysfunc-
in any cell-mediated immune response, includ- tional or immature immune system, and the
ing Ab-mediated FcR activation (i.e., ADCC). effector functions expected to accompany
An activated complement response thereby has Ab therapies are similarly compromised.
the potential to act synergistically with ADCC, Immune function assessment and coadmin-
especially by the C5a-dependent upregulation istration of complement sources or adjuvants
of activating FcgRIII on macrophages (34, 35). could therefore be necessary to achieve ex-
pected drug effects (7).
A central property of the complement
MANIPULATING AB-MEDIATED system is its fast-acting nature. Due to its
COMPLEMENT RESPONSES efficient amplification, a response rapidly
develops and exerts its function. Within a
Many of the MAbs already in clinical use relatively short period, the plasma becomes
do not seem to exploit the full potential of drained for native complement proteins, and
70 WIBROE ET AL.

the constant activity of the nonconsumed have positive effects of the recruitment and
regulatory proteins ensures down-regulation activation of phagocytes necessary for ADCC,
of the response. Indeed, in the case of ritux- but recent studies with cancer MAb therapies
imab infusions against chronic lymphocytic have questioned the positive contributions
leukemia (CLL), the least-abundant comple- from complement. In an in vitro study (42),
ment plasma protein, C2, is rapidly consumed the activation of complement was found to
and is the limiting factor of rituximab cell- inhibit NK cell-mediated cytotoxicity brought
killing efficiency (37). The same group demon- on by ADCC. This inhibition was due to a C3b-
strated that boosting the rituximab-initiated dependent interference of the binding of NK
complement response by administering a sec- cells to rituximab, thus preventing the activa-
ondary Ab toward cell-bound C3b and its tion of NK cells. In the same study, the
breakdown products greatly enhanced C3b depletion of C3b components in an in vivo
coating (38). Based on these findings, it was model was able to enhance the ability of
recently shown that the effect of rituximab on rituximab-coated target cells to activate
five patients with CLL was markedly improved human NK cells and improve the efficacy of
when coadministered with fresh plasma as a the MAb. Yet another study showed that the
source of uncleaved complement proteins (39). presence of C5a in the tumor microenviron-
It should be noted that CLL is well associated ment could suppress the antitumor CD8+ T-
with low levels of complement proteins, espe- cell-mediated response, which would normally
cially classical pathway components and pro- recruit myeloid-derived suppressor cells into
perdin. However, since long, repetitive tumors and augment T-cell-directed suppres-
infusions are not uncommon, consumption of sive abilities (43). The same study showed that
complement proteins is likely to occur, deficiency of complement in mice was coupled
resulting in a reduced therapeutic efficiency with a hindered tumor growth, a suppression
and increased risk of infectious diseases. that was comparable to pharmacologically
Furthermore, most MAb therapies target ex- blocking C5a receptors (C5aRs). The inhibition
travascular spaces, where complement protein of C5aR signaling was also associated with
levels may be reduced or unevenly distributed enhanced CD8+ T cell antitumor response (43).
compared to plasma. This may explain why Since CDC depends on late-stage complement
many in vitro experiments performed in serum development, attempts that block complement
show high complement contributions, whereas activation or deficiency of complement pro-
in vivo data are less convincing. If target cells teins in specific target tissues or in some
are located in extravascular spaces, it is individuals is likely to reduce ADCC without
therefore imperative to evaluate the availability gaining any effect from CDC. Accordingly,
of complement proteins and potentially con- complete blocking of complement may not be
sider coadministration of a complement pro- a plausible approach in antimicrobial therapies
tein source at the target site (37, 40). with Abs.

Reducing Negative Effects


COMPLEMENT EVASIVE PROPERTIES
from Complement
OF PATHOGENS
The mechanism of action of MAb therapies
is likely to be a combination of ADCC, CDC, Pathogens generally lack complement regula-
and CDCC (Fig. 2). However, the exact tory proteins and are susceptible to opson-
nature of their interaction, i.e., whether it is ization and complement attack. However,
synergistic, additive, or antagonistic, re- millions of years of evolution have allowed
mains uncertain (27, 41). Activated comple- pathogenic microorganisms to develop sophis-
ment components would be expected to ticated strategies to dampen or even overcome
CHAPTER 4 The Role of Complement in Antibody Therapy 71

complement activity (1, 44, 45, 46). Some have ment activation may overcome the pathogens
successfully employed surface strategies to immune evasive properties.
mask or camouflage antigenic components on
their surfaces, while others can express surface
proteins to bind to, recruit, or mimic host CONCLUSIONS AND FUTURE
complement regulators (44). It is not unusual PERSPECTIVE
for pathogens to apply several defense strate-
gies, exploiting complement cascades at differ- The complement system protects the host
ent stages (47). Staphylococcus aureus is an from pathogenic invaders in a very effective
excellent example of a pathogen that can evade manner. Although defined as a part of the
several different points of a complement attack innate immune system, there is a large overlap
by preventing activation, degrading opsonin area shared with the adaptive effector func-
C3b, and inhibiting C3 convertase, thereby tions. Complement is able to induce inflam-
preventing downstream complement effector mation responses that permit the functions of
responses (44). This pathogen also expresses the adaptive system as well as stimulate B-cell-
staphylococcal protein A (SpA) with the ability specific Ab production and differentiation of
to bind to the Fc fragments of IgGs, thereby memory B cells. Likewise, target-bound Abs
reducing the FcR-mediated phagocytosis (48). can act as pattern recognition molecules and
Other mechanisms include the expression of activate complement. Further, it interacts
adhesins by pathogens to anchor to host cell dynamically with multiple systems, which
surfaces or to present invasin to mediate uptake highlights the complexity of both the biolo-
into host cells (49). gical system and the complement system. The
All surface strategies serve the purpose of poor understanding of this complexity has led
avoiding recognition, opsonization, and clear- to past failures in the pharmaceutical devel-
ance by host complement response, which lead opment in the field. A particular challenge
to the survival of pathogens in an otherwise lies in the in vitro/in vivo correlation, in
immunocompetent host (4, 49). Over and which theory obtained from the linear and
above, mutating or new merging pathogenic often well-controlled experimental environ-
microorganisms persist to accomplish new ments fails to apply to in vivo performances,
ways to counterstrike complement defense, where multiple systems are at play. For
presenting a real challenge for the host and example, it might be difficult to predict the
the application for MAb treatments against biological response in tissues where certain
certain infectious diseases (5). effector proteins are dysregulated or in immu-
Persisting malignant cells have the inherent nocompromised patients.
ability to express high levels of endogenous Our immune system is in constant battle
membrane-bound complement regulators, with invading and opportunist pathogens.
including CD46, decay-accelerating factor, A lowering of this guard, for example, in
and CD59, which efficiently prevent com- immune-compromised individuals, could
plement amplification and MAC formation. have consequences that might require phar-
They can further block complement activa- maceutical interventions. However, the pres-
tion by recruiting fluid-phase complement ent anti-infectious development is not
regulators, contributing to the evasion of advancing at the same pace as the rapid
complement surveillance that leads to pro- emerging strategies of pathogens to counter-
gression in malignant growth (4). However, strike both the immune system and the
considering the success of CDC-dependent current broad-spectrum antibiotic treatments.
MAb treatments against malignant cells with On the other hand, particulate nanoparticles
upregulated complement regulators, it is plau- are receiving increasing attention to enhance
sible to speculate that a well-targeted comple- the immunogenicity of subunit vaccines
72 WIBROE ET AL.

through both antigen protection and targeting 3. Mastellos D, Germenis AE, Lambris JD. 2005.
to antigen-presenting cells as well as Complement: an inflammatory pathway fulfilling
multiple roles at the interface of innate immunity
immunostimulation (50). The latter may in- and development. Curr Drug Targets Inflamm
volve the complement system (for instance, Allergy 4:125127.
some complement activation products can 4. Markiewski MM, Nilsson B, Nilsson-Ekdahl
induce B cell activation) or direct activation K, Mollnes TE, Lambris JD. 2007. Comple-
ment and coagulation: strangers or partners in
of the NALP3 inflammasome complex (apo-
crime? Trends Immunol 28:184192.
ptosis-associated speck-like protein and 5. Rittirsch D, Flierl MA, Ward PA. 2008.
caspase 1 protease), which in turn cleave and Harmful molecular mechanisms in sepsis. Nat
activate the immunostimulatory cytokine in- Rev Immunol 8:776787.
terleukin 1b (51). 6. Zipfel PF, Skerka C. 2009. Complement regulators
Ab treatment holds the potential to open and inhibitory proteins. Nat Rev Immunol 9:729
740.
a new approach to fight infectious diseases.
7. ter Meulen J. 2011. Monoclonal antibodies
Properties like direct targeting and selec- in infectious diseases: clinical pipeline in 2011.
tivity, even for mutant strains, should be Infect Dis Clin N Am 25:789802.
taken advantage of. However, issues such as 8. Saylor C, Dadachova E, Casadevall A. 2009.
efficiency and precision of diagnosis, cost of Monoclonal antibody-based therapies for micro-
bial diseases. Vaccine 27(Suppl 6):G38G46.
drug discovery, and the lack of comprehen-
9. Krilov LR. 2011. Respiratory syncytial virus
sive understanding of the mode of action of disease: update on treatment and prevention.
development candidates signify that much Expert Rev Anti Infect Ther 9:2732.
more attention is needed to succeed in 10. Martnez I, Melero JA. 1998. Enhanced neutral-
building this new platform. When their ization of human respiratory syncytial virus by
potentials are fully explored, they would mixtures of monoclonal antibodies to the attach-
ment (G) glycoprotein. J Gen Virol 79:22152220.
definitely provide a promising future for
11. Moghimi SM, Hamad I. 2008. Liposome-
complement in Ab treatments. mediated triggering of complement cascade.
J Liposome Res 18:195209.
ACKNOWLEDGMENTS 12. Murphy K, Travers P, Walport M. 2008.
Janeways Immunobiology, 7th ed. Garland
Financial support from the Danish Agency Science, New York, NY.
for Science, Technology, and Innovation 13. Favoreel HW, Van de Walle GR, Nauwynck
HJ, Pensaert MB. 2003. Virus complement
(Det Strategiske Forskningsrd), reference
evasion strategies. J Gen Virol 84:115.
09-065746, is gratefully acknowledged. 14. Ehrnthaller C, Ignatius A, Gebhard F, Huber-
Conflicts of interest: We declare no Lang M. 2011. New insights of an old defense
conflicts. system: structure, function, and clinical relevance
of the complement system. Mol Med 17:317329.
15. Sellborn A, Andersson M, Hedlund J,
CITATION
Andersson J, Berglin M, Elwing H. 2005.
Wibroe PP, Helvig SY, Moghimi SM. 2014. Immune complement activation on polystyrene
and silicon dioxide surfaces. Impact of reversible
The role of complement in antibody therapy
IgG adsorption. Mol Immunol 42:569574.
for infectious diseases. Microbiol Spectrum 16. Pedersen MB, Zhou XF, Larsen EKU, Sorensen
2(2):AID-0015-2014. US, Kjems J, Nygaard JV, Nyengaard JR, Meyer
RL, Boesen T, Vorup-Jensen T. 2010. Curvature
of synthetic and natural surfaces is an important
REFERENCES
target feature in classical pathway complement
1. Ricklin D, Hajishengallis G, Yang K, Lambris JD. activation. J Immunol 184:19311945.
2010. Complement: a key system for immune sur- 17. Salfeld JG. 2007. Isotype selection in antibody
veillance and homeostasis. Nat Immunol 11:785797. engineering. Nat Biotechnol 25:13691372.
2. Carroll MC. 2004. The complement system in 18. Beenhouwer DO, Yoo EM, Lai C-W, Rocha
regulation of adaptive immunity. Nat Immunol MA, Morrison SL. 2007. Human immunoglob-
5:981986. ulin G2 (IgG2) and IgG4, but not IgG1 or IgG3,
CHAPTER 4 The Role of Complement in Antibody Therapy 73

protect mice against Cryptococcus neoformans 32. Vetvicka V, Thornton BP, Ross GD. 1996.
infection. Infect Immun 75:14241435. Soluble beta-glucan polysaccharide binding to
19. Berry JD, Gaudet RG. 2011. Antibodies in infec- the lectin site of neutrophil or natural killer
tious diseases: polyclonals, monoclonals and niche cell complement receptor type 3 (CD11b/CD18)
biotechnology. Nat Biotechnol 28:489501. generates a primed state of the receptor capa-
20. Roos A, Bouwman LH, van Gijlswijk-Janssen ble of mediating cytotoxicity of iC3b-opsonized
DJ, Faber-Krol MC, Stahl GL, Daha MR. target cells. J Clin Investig 98:5061.
2001. Human IgA activates the complement 33. Klos A, Tenner AJ, Johswich KO, Ager RR,
system via the mannan-binding lectin pathway. Reis ES, Kohl J. 2009. The role of the anaphyl-
J Immunol 167:28612868. atoxins in health and disease. Mol Immunol
21. Janoff EN, Fasching C, Orenstein JM, Rubins 46:27532766.
JB, Opstad NL, Dalmasso AP. 1999. Killing of 34. Shushakova N, Skokowa J, Schulman J,
Streptococcus pneumoniae by capsular polysac- Baumann U, Zwirner J, Schmidt RE, Gessner
charide-specific polymeric IgA, complement, JE. 2002. C5a anaphylatoxin is a major regulator
and phagocytes. J Clin Investig 104:11391147. of activating versus inhibitory FcgammaRs in
22. Hiemstra PS, Gorter A, Stuurman ME, Van immune complex-induced lung disease. J Clin
Es LA, Daha MR. 1987. Activation of the Investig 110:18231830.
alternative pathway of complement by human 35. Schmidt RE, Gessner JE. 2005. Fc receptors
serum IgA. Eur J Immunol 17:321326. and their interaction with complement in auto-
23. Boackle RJ, Nguyen QL, Leite RS, Yang X, immunity. Immunol Lett 100:5667.
Vesely J. 2006. Complement-coated antibody- 36. Logtenberg T. 2007. Antibody cocktails: next-
transfer (CCAT); serum IgA1 antibodies inter- generation biopharmaceuticals with improved
cept and transport C4 and C3 fragments and potency. Trends Biotechnol 25:390394.
preserve IgG1 deployment (PGD). Mol Immunol 37. Kennedy AD, Beum PV, Solga MD, DiLillo
43:236245. DJ, Lindorfer MA, Hess CE, Densmore JJ,
24. van Egmond M, van Spriel AB, Vermeulen H, Williams ME, Taylor RP. 2004. Rituximab
Huls G, van Garderen E, van de Winkel JG. infusion promotes rapid complement depletion
2001. Enhancement of polymorphonuclear and acute CD20 loss in chronic lymphocytic
cell-mediated tumor cell killing on simulta- leukemia. J Immunol 172:32803288.
neous engagement of fcgammaRI (CD64) and 38. Kennedy AD, Solga MD, Schuman TA, Chi AW,
fcalphaRI (CD89). Cancer Res 61:40554060. Lindorfer MA, Sutherland WM, Foley PL, Taylor
25. Jarvis GA, Griffiss JM. 1991. Human IgA1 RP. 2003. An anti-C3b(i) mAb enhances comple-
blockade of IgG-initiated lysis of Neisseria men- ment activation, C3b(i) deposition, and killing of
ingitidis is a function of antigen-binding fragment CD20+ cells by rituximab. Blood 101:10711079.
binding to the polysaccharide capsule. J Immunol 39. Klepfish A, Gilles L, Ioannis K, Rachmilewitz
147:19621967. EA, Eliezer R, Schattner A, Ami S. 2009.
26. Griffiss JM. 1975. Bactericidal activity of Enhancing the action of rituximab in chronic
meningococcal antisera. Blocking by IgA of lymphocytic leukemia by adding fresh frozen
lytic antibody in human convalescent sera. plasma: complement/rituximab interactions
J Immunol 114:17791784. and clinical results in refractory CLL. Ann N
27. Weiner LM, Surana R, Wang S. 2010. Mono- Y Acad Sci 1173:865873.
clonal antibodies: versatile platforms for cancer 40. Wang S-Y, Veeramani S, Racila E, Cagley J,
immunotherapy. Nat Rev Immunol 10:317327. Fritzinger DC, Vogel C-W, St John W, Weiner
28. Scott AM, Wolchok JD, Old LJ. 2012. Antibody GJ. 2009. Depletion of the C3 component of
therapy of cancer. Nat Rev Cancer 12:278287. complement enhances the ability of rituximab-
29. Wright SD, Silverstein SC. 1983. Receptors coated target cells to activate human NK cells
for C3b and C3bi promote phagocytosis but not and improves the efficacy of monoclonal anti-
the release of toxic oxygen from human phago- body therapy in an in vivo model. Blood 114:
cytes. J Exp Med 158:20162023. 53225330.
30. Zhou X, Hu W, Qin X. 2008. The role of 41. Wang S-Y, Weiner G. 2008. Complement and
complement in the mechanism of action of cellular cytotoxicity in antibody therapy of
rituximab for B-cell lymphoma: implications cancer. Expert Opin Biol Ther 8:759768.
for therapy. Oncologist 13:954966. 42. Wang SY, Veeramani S, Racila E, Cagley J,
31. Gelderman KA, Tomlinson S, Ross GD, Fritzinger DC, Vogel CW, St John W, Weiner
Gorter A. 2004. Complement function in mAb- GJ. 2009. Depletion of the C3 component of
mediated cancer immunotherapy. Trends Immunol complement enhances the ability of rituximab-
25:158164. coated target cells to activate human NK cells and
74 WIBROE ET AL.

improves the efficacy of monoclonal antibody 48. Peterson PK, Verhoef J, Sabath LD, Quie
therapy in an in vivo model. Blood 114:53225330. PG. 1977. Effect of protein A on staphylococcal
43. Markiewski MM, DeAngelis RA, Benencia F, opsonization. Infect Immun 15:760764.
Ricklin-Lichtsteiner SK, Koutoulaki A, 49. Finlay BB, McFadden G. 2006. Anti-immunol-
Gerard C, Coukos G, Lambris JD. 2008. ogy: evasion of the host immune system by
Modulation of the antitumor immune response bacterial and viral pathogens. Cell 124:767782.
by complement. Nat Immunol 9:12251235. 50. Moghimi SM. 2009. The innate immune
44. Sarma JV, Ward PA. 2011. The complement responses, adjuvants and delivery systems, p
system. Cell Tissue Res 343:227235. 113127. In Jorgensen L, Nielsen HM (ed),
45. Rooijakkers SH, van Strijp JA. 2007. Bacterial Delivery Technologies for Biopharmaceuticals.
complement evasion. Mol Immunol 44:2332. Peptides, Proteins, Nucleic Acids and Vaccines.
46. Lambris JD, Ricklin D, Geisbrecht BV. 2008. Wiley, Chichester, United Kingdom.
Complement evasion by human pathogens. Nat 51. Li H, Willingham SB, Ting JP, Re F. 2008.
Rev Microbiol 6:132142. Cutting edge: inflammasome activation by
47. Szebeni J. 2004. The Complement System. alum and alums adjuvant effects are mediated
Kluwer Academic Publisher, Boston, MA. by NLRP3. J Immunol 181:1721.
Immunoglobulin E and Allergy:
Antibodies in Immune Inammation
and Treatment

SOPHIA N. KARAGIANNIS,1 PANAGIOTIS KARAGIANNIS,1


DEBRA H. JOSEPHS,1 LOUISE SAUL,1 AMY E. GILBERT,1 NADINE UPTON,2
5
and HANNAH J. GOULD2

IMMUNOLOGICAL MECHANISMS OF ALLERGIC DISEASES AND THE


ROLE OF IGE ANTIBODIES

Clinical Perspective on Allergic Inammation


Allergic inflammation, caused by development of an allergen-induced
immune response, is largely driven via immunoglobulin E (IgE)-dependent
mechanisms. It manifests clinically as asthma, rhinoconjunctivitis (more
commonly known as hay fever), allergic skin inflammation (the main
example of which is atopic dermatitis), food allergy, urticaria, and/or
anaphylaxis, with several known disease variants caused by different
underlying cellular and molecular mechanisms (1). Increased levels of
circulating IgE, allergen-specific IgE reactivity profiles measured with
radioallergosorbent tests and positive skin prick tests for specific allergens,

1
Cutaneous Medicine and Immunotherapy Unit, St. Johns Institute of Dermatology, Division of Genetics and
Molecular Medicine & NIHR Biomedical Research Centre at Guys and St. Thomass Hospitals and Kings College
London, Kings College London School of Medicine, Guys Hospital, Kings College London, London SE1 9RT,
United Kingdom; 2Randall Division of Cell and Molecular Biophysics, Division of Asthma, Allergy, and Lung
Biology, MRC and Asthma UK Centre for Allergic Mechanisms of Asthma, Kings College London, New Hunts
House, Guys Campus, London SE1 1UL, United Kingdom.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0006-2012

75
76 KARAGIANNIS ET AL.

together with auxiliary ex vivo and in vitro numbers of activated CD4+ T helper type 2
mast cell and basophil activation functional (Th2) lymphocytes.
readouts, support the importance of IgE Production of allergen-specific IgE re-
antibodies in the clinical manifestation of quires that allergens are taken up by dendritic
allergies (2, 3). Allergic inflammation can be cells (DCs), monocytes, B cells, or other
local (that is, within the target organ), as is antigen presenting cells (APCs), which, in
the case for allergic rhinoconjunctivitis and the presence of cytokines such as interleukin-
allergic asthma, or systemic, as is the case 4 (IL-4) or IL-13 present the processed
for anaphylaxis. The etiology of allergic antigens to cognate naive T cells that then
immune responses has been shown to be acquire a Th2 cell phenotype (9). Th2 cells
influenced by several factors, including then engage cognate B cells through both B
genetic susceptibility (4), route of exposure, cell major histocompatibility complex class II
dose of the allergen, and in some cases, and costimulatory molecules and secrete IL-4
structural characteristics of the allergen (5). and IL-13, inducing B cells to undergo class-
The allergic inflammatory cascade is switch recombination, resulting in the vari-
thought to have evolved from the natural able, diverse, and joining segments that were
immune defenses to parasite and worm infec- initially linked to another constant (C) region
tions. This response can provide swift protec- in the immunoglobulin heavy chain locus to
tion against these organisms but also instead be linked to the C region (10). Class-
participates in wound healing following tissue switch recombination can also be induced by
damage accrued as a result of the infection or IL-4 and/or IL-13 derived from cells other
accumulation of toxins. The same inflammatory than Th2 cells, which may include mast cells
cascades, originally evolved to neutralize invad- and basophils (11). Allergen sensitization was
ing parasites, are activated in response to previously thought to occur primarily in
innocuous antigens such as house dust mite lymphoid germinal centers; however, IgE-
and pollen proteins in the context of allergic producing B cells that undergo clonal selec-
diseases, triggering mast cell degranulation and tion and affinity maturation can also be
eosinophil inflammation to the sites of allergen generated locally at sites of antigen challenge
challenge. Furthermore, acute protective im- such as the respiratory mucosa, gastrointes-
mune responses to infection evolve into chronic tinal tract, and lymph nodes of individuals
inflammatory cascades with long and persistent with food allergy, seasonal or perennial
exposure to allergens. The consequences are allergic rhinitis, or atopic or nonatopic asth-
persistent inflammation, tissue remodeling, and ma (11).
undesirable pathologies (6).
A Glance at IgE Receptors
IgE Antibodies Activate Immune
IgE antibodies have two known cell surface
Responses in the Presence of Allergens
receptors, FcRI (the high-affinity receptor)
The discovery and characterization of IgE, and CD23 (otherwise known as FcRII, the
culminating in the independent descriptions low-affinity receptor) (10). Each Fc receptor
of this class of antibody by Ishizaka et al. and recognizes a distinct epitope located on oppo-
Johansson and Bennich, arguably represents site sites of the Ce3 domain of the IgE constant
the most crucial advance in our understanding region, and binding of one receptor to this
of the immunological basis of allergic disorders domain prevents binding of the other to IgE
(7, 8). Allergic inflammation is characterized by (12). IgE antibodies also recognize the IgE- and
IgE-dependent activation of mast cells and an FcRI-binding protein galectin-3 (10).
infiltration of inflammatory cells to sites of FcRI, which has affinity for IgE in the
allergen challenge, orchestrated by increased nanomolar range (Ka = 108 to 1010 M1) is
CHAPTER 5 Immunoglobulin E and Allergy 77

expressed on basophils and mast cells as an Galectin-3, the beta-galactoside-binding


2 tetramer and on Langerhans cells, lectin which recognizes both IgE Fc and
myeloid DCs, plasmacytoid DCs, monocytes, FcRI, exists in a secreted form and is also
and eosinophils as an 2 trimer (13). The stored intracellularly. Its expression is as-
subunit has two extracellular immunoglob- sociated with activated IgE immune cells
ulin domains, which form the binding site of such as monocytes/macrophages, DCs, mast
IgE to the receptor, and regulates movement cells, and eosinophils and also with B cells
of the receptor from the endoplasmic retic- following stimulation by IL-4 and CD40-
ulum, where the subunits of the receptor are CD40L engagement (19).
assembled to the cell surface membrane via
an immunoreceptor tyrosine-based activa-
IgE Effector Cells and Their Roles
tion motif. The chain which is shared with
in IgE Responses
FcRs is necessary for receptor assembly and
cell surface expression, while the subunit is Mast Cells: Hallmarks of IgE-Driven
thought to enhance receptor functions and to Allergic Response
also be responsible for enhanced receptor The hallmark of an acute allergic response,
(2 tetramer) expression on the surface of mediated by allergen-IgE-FcRI complexes
mast cells and basophils. The downstream cross-linking FcRI on the surface of mast
signaling events that culminate in degranu- cells, is immediate (type I) hypersensitivity.
lation in mast cells and basophils are medi- Cross-linking can occur when the targeted
ated by the associated and subunits. antigen is multivalent, as two or more re-
The low-affinity receptor, CD23, can be ceptors need to be engaged to activate the
induced on a broad range of immune cells, ensuing signaling cascade, although the pre-
such as activated B cells, activated monocytes sence of cytokinergic IgEs capable of activat-
and macrophages, eosinophils, natural killer T ing mast cells in the absence of multivalent
cells, T cells, follicular DCs, and platelets, and antigen have been reported (20). Cross-linking
also on nonimmune cells, such as airway of IgE-FcRI complexes on the mast cell
epithelial cells and smooth muscle cells (10, surface by allergens leads, within minutes, to
14, 15). Unlike the high-affinity receptor, the the so-called early phase of the allergic
low-affinity receptor, CD23, displays a micro- response, involving mast cell degranulation
molar affinity (Ka = 107 to 108 M1) for IgE as a and the rapid release of preformed vasoactive
monomer, but trimeric fragments are released amines (mainly histamine), lipid mediators
from the cell membrane by metalloproteases (such as prostaglandin D, platelet-activating
and bind with 10 nM avidity (16). The CD23a factor, leukotriene C4 [LTC4], LTD4, and
splice variant is exclusively expressed on B LTE4), chemokines (CXC-chemokine ligand
cells activated by antigen, and it plays a key 8, CXC-chemokine ligand 10, CC-chemokine
role in antigen endocytosis, processing, and ligand 2 [CCL2], CCL4, and CCL5), and
presentation and in the regulation of IgE cytokines (such as IL-4, IL-5, and IL-13).
synthesis. Expression of the CD23b form can Mast cell activation plays a key role in
be triggered by IL-4 on a variety of effector generating the symptoms of allergy; the cyto-
cells, including B cells and macrophages. kines and chemokines liberated in this early
Engagement of CD23 by IgE-antigen com- phase initiate the late phase, which peaks
plexes is thought to induce nitric oxide some hours later and involves the recruitment
synthase and the release of cytokines such as and activation of inflammatory cells (neutro-
tumor necrosis factor alpha (TNF-) by phils, followed by eosinophils, monocytes, and
macrophages, and these events have been lymphocytes) at sites sensitive to allergen.
reported to participate in phagocytosis and Similarly, but without overt symptoms, aller-
killing of parasites (17, 18). gens activate the IgE-sensitized APCs, which
78 KARAGIANNIS ET AL.

in turn promote de novo immunoglobulin sentation (FAP). The association between


production by B cells, thereby maintaining CD23 and HLA-DR in the cell membrane is
mast cell and APC sensitization (10). involved in the trafficking of allergen-IgE-
CD23 complexes to endosomes, where the
APCs Trigger Th2 Responses allergen-derived peptides are loaded onto
and IgE Inammation the HLA-DR molecules for presentation at
Th2 cells are recruited and activated at the the B-cell surface (29). An antigen-activat-
sites of allergic inflammation. A key re- ed B cell expressing CD23 can simulta-
search focus aims to understand the mech- neously process unrelated antigens through
anisms by which allergens regulate Th2 cells FcRs and cause epitope spreading to other
through antigen presentation by professional antigens. Indeed, CD23-mediated FAP is
APCs such as DCs. It has been proposed known to be as efficient as FcR-mediated
that allergic inflammation is initiated fol- antigen presentation by DCs (30), orders of
lowing allergen-epithelial cell contact, lead- magnitude more efficient than B-cell inter-
ing to the promotion of Th2 immunity via nalization via surface immunoglobulin (the
the production of a number of cytokines. Of BCR).
key importance among these is epithelial B cells in draining lymph nodes partici-
cell-derived thymic stromal lymphopoietin, pate in allergic inflammation through IgE
which conditions DCs to favor Th2 induc- synthesis. However, they are also present in
tion (21), while other epithelial cell-derived the respiratory mucosa, and their capacity
cytokines, namely IL-25 and IL-33, are also to produce IgE locally in tissues has been
capable of inducing and maintaining Th2 demonstrated (31). Following exposure to
immunity (22, 23). In addition, by secreting allergens, production of IgE is triggered by
the chemokines CCL17 and CCL22, DCs key Th2 cytokines IL-4 and IL-13, with IL-9
possess definitive roles in the recruitment participating by enhancing IgE synthesis in
and activation of Th2 cells, while other cells situ (32, 33). Regulatory functions have also
secrete IL-4 which is necessary for Th2 cell been attributed to B cells through secretion
differentiation (24). There is increasing of IL-10, but the role of different subsets of
evidence that basophils express FcRI, B cells in allergic diseases remains to be
major histocompatibility complex class II, determined (6).
and costimulatory molecules CD80/CD86,
may participate in antigen recognition and Monocytes and Eosinophils Mediate IgE
processing, and can induce Th2 cell differ- Effector Functions and Tissue Remodeling
entiation through the release of IL-4 (25, 26, Monocytes and eosinophils are among the
27). Other cells may also possess antigen IgE effector cell types that are drawn to sites
presentation properties and can also secrete of allergic inflammation. IgE upregulates the
Th2 cytokines when activated by IL-25 and expression of FcRI, whereas IL-4 and IL-13
IL-33; these include mast cells, macro- can stimulate the expression of CD23 by
phages, eosinophils, and natural helper these cells. This in turn arms the cells for
cells (28). functions such as clearance of antigen-IgE
complexes, and killing and phagocytosis of
B Cells and CD23 Participate in pathogens (for example, helminth parasites)
Antigen Presentation and tumor cells that bear foreign antigens
Allergen-IgE complexes bound to CD23 (34, 35). In carrying out their functions,
expressed by allergen-activated B cells can monocytes and eosinophils inevitably also
facilitate antigen presentation to T cells. inflict some damage on bystander cells in the
The process of antigen presentation by way tissue. This side effect may ultimately con-
of CD23 is termed facilitated antigen pre- tribute to tissue remodeling and exacerbates
CHAPTER 5 Immunoglobulin E and Allergy 79

the symptoms of chronic asthma. The Upon multivalent antigen-IgE complex


IgE-binding protein galectin-3, which is binding to the receptor, cross-linked FcRI
expressed by monocytes, has both IgE- is mobilized in lipid rafts, where downstream
dependent and IgE-independent proinflam- signaling events potentiated by the associated
matory activities in the allergic response. Its and chains activate phosphorylation by
expression is elevated in peribronchial mono- Src kinases (38), leading to degranulation,
cytes, and it is released into the extracellular release of proinflammatory mediators, and
space. It contributes to cell survival and ac- the onset of allergic responses. In mast cells,
tivation and to the retention of cells in the these events are mediated through activation
extracellular matrix in allergic inflammation of RhoGTPases and mitogen-activated pro-
(10). Galectin-3 also enhances the hypersensi- tein kinase pathways (39, 40). As well as the
tivity of mast cells and the phagocytic activity of activation of signaling cascades to pro-
monocytes, perhaps by cross-linking additional mote degranulation, the receptor also has
IgE and FcRI molecules on these cells when sequences which interact with the actin
they have engaged their specific targets. The cytoskeleton and microtubules, initiating sig-
pentameric molecule galectin-3 may by itself naling cascades that are calcium dependent
contribute through FcRI to airway smooth and calcium independent, respectively (41,
muscle cell contractile responses and thus to 42).
airway hyperresponsiveness and remodeling in The central role of FcRI signaling in
asthma. triggering mast cell activation renders this
receptor an attractive target for therapeutic
approaches, which include neutralizing
Interactions Between Allergens, IgE,
monoclonal antibodies.
and FcRI Trigger Hypersensitivity
Responses
Allergen-Specic IgG Antibodies Can
The high-affinity IgE receptor, FcRI, is of
Trigger Hypersensitivity Responses in
primary interest to researchers investigating
Murine Models
allergy, since its activation through engage-
ment of IgE-multimeric antigen complexes IgE antibodies can mediate allergies and
mediates allergic reactions and could in anaphylactic reactions, and several cell
some instances trigger anaphylactoid re- types have been described to be involved in
sponses (36). Disrupting IgE-FcRI interac- these processes; among them are mast cells,
tions is expected to reduce IgE-mediated basophils, and macrophages, which express
allergic responses and may result in loss of Fc receptors, and also neutrophils, which
allergic inflammatory cascades, including do not express Fc receptors. Interestingly,
anaphylactic functions. Mice lacking FcRI studies with mice have demonstrated that
showed normal development and differenti- effector cells are not exclusively activated by
ation of mast cells, B cells, and T cells, as well the combination of IgE, its high-affinity
as a normal level of expression of the low- receptor, and antigens and that IgG anti-
affinity IgE receptor, CD23 (37). However, bodies can trigger cell activation and de-
these mice were resistant to systemic and granulation which can lead to anaphylactoid
cutaneous anaphylaxis induced by intrave- responses. It has been reported that active
nous and subcutaneous injection of allergen- systemic anaphylaxis (ASA) can be induced
specific IgE and the allergen shortly after- through activation of FcRs in vivo via IgE-
wards. These data demonstrated that the independent effector mechanisms in mice
interaction between IgE and the FcRI could (43, 44). Human neutrophils transferred into
be interrupted to prevent escalation of the mice that lack active IgG Fc receptors on
allergic response. mouse immune cells were described to
80 KARAGIANNIS ET AL.

restore ASA. FcRIIA and FcRIIIB are ex- challenge infection with S. mansoni when
pressed on human neutrophils, but only passively transferred into naive recipient
FcRIIA can bind mouse IgG, indicating that rats. Furthermore, in another study, induc-
this receptor was responsible for restored ASA tion of resistance to infection by adoptive
after human neutrophil transfer into mice (45, transfer of eosinophils or platelets bearing
46). In support of the role of FcRIIA in IgE indicated that the presence of IgE on
anaphylactic reactions, a recent study using an these effector cells was crucial (51).
FcRIIA transgenic mouse model demonstrat- Subsequently, support for a role of IgE in
ed that induction of FcRIIA engagement and parasite immunity was found when it was
signaling was sufficient to trigger active and demonstrated that human eosinophils, plate-
passive anaphylaxis as well as airway inflam- lets, and macrophages could harness IgE in
mation. Additionally, blocking FcRIIA in the vitro to mediate cytotoxicity and phagocytosis of
absence of IgE abolished the reactions induced Schistosoma mansoni or Leishmania major via
by IgG antibodies in vivo (47). Further inves- FcRI and CD23, respectively (17, 18, 52). These
tigations revealed that IgG1 induced passive observations were subsequently established to
systemic anaphylaxis by activating basophils, be relevant to human immunity when epidemi-
whereas IgG2 induced passive systemic ana- ological studies with Schistosoma haematobium
phylaxis via neutrophils, while ASA depended provided evidence that host protection against
on monocytes/macrophages or neutrophils reinfection in S. haematobium-infected popula-
and not on mast cells and basophils (47, tions was associated with high levels of para-
48). site-specific IgE (53), and subsequently, IgE
Thus, emerging evidence in murine models antibodies against Schistosoma mansoni were
suggests that IgG and FcR complexes contrib- shown to positively predict resistance against
ute to allergic and anaphylactoid processes, reinfection with this blood fluke (54).
although the precise requirements and path- More recently, studies have demonstrated
ways involved need to be elucidated. evidence that IgE antibodies are capable of
activating a different cell type, namely mast
cells to induce elimination of parasites via the
release of toxic granules (55). Trichinella
IGE ANTIBODIES IN INFECTIOUS
spiralis infection induces intestinal masto-
DISEASES
cytosis and heightened IgE responses, and
elimination of this parasite requires expulsion
IgE Mechanisms of Protection in
of the adult worms from the gut and destruc-
Parasitic Infections
tion of the larval cysts deposited in the muscles
Besides their critical role in allergy, IgE (56). In IgE-sufficient animals, intense deposi-
antibodies play a key physiological role in tion of IgE around the necrotic larval cysts was
immunity against multicellular parasitic demonstrated with associated accelerated re-
infections by a number of different mecha- moval of worms from the intestine and a
nisms and via a number of IgE receptor- reduction in the viability of larval parasites in
expressing cell types (49). The first in vivo muscle (56). Indeed, T. spiralis infection drove
evidence that IgE antibodies could be an a marked splenic mastocytosis and elevated
essential component of the protective im- serum levels of mouse mast cell protease-1
munity against parasites was provided by (MMCP-1), consistent with a systemic expan-
passive transfer of monoclonal IgE anti- sion of mast cells driven by the parasite. This
bodies directed against schistosomes (50). mast cell increase was dramatically attenuated
In this study, a rat monoclonal IgE antibody in IgE/ mice, implicating IgE antibodies in
raised against Schistosoma mansoni afforded this mast cell protection from parasitic infec-
a significant level of protection against a tions. Furthermore, protective roles for mast
CHAPTER 5 Immunoglobulin E and Allergy 81

cells during T. spiralis infection have also been HIES is a direct consequence of STAT3 muta-
observed using mast cell-deficient mice and by tions (59, 64). STAT3 regulates multiple fam-
antibody inhibition of the mast cell marker, ilies of cytokines, including IL-6, IL-21, IL-22,
c-Kit (56). and IL-23, which are involved in the genera-
tion of Th17 cells (65, 66). Th17 cells are
essential for clearance of bacterial and fungal
Hyper-IgE Syndrome and Sensitivity
infections via induction and expansion of
to Infections
neutrophilic and antimicrobial responses
Elevated serum IgE is a major hallmark of (67). Impaired Th17 immune responses are a
hyper-IgE syndrome (HIES), a rare primary likely explanation for recurrent infection in
immunodeficiency disease. Common clini- AD-HIES (68).
cal manifestations include eczema and skin The precise role, if any, of IgE in recurrent
abscesses, often described as cold boils infections in HIES is unknown. It is likely that
(57). HIES is clinically subdivided into elevated serum IgE in nonatopic HIES patients
autosomal dominant HIES (AD-HIES) and is simply a marker of deregulated immune
autosomal recessive HIES (AR-HIES). AD- responses, with a dominating Th2 lineage
HIES is associated with additional somatic driving the generation of IgE cells. In addition,
abnormalities of the vascular, connective impaired STAT3 induction of IL-21, a critical
tissue, and skeletal systems, whereas AR- negative regulator of IgE class switching and B-
HIES is limited to skin conditions, including cell death, is evident in animal models of AD-
eczema, and is frequently more severe (58). HIES (69) and may largely contribute to the
HIES is further classified according to associated high serum IgE levels observed in
underlying mutations in key proteins in- patients with HIES. As allergy and asthma are
volved in intracellular signaling networks, common in patients with DOCK8 mutations,
namely STAT3 and DOCK8. Dominant the role of allergen-specific IgE may be more
negative mutations in STAT3 are more prominent in both the underlying pathogenesis
frequent in AD-HIES (59) and DOCK8 of AR-HIES and its role in recurrent infections.
mutations are more frequent in AR-HIES Recent reports demonstrate that in nasal polyps
(60, 61), which may be directly related to from patients with chronic rhinosinusitis,
the clinical and underlying immunological which are frequently associated with Staphylo-
differences observed. In terms of allergy, coccus aureus infections, ongoing local receptor
patients with DOCK8 mutations frequently revision and class switching to IgE occurs (70).
suffer from symptoms of atopic disease such It is possible that such events occur during
as asthma (62). It has been suggested that Staphylococcus aureus infection in AR-HIES,
despite elevated serum IgE being a feature which could suggest a mechanism of interplay
of both forms of HIES, differences in between infection and allergy in AR-HIES.
mechanisms of IgE regulation may be Current strategies to prevent recurrent
apparent. Xiong et al. suggested that affinity Staphylococcus aureus infection in HIES
maturation pathways of IgE-producing cells involve administration of antibiotics and
may differ between atopic and nonatopic regular steroid treatment (58). Treatment
HIES patients, and such investigation of a patient with HIES with omalizumab, an
would be very informative (63). anti-IgE antibody that blocks binding of
Recurrent Staphylococcus aureus infec- circulating IgE to its Fc receptors on effector
tions in the lungs and skin is common in cells, resulted in significant clinical im-
HIES and, to a lesser degree, Haemophilus provement (71). This report indicates that
influenzae and Streptococcus pneumoniae targeting IgE in AR-HIES may prove to be a
infections (58). It has been suggested that beneficial alternative to current treatment
increased susceptibility to infection in AD- strategies.
82 KARAGIANNIS ET AL.

Virus-Induced IgE Antibodies during rhinovirus infection could spill


over into the circulation, giving rise to the
Multiple reports demonstrate that viral
detected increase in serum IgE during
infection can induce IgE production. Infec-
infection in asthma. In 1998, Rager et al.
tion of human tonsil B cells with measles
demonstrated that infection of Ramos B cell
virus induced germ line epsilon expression
line with rhinovirus 14 and 16 strains (RV14
(72), which suggests that direct viral infec-
and RV16) induced germ line epsilon ex-
tion triggers IgE class switch recombination.
pression (73). Whether in vivo rhinovirus
It has been suggested that virus-induced
infection induces IgE class switching or B-
activation of B-cell antiviral protein kinase
cell proliferation and differentiation into
R drives virus-induced class switching to
IgE plasma cells would be informative.
IgE, via cellular detection of viral double-
The role of respiratory syncytial virus
stranded RNA (73).
(RSV) in the development of asthma and
In allergic disease, viral infection may
allergies in children has been well estab-
increase production of allergen-specific IgE
lished (84). Children with recurrent RSV
antibodies and so increase susceptibility to
infections developed high titers of serum
allergen-induced IgE reactions such as mast
IgE and tested positive with allergen skin
cell degranulation. In atopic asthma, respi-
prick tests (85). The risk of developing
ratory viral infections induce over 80% of
asthma was significantly greater in these
asthma exacerbations, and 60% of these are
children, highlighting RSV as a major risk
strains of rhinovirus (74, 75), also known as
factor for the development of allergic asth-
the common cold virus. The risk of hospi-
ma. RSV has been shown to induce produc-
talization is greatest in asthmatics infected
tion of RSV-specific IgE (86). Dakhama et
with rhinovirus who are both sensitized and
al. demonstrated a positive correlation be-
have been exposed to allergen (76, 77),
tween RSV-IgE production and severity of
indicating an additive effect of viral infection
airway hyperresponsiveness in mice (86).
on underlying allergic inflammatory reac-
Investigation into mechanisms of virus-
tions. In addition, serum levels of total IgE
induced IgE antibodies in allergic disease
(78) and dust mite IgE (79) are elevated
will prove very informative and could aid the
during rhinovirus infection in atopic asth-
development of novel preventative therapies
matics, which suggests that rhinovirus
for both virus-induced asthma exacerbations
infection increases susceptibility to IgE-
and the development of asthma.
mediated reactions. The precise mecha-
nisms for how in vivo rhinovirus infection
increases production of IgE are unknown;
ANTIBODIES IN THE TREATMENT
however, an advanced understanding of
OF ALLERGIES
rhinovirus-induced inflammation has gen-
erated logical hypotheses. Both in vitro and
Recombinant Antibodies Targeting IgE
in vivo rhinovirus infection enhance Th2-
Interactions with Fc Receptors
mediated airway inflammation in asthma
(80, 81, 83). It is possible that rhinovirus- Designing therapeutic antibodies targeting
induced inflammation drives germinal cen- IgE represents an approach for the treat-
ter reactions within the bronchial mucosa. ment of patients with allergies. The primary
Local germinal center reactions could gen- aim of anti-IgE therapy is to reduce the
erate B cells within the lungs to class switch amount of circulating IgE in patients and to
to IgE and induce differentiation into IgE- neutralize the interactions between free IgE
secreting plasma cells. It is possible that and immune cells responsible for mediating
this excessive local production of IgE allergic reactions (87). One approach entails
CHAPTER 5 Immunoglobulin E and Allergy 83

blocking free IgE from binding to its Fc design of IgE immunotherapy to ameliorate
receptors expressed on immune effector the risk of any acute allergic reactions such as
cells, such as basophils and mast cells, to type 1 hypersensitivity in patients (Table 1).
prevent the release of inflammatory me- Anti-IgE therapy has widespread appli-
diators, such as histamines and tryptases, cability in asthma. Currently, omalizumab is
from these cells which lead to hypersensi- being evaluated for the treatment of many
tivity. Another would be to downregulate other IgE-mediated allergic diseases, in-
the production of IgE by B cells. cluding atopic dermatitis, seasonal allergic
rhinitis, food allergy, and urticaria (Table 2).
IgE-Blocking Antibodies for Passive Although discontinued for clinical develop-
Immunotherapy: Omalizumab ment, due to partnership agreements rather
There have been numerous anti-IgE anti- than clinical utility, the anti-IgE antibody
bodies in preclinical and clinical develop- talizumab (TNX-901) demonstrated the
ment, with omalizumab (Xolair) being the ability to increase the sensitivity threshold
first licensed anti-IgE therapy approved for to peanuts, supporting the use of anti-IgE
the treatment of allergic bronchial asthma therapy for some food allergies to prevent
in 2003 (88). Omalizumab represents an mast cell/basophil degranulation (95).
important breakthrough in the development Talizumab can also inhibit the presentation
of antibodies for anti-IgE immunotherapy of allergens, since it was shown to block
in allergy. Omalizumab is a humanized IgG1 allergen-CD23 complex formation on the
monoclonal antibody that recognizes an surface of B cells, and can prevent prolifer-
epitope in the C3 region of IgE, the IgE ation of allergen-specific T cells (96).
Fc region which contains the binding sites
for both the high- and low-affinity IgE Fc Antibodies Blocking CD23 Functions
receptors (10, 89). This anti-IgE antibody and B Cells
can block the binding of circulating IgE to Another approach centers on targeting CD23
both of its Fc receptors, which are ex- with the aim of regulating IgE production
pressed on the surfaces of various effector through blocking the functions of the low-
cells, thus preventing IgE-mediated allergic affinity IgE receptor, CD23, on B cells. A
immune responses (Fig. 1). prominent agent featuring this approach is
Treatment with omalizumab also has been primatized anti-CD23 macaque/human chi-
shown to result in downregulating the expres- meric antibody, lumiliximab (IDEC-152),
sion of FcRI on immune cells (Table 1) (90, 91, which was designed to modulate antigen
92). Notably, omalizumab is not able to bind to presentation, reduce activation of Th2 re-
IgE in the complex with its receptors, possibly sponses, and hinder IgE production by B
due to allosteric inhibition. It was recently cells (97). This agent reached clinical eval-
shown that engagement of IgE with omali- uations for the treatment of allergic asthma,
zumab triggers a lower degree of bending in but it is now evaluated for use in hematolog-
IgE compared to that adopted through recog- ical oncology indications such as chronic
nition of FcRI, inducing a conformational lymphocytic leukemia.
change that is incompatible with FcRI recog- New anti-IgE antibodies are undergoing
nition (93). Due to these properties, the preclinical evaluations and development and
antibody is not capable of cross-linking IgE include agents aimed at downregulating the
already bound to receptors on the surfaces of production of IgE by B cells. One emerging
mast cells and basophils, and therefore, any strategy is the engineering of an antibody
unwanted immune cell activation and release (8D6) which, like omalizumab, prevents the
of inflammatory mediators is avoided (94). binding of free IgE to cell surface FcRI but
Examination of cross-linking is essential in the additionally interacts with CD23-bound IgE
84 KARAGIANNIS ET AL.

FIGURE 1 Omalizumab blocks IgE-mediated mast cell activation. The allergic response mediated by
multivalent allergen-IgE-FcRI complex formation on the surface of mast cells triggers cross-linking of
FcRI, leading to downstream signaling events potentiated by the and chains (left). These entail
phosphorylation (P) by Src kinases and cellular activation through RhoGTPases and mitogen-activated
protein kinase pathways, leading to mast cell degranulation and the rapid release of a range of
vasoactive amines (e.g., histamine), prostaglandins, leukotrienes, cytokines, and chemokines, inducing
and maintaining allergic inammatory responses. Omalizumab, a humanized IgG1 monoclonal antibody
that recognizes an epitope in the C3 region of IgE, can block the binding of circulating IgE to FcRI,
sequestering free and allergen-bound IgE (right). These interactions prevent allergen-IgE-FcRI complex
formation on the surface of mast cells and interfere with the signaling cascades that trigger
degranulation and the onset of IgE-mediated allergic inammatory cascades. Syk, spleen tyrosine
kinase; AKT, Ak strain thymoma serine/threonine-specic protein kinase; ERK, extracellular signal-
regulated kinase; JNK, Jun N-terminal protein kinase; P, phosphorylation. doi:10.1128/microbiolspec.
AID-0006-2012.f1

with the aim of also inhibiting IgE production total IgE in circulation in vivo while neutraliz-
on B cells, thereby controlling circulating IgE ing IgE in a manner similar to omalizumab. The
levels (98). Another approach encompasses an two complementary properties resulted in
anti-IgE antibody, XmAb7195, with modified prolonged efficacy for XmAb7195 compared
IgG Fc domains that feature increased affinity to antibodies exerting only one of the two
to the inhibitory FcRIIB expressed on B cells mechanisms.
and also the ability to suppress B-cell activa- Another strategy involved use of the anti-
tion. Like omalizumab, this agent was recently CD20 chimeric antibody rituximab, which
reported to target the C3 domains of free IgE has been approved for the B-cell malignancy
but to also cointeract with IgE in the complex non-Hodgkin lymphoma and has also dem-
with FcRIIB on the surface of IgE-expressing onstrated efficacy in some autoimmune
B cells (99). This antibody was shown to disorders (100). The antibody was examined
suppress B-cell signaling and prevent matura- as a potential tool to eliminate B cells and
tion of B cells into IgE+ plasma cells, reducing therefore remove allergen-induced B-cell
TABLE 1 Interactions of omalizumab with IgE, the resulting mechanisms of action, and clinical effects of treatment in patients with allergies
Monoclonal antibody property Mechanism Biological function Immunological effects Clinical impact

Binds to the C3 region Sequesters free IgE Reduction of IgE titers in human Reduced mast cell and basophil Reduced allergic symptoms/efcacy
of free circulating IgE sera and reduced levels of activation, reduced antigen
cell-bound IgE presentation
Prevents IgE-FcRI Decreased FcRI density on Decreased basophil/mast cell Reduced allergic symptoms/efcacy
interactions on cell surfaces circulating basophils responsiveness upon challenge
with IgE-allergen complexes
Decreased FcRI density on APCs Inhibition of antigen processing Reduced allergic inammation/
(e.g., DCs) and presentation to T cells, efcacy
reduced Th2 cellular and cytokine
responses
Prevents IgE-FcRII Inhibition of allergen-IgE-FcRII Decreased antigen presentation Reduced chronic allergic responses/
interactions on cell complex formation to T cells efcacy
surfaces
Cross-linking and IgE-mediated Decreased de novo synthesis
endocytosis of IgE

CHAPTER 5 Immunoglobulin E and Allergy


Does not interfere \with Circulating IgEs of any Blocks IgE-FcR-IgE functions Inhibition of allergic responses Broad applicability in allergic
variable regions of IgE specicity sequestered mediated by any allergen irrespective of allergen specicity diseases
Blocks cell surface interactions
of FcRs with any IgEs
Binds to a C3 epitope in Does not recognize IgE bound Addition of antibody could not Reduced potential for type 1 Improved safety prole
proximity to both binding to cell surface IgE-FcRs trigger FcRI cross-linking and hypersensitivity responses that
sites recognized by FcRI mast cell/basophil degranulation may lead to onset of anaphylaxis
and FcRII
High specicity for IgE Fc Sequesters free IgE only Reduction of IgE titers but no Normal immunity is unaffected No off-target effects/safety
reduction of IgG

85
86
KARAGIANNIS ET AL.
TABLE 2 Active phase II, III, and IV interventional clinical studies for antibody immunotherapies of allergic diseases in 2012a
Trial title Intervention Target Phase Indication Primary endpoint(s) Sponsor Status
b
Oral immunotherapy combined with MAb + OIT : IgE II Milk allergy Efcacy: percentage of National Institute of Recruiting
humanized monoclonal anti-IgE omalizumab + milk subjects developing clinical Allergy and Infectious
antibody Xolair (omalizumab) in the OIT tolerance to milk Diseases (NIAID)
treatment of cows milk allergy
Peanut oral immunotherapy and MAb + OIT: IgE I/II Peanut Efcacy: percentage of University of North Active, not
anti-IgE for peanut allergy omalizumab + hypersensitivity subjects developing clinical Carolina (collaborator, recruiting
peanut OIT tolerance to peanuts Genentech)
Effect of KB003 in subjects with MAb: KB003-04 GM-CSFc II Moderate-to-severe Safety, tolerability, and KaloBios Recruiting
asthma inadequately controlled by asthma efcacy: effects on lung Pharmaceuticals
corticosteroids function measured by FEV1d
A phase 2b, randomized, double-blind MAb: tralokinumab IL-13 IIb Asthma Efcacy: asthma exacerbation MedImmune LLC Recruiting
study to evaluate the efcacy of rate
tralokinumab in adults with asthma
Dose ranging pharmacokinetics and MAb: mepolizumab IL-5 II Asthma Pharmacokinetics, GlaxoSmithKline Recruiting
pharmacodynamics study with (SB-240563) pharmacodynamics,
mepolizumab in asthma patients with immunogenicity analysis;
elevated eosinophils effects on blood eosinophil
levels, drug plasma levels
(Continued on next page)
TABLE 2 Active phase II, III, and IV interventional clinical studies for antibody immunotherapies of allergic diseases in 2012a (Continued)
Trial title Intervention Target Phase Indication Primary endpoint(s) Sponsor Status

Omalizumab in patients with MAb: omalizumab IgE III Persistent allergic Efcacy: change from Novartis Recruiting
moderate to severe persistent allergic asthma baseline in mean Pharmaceuticals
asthma not adequately controlled morning PEFe (collaborator,
despite GINA (2009) step 4 therapy Genentech)
A study of lebrikizumab in patients MAb: lebrikizumab IL-13 III Asthma Efcacy: rate of asthma Genentech Recruiting
whose asthma is uncontrolled with exacerbations
inhaled corticosteroids and a second
controller medication (LUTE)
A study of lebrikizumab in patients MAb: lebrikizumab IL-13 III Asthma Efcacy: rate of asthma Genentech Recruiting
with uncontrolled asthma who are on exacerbations
inhaled corticosteroids and a second
controller medication (VERSE)
Effect of Xolair on airway MAb: omalizumab IgE IV Allergic asthma Efcacy: reduction in Creighton University Active, not
hyperresponsiveness abnormal increase in (collaborator, recruiting
limitation to airow in Genentech)
patients with asthma;
decrease the amount of
inammation in the lungs
a
Source: ClinicalTrials.gov; U.S. National Institutes of Health.
b
MAb, monoclonal antibody; OIT, oral immunotherapy.
c
GM-CSF, granulocyte-macrophage colony-stimulating factor.
d

CHAPTER 5 Immunoglobulin E and Allergy


FEV1, forced expiratory volume in 1 s.
e
PEF, peak expiratory ow.

87
88 KARAGIANNIS ET AL.

class switching and synthesis of IgE and also allergic inflammatory cascades in animal lungs
IgG autoantibodies, for applications in atopic (103). Phase I and II clinical studies in patients
dermatitis and urticaria, and anecdotal suc- with asthma using a humanized antibody
cesses for individual patients treated with (pascolizumab, SB-240683) which blocks IL-4
this agent have been reported (101, 102). IL-4 receptor interactions did not result in
As demonstrated by the success of oma- clinical improvements in patients (104).
lizumab and the other anti-IgE therapies By sharing a receptor with IL-4 in the form
undergoing preclinical and clinical evaluations, of IL-4R subunit , IL-13 is thought to be
monoclonal antibodies targeting IgE are involved in class switching to IgE but also has
emerging as important therapeutic modalities functions distinct from IL-4, such as regulation
for the treatment of allergic diseases, while new of epithelial cell maturation, muscle contrac-
approaches are being examined which aim to tility, production of extracellular matrix pro-
improve the efficacy of omalizumab. Other teins, and recruitment of monocytes, T cells,
strategies with indirect effects on IgE produc- and eosinophils. Targeting IL-13 is presently an
tion, such as those aimed at neutralizing B cells area of translational and clinical activity.
and targeting CD23, have been less successful Tralokinumab (CAT-354), a human IgG4 anti-
to date. body which can neutralize IL-13, was associat-
ed with improved lung function when tested in
patients with asthma, although results from
Antibodies Targeting Key Cytokines,
phase IIb clinical studies are pending (Table 2)
Chemokines, and Their Receptors
(105). Another agent presently undergoing
Th2-type cytokines (e.g., IL-4, IL-5, IL-9, clinical evaluations in phase III clinical trials
and IL-13) are produced by mast cells, is the IgG4 humanized anti-IL-13 antibody,
basophils, Th2 lymphocytes, and eosinophils lebrikizumab. Small improvements in lung
but also by nonimmune cells such as lung functions were observed when this antibody
airway epithelial cells. They play key roles in was administered in asthmatics with inhaled
triggering and maintaining allergic inflam- glucocorticoid-resistant disease, and clinical
mation and contribute to the resulting pa- responses were associated with increased
thologies. Targeting cytokines associated levels of serum periostin, suggesting the merits
with allergies with antibodies has been of utilizing biomarkers to select patients most
explored as a treatment option, and various likely to respond (106).
strategies are being examined in preclinical Eosinophilia comprises an important com-
studies and in clinical trials, with asthma ponent of the inflammatory infiltrate in asth-
being a major therapeutic indication for matic lungs, and increased levels of eosinophils
these efforts. are found in the blood of allergic subjects.
Production of IL-4 by basophils, mast cells, Because of its role in potentiating activation and
NK cells, or eosinophils and subsequently also proliferation of eosinophils and their terminal
by Th2 lymphocytes is linked to a number of differentiation in tissues, IL-5 constitutes an
processes in allergic inflammation, such as attractive therapeutic target for allergic condi-
upregulation of FcRI and CD23 on the surface tions characterized by eosinophilia (107). Three
of effector cells and class switching from IgM humanized antibodies targeting IL-5 are in
to IgE by B lymphocytes. Therefore, neutraliz- clinical trials for the treatment of eosinophilia-
ing IL-4 would be expected to inhibit IgE associated allergic conditions. Clinical studies
synthesis, formation of Th2-type T cells, and of patients with hypereosinophilic conditions
upregulation of IgE receptors. Preclinical stud- reported that treatment with mepolizumab
ies in a murine model of asthma demonstrated (SB-240563) resulted in increased titers of
that an anti-IL-4 antibody could inhibit IgE serum IL-5 and IL-5 receptor, while reduced
production, but this agent did not abrogate eosinophil activation and lower blood eosino-
CHAPTER 5 Immunoglobulin E and Allergy 89

phil levels were observed (108). One study may reflect the importance of considering the
demonstrated disease management with lower complex balance between mounting natural
doses of corticosteroid treatment and reduced protective immune functions and reducing
bronchial mucosal eosinophilia in asthmatic allergic inflammatory environments when
subjects receiving this antibody. Others designing therapeutics for allergic diseases
reported reduced exacerbation of symptoms (117).
in asthmatics, with further clinical trials with
this agent presently in progress (Table 2) (109,
Active Allergen Immunotherapy
110). Two other humanized antibodies, the
Induces Neutralizing Antibody
anti-IL-5 antibody reslizumab (CTx55700)
Responses in Patients
and the anti-IL-5 receptor-specific antibody
benralizumab (MEDI-563), have demonstrated An attractive therapeutic direction entails
favorable safety profiles and reductions in strategies to interrupt IgE binding to allergens
eosinophil counts in patients. So far, clinical to reduce IgE-mediated responses in patients
utility has not been proven for anti-IL-5 with allergies. For known allergens, it is
therapies, although clinical studies are still possible to produce a vaccine that would
underway (111, 112, 113). reduce the inflammatory response of antigen-
Studies of murine models of asthma dem- bound IgE binding to mast cells by disrupting
onstrated that IL-9 provides activatory and the binding of the Fab region of IgE to
proliferative signals to mast cells, contributing allergenic epitopes, and this has formed the
to airway hyperresponsiveness and airway focus of recombinant allergen immunotherapy.
fibrosis. Targeting this cytokine with an anti- This therapy has been examined now for
body reduced these allergen-induced inflam- decades; the availability of allergen sequences
matory symptoms, and antibody treatment was has led to the expression of recombinant
associated with reduced levels of known pro- allergens and production of synthetic allergen-
fibrotic cytokines transforming growth factor ic peptides, and there now exists a database
, vascular endothelial growth factor, and where these sequences have been deposited for
fibroblast growth factor 2 (114). A humanized comparison (118). Immunization with recom-
antibody targeting IL-9 (MEDI-528) has been binant allergen induces the production of
tested in clinical trials in healthy individuals allergen-specific IgGs, which compete with
and patients with asthma with some encour- IgEs for binding to the allergen, thus blocking
aging clinical responses (115). downstream IgE cross-linking and suppressing
Expressed by macrophages and mast cells allergic responses (119).
following activation with different stimuli, There are a number of subclasses of these
including IgE-FcRI associations, TNF- recombinant and synthetic allergens. Of par-
triggers infiltration of immune effector cells ticular interest to those who study the allergic
in tissues and has a role in inflammatory response stimulated by the cross-linking of IgE
processes, including those associated with are the wild-type hypoallergens. These are
asthma (116). Anti-TNF- antibodies tested in designed to be allergen mimics and may consist
the clinic include the chimeric infliximab, the of a fragment of the allergen, a mutant of the
humanized certolizumab pegol, the human allergen, or the whole recombinant allergen.
antibodies adalimumab and golimumab, and The net result of this subclass is reduced IgE
the soluble TNF- receptor 2-Fc fusion protein, activity in patients. The first clinical trial of the
etanercept. All of these agents are expected to wild-type hypoallergen was with the Birch
neutralize TNF-. However, any moderate pollen allergen, Bet v 1, with patients who
clinical responses observed have been showed an allergic response to the crude
overshadowed with reports of induction of extract. Application of rBet v 1, as well as
opportunistic infections and cancer, which fragments of the Bet v 1, showed efficacy in
90 KARAGIANNIS ET AL.

reducing histamine release in basophils, an highlighted a potentially important residue


important step in the progression of an allergic (124). When this residue was mutated, in two
response (120). The same trial also showed that single-point mutant forms of the allergen, it
there was an increased production of allergen- resulted in very different circular dichroism
specific IgG1 and IgG4 with both the full- spectra, suggesting a change in conformation of
length allergenic recombinant and smaller the allergen based on altering this site. Both of
fragments, suggesting that a few allergenic the mutants had reduced ability to bind IgE and
residues are all that are required to induce a activate basophils but could be recognized by
clinical regression of symptoms in patients. Not allergen-specific T cells. These findings suggest
only was there a notable lack of histamine that site-directed mutagenesis retains the im-
reaction to the use of recombinant therapies munogenicity of these allergens while
but there were also no T-cell-mediated toxic counteracting initiation of an allergic response.
effects noted. In 2011, there were 11 clinical trials running
The efficacy of recombinantly expressed in the United Kingdom using recombinant
allergen has been characterized and shown to allergens to combat allergic diseases, with 4
be capable of inducing desensitization to of these being in phase III (125). The relative
specific allergens. The Der p 2 allergen is the success of this therapy suggests that targeting
major allergen responsible for dust mite aller- the interaction of the antibody with the
gies, and there are a number of allergenic allergen may be an effective therapeutic alter-
isoforms. One of these isoforms was selected native to or complementary to the use of anti-
and expressed in the yeast strain Pichia pastoris IgE antibodies.
and as inclusion bodies in Escherichia coli,
where they were refolded to a stable tertiary
Triggering Protective Immunity with
structure. The secondary structures of these
Immunotherapythe Role of IgG4
recombinant products were compared to the
native allergen, obtained from dust mites by Specific immunotherapy (SIT) involves the
circular dichroism, and showed that different administration of allergens to achieve clin-
structures were obtained for each of the ical tolerance, with the aim of easing the
products. However, in a mouse model, both symptoms in patients with allergic condi-
of the recombinant products were capable tions. Long-term clinical benefits of SIT
of inducing desensitization to the Der p 2 persist even after discontinuation of thera-
allergen, despite differential structural prop- py, indicating the involvement of a cellular
erties. These findings suggest that potentially memory component to therapy. The mech-
linear epitopes may be sufficient for allergenic anism behind the therapy has extensively
vaccine therapy (121). been studied but still remains a matter of
Based upon the success of recombinant research and debate.
allergens, rational design of allergenic epitopes Research on SIT has to date focused on
was the next logical step, since, as far back as altered T-lymphocyte responses and induction
1970, allergens were being chemically modified of allergen-specific IgG4 antibodies. Two indi-
to reduce IgE binding capacity while retaining vidual patterns of change, which may occur
the ability to prompt the production of antigen- sequentially, have been described. The first
specific IgGs (122, 123). More recently, the use event is induced within 1 to 4 weeks and entails
of recombinant technology yielded new tools the generation of regulatory T cells secreting
with which to mutate specific residues that IL-10 and transforming growth factor , ac-
alter the activity of the allergen. An example of companied by suppression of allergen-induced
this approach was the use of mutated forms of late cutaneous responses. Subsequently,
the mouse allergen Mus m 1. The high resolu- around week 10 postchallenge, elevated
tion crystal structure of a domain of the allergen serum titers of allergen-specific IgG4 and IgA
CHAPTER 5 Immunoglobulin E and Allergy 91

are observed. Although these titers appeared The IgE-neutralizing and -tolerogenic prop-
proportional to the dose of administered erties of IgG4 may be partly due to competing
antigen rather than to clinical improvements, with allergen-specific IgE for antigen specific-
recent studies reported that the functional ity. This would interrupt IgE-FcRI-multivalent
capacity of IgG4 antibodies to block IgE allergen complex formation on the surface of
functions correlates with clinical responses immune effector cells, preventing downstream
(126, 127). These events coincide with a signaling and effector cell activation (Fig. 2).
decrease of allergen-specific IgE antibodies However, since only a fraction of IgG4 anti-
and a shift in the allergen-specific T-cell bodies are allergen specific after immuno-
response from predominantly Th2 to Th1 (126). therapy, other mechanisms may also be
Serum obtained from patients following involved. van der Neut Kolfschoten and col-
SIT has been shown to inhibit allergen-IgE leagues used recombinant IgG4 antibodies
binding to B cells, an effect mediated largely by against grass and cat allergens, which when
IgG4 antibodies in patient sera. This in vitro coinjected in a mouse graft, became bispecific in
system introduced the idea that SIT triggers a process termed Fab-arm exchange. Although
production of blocking antibodies that in- this process was not observed when antibodies
hibit IgE functions, such as IgE-FAP on B cells. were combined in vitro, these findings raise the
Similarly, basophil histamine release assays or possibility that IgG4 antibodies may also oper-
basophil activation assays have demonstrated ate by Fab-arm exchange (132). If this is true in
the functional ability of IgG4 to inhibit IgE- patients receiving allergen immunotherapy,
dependent activation and mediator release, IgE/IgG4 bispecific antibody formation may
either by competing with IgE for the antigen reduce the affinity of IgE antibodies for
and/or by stimulating cell surface IgG-inhib- allergens and interrupt IgE-FcRI-multivalent
itory receptors present on basophils and mast allergen complex formation on the surface of
cells (127, 128). Interestingly, IgA antibodies immune effector cells. These properties would
could not block allergen-IgE binding to B cells. therefore moderate IgE-mediated inflammato-
The contribution of IgA in the responses to SIT ry cascades. This latter possible mechanism by
may lie in engagement of surface IgA receptors which IgG4 antibodies may function would be
and release of the inhibitory cytokine IL-10, consistent with the observations that IgE
which may participate in the induction of effector cells such as eosinophils, mast cells, or
immune tolerance to allergens (129). basophils are reduced after SIT against house
The mechanisms triggering elevated IgE- dust mite and grass pollen (133). An additional
neutralizing antibodies following SIT are not mechanism by which SIT can tolerize allergic
completely understood, although it is known immune responses lies with the ineffective Fc-
that prolonged exposure to antigenic stimuli mediated functions of IgG4 in lacking comple-
can direct production of IgG4 by B cells. B cells ment activator properties and mediating inef-
can switch from IgG4 to IgE but not vice versa, fective FcR signals, reducing the capacity of this
as these sections of the gene are spliced out antibody class to trigger cytotoxicity or phago-
during class switching. Thus, IgG4 must be cytosis.
produced by switching from IgM, IgG13, or Additionally, involvement of IgG4 and also
IgA1 or by proliferation of preexisting IgG4+ IgA antibodies in natural induction of tolerance
B cells during SIT. It has been suggested that is supported by reports of increased allergen-
Th2 cytokine environments with elevated specific IgG4 serum levels in patients who
levels of IL-10 can drive the differentiation of spontaneously become tolerant to cows milk
IgG4-switched B cells to IgG4-secreting plasma and from findings that these serum levels are
cells (130). In addition to IL-10, the immuno- maintained and remain higher than those
regulatory cytokine, IL-21, has been found to measured from individuals with diagnosed
increase IgG4 production in vitro (131). active cows milk allergies (134, 135). Finally,
92 KARAGIANNIS ET AL.

FIGURE 2 IgG4 antibodies induced by allergen immunotherapies modulate IgE-mediated activation of


effector cells. The allergic response mediated by multivalent allergen-IgE FcRI complex assembly and
downstream signaling events leading to release of inammatory mediators (left) may be blocked by
adaptive immune responses triggered in response to SIT with recombinant antigens. IgG4 antibodies,
induced following SIT, could compete with IgE for binding to allergens and prevent the formation of
allergen-IgE-FcRI complexes on the surface of effector cells, blocking effector functions such as
degranulation (right). Syk, spleen tyrosine kinase; AKT, Ak strain thymoma serine/threonine-specic
protein kinase; ERK, extracellular signal-regulated kinase; JNK, Jun N-terminal protein kinase; P,
phosphorylation. doi:10.1128/microbiolspec.AID-0006-2012.f2

favored production of IgG4 antibodies was efforts to enhance the efficacy of these
recently demonstrated in patients with mela- antibodies by engineering Fc regions with
nomas and extrahepatic cholangiocarcinomas, enhanced affinity to Fc receptors to create
suggesting the association of IgG4 antibodies antibodies with desirable pharmacokinetic
with immune tolerance favored by IL-10-rich properties and improved cellular immune
environments such as those in tumors (136, 137). functions. Our groups approach has been to
enhance antibody Fc-mediated functions by
engineering therapeutic antitumoral anti-
THE OPPOSITE SIDE OF THE COIN: bodies with Fc regions of the IgE class
HARNESSING THE ALLERGIC RESPONSE (138). Employing this strategy, therapeutic
AGAINST CANCER AND THE EMERGENCE antibodies of the IgE class may confer
OF ANTIBODIES OF THE IGE CLASS FOR several advantages over antibodies of the
CANCER THERAPY IgG class, particularly with respect to the
treatment of solid tumors. The advantages of
Monoclonal antibodies of the IgG class have IgE would include (i) high affinity of IgE to
emerged as an important therapeutic modal- its Fc receptors; (ii) lack of IgE inhibitory Fc
ity over the last 20 years, and a number of receptors; (iii) natural immune activatory
these agents are in clinical use for the functions in tissues; (iv) presence of IgE
treatment of cancer. There have been many effector cells in tumors; (v) the activation of
CHAPTER 5 Immunoglobulin E and Allergy 93

Fc receptors (i.e., FcRI and FcRII) other cell infiltrates bearing Fc receptors capable of
than Fc receptors on a set of effector cells (e.g., mediating potent cytotoxic effects. The system-
mast cells, eosinophils, and monocytes/ ic administration of IgE therapeutic antibodies
macrophages) different from those bearing also warrants immunological monitoring of
IgG receptors; and (vi) desirable pharmaco- patients, to ensure allergic responses are pri-
kinetic properties such as fast clearance marily harnessed in solid tumors and do not
from circulation, reducing the chance of lead to potentially harmful type 1 hypersensi-
antibody-neutralizing antibody responses tivity responses in patient circulation. Here,
(35, 139, 140, 141). since only soluble multimeric antigens at high
Exploiting IgE as a novel class of therapeutic concentrations in blood would have the poten-
antibodies may prove successful in harnessing tial to cross-link IgE-FcRI complexes that may
a potent allergic response against tumor cells. give rise to type 1 hypersensitivity, the design of
The multimerization of therapeutic IgE anti- antibodies to antigenic targets known to be
bodies bound to multimeric antigens or mul- shed in multimeric form in the patient circula-
tivalent antigens expressed on tumor cells can tion should be avoided. Of note, our most
result in the cross-linking of antibody recep- advanced IgE class therapeutic antibody can-
tors on immune cells, high-avidity binding and didate, MOv18 IgE, is designed against the cell
cell activation, and the release of inflammatory surface tumor antigen FR which is shed in low
mediators such as histamines, leukotrienes, levels and in a monomeric form in human sera.
tryptases, inflammatory cytokines, and cyto- To address concerns of potential type 1 hyper-
toxic granules which would result in tumor sensitivity responses upon administration of
cell death. We have demonstrated that IgE antibody in patients, we have evaluated the
antibodies targeting the tumor antigens folate activation of human immune cells involved in
receptor alpha (FR) and the human epider- systemic allergic responses, such as basophils
mal growth factor receptor 2 (HER2/neu) are and mast cells. We conducted these experi-
capable of mediating effective cytotoxicity ments using two ex vivo and in vitro functional
against tumor cells through IgE Fc-mediated readouts of effector cell activation following the
interactions with Fc receptors present on addition of therapeutic IgE antibodies in blood
frequently tumor-resident immune cells such and sera from patients with cancer and healthy
as monocytes, mast cells, and eosinophils (34, volunteers. In both of these readouts, we have
35, 139, 140, 142). Furthermore, in a number of found a lack of human immune cell activation,
animal models of ovarian carcinoma, the even in the presence of soluble forms of tumor
administration of MOv18 IgE antibodies, antigens. These findings suggest that this
which target the tumor antigen FR, resulted approach bears low risk of systemic type 1
in increased survival and restriction of tumor hypersensitivity (143). This is likely due to the
growth compared to dosing with the IgG1 absence of IgE cross-linking in blood, even in
counterparts (35, 139, 140). Thus, IgE anti- the presence of soluble tumor antigens and
bodies targeting tumor antigens are capable of circulating tumor cells at the highest reported
mediating allergic responses in solid tumors physiological amounts in patients. These stud-
by the activation of immune cells generally ies provided support for the potential safety of a
involved in allergic responses, and such a tumor antigen-specific IgE antibody adminis-
strategy resulted in the redirection of the tered in the patients circulation (138, 144).
cytotoxic effects of these immune cells against In conclusion, we propose that therapeutic
tumor cells (141). IgE antibodies may harness allergic responses
We envisage that a successful clinical utility in individuals with cancer, resulting in signif-
setting for therapeutic IgE antibodies includes icant antitumor effects, and that this antibody
the selection of appropriate disease settings class has the potential to emerge as an impor-
such as those of solid tumor types, with immune tant therapeutic modality to direct IgE immune
94 KARAGIANNIS ET AL.

responses against cancer. We await the testing gent design of new more effective therapeutics
of the first therapeutic IgE in humans and with improved functional properties or on the
continue in the design and development of IgE design of agents with multimodal actions, all
antibodies for the treatment of many malig- aiming at achieving better efficacy than that
nancies, including breast cancer, ovarian can- shown with application of omalizumab in the
cer, and melanoma. clinic. Such strategies would include mono-
clonal antibodies but also small molecules with
the capacity to interfere with IgE-FcR inter-
CONCLUDING REMARKS AND FUTURE actions (145).
ROLES OF ANTIBODIES IN THERAPY The significance of antibodies in the treat-
ment of allergic diseases has also been sug-
Key roles of IgE class antibodies in allergic gested in a different context, through the
diseases, in the associated allergic inflamma- pivotal findings that allergen immunotherapy
tion, and in parasitic and infectious diseases triggers increased levels of protective humoral
have emerged over 4 decades of research. responses in patients. These responses manifest
Increased levels of IgE antibodies specific to in the form of IgG4 subclass antibodies. All
allergens are central to the pathogenesis, these developments highlight the central role of
exacerbation, and diagnosis of many allergic antibodies in passive but also active immuno-
conditions. Increased levels of IgE are also therapy of allergic diseases. Furthermore, the
associated with sensitivity to infections in mechanisms by which IgG4 antibodies operate
patients with HIES, while certain viral infec- are currently an area of intense research
tions also trigger class switching and produc- activity and may themselves lead to the rational
tion of IgE that can lead to symptom design of active immunotherapies designed to
exacerbations in allergic individuals. On the trigger more effective humoral responses capa-
other hand, IgE antibodies are also thought to ble of regulating IgE-mediated inflammation.
confer protective roles in parasitic infections. Another potential future strategy may be the
Potential functions for the IgG class are also design of engineered IgG4 subclass antibody
reported in some anaphylactic responses, therapeutics capable of passively blocking IgE-
while IgG4 subclass and IgA antibodies are mediated responses in patients with allergies.
associated with development of natural and Finally, IgE antibodies and their potent
specific allergen immunotherapy-induced tol- effector functions, known to confer protection
erance to allergen exposure. These provide in parasitic infections and to contribute to
support for the important contributions of allergic diseases, may be utilized as therapeu-
antibodies in allergic and infectious diseases. tics for cancer. A number of groups have
Importantly, the approval of omalizumab for examined the concept of raising IgE antibodies
the treatment of a number of IgE-mediated through a number of active immunization
allergies constituted a proof-of-principle agent approaches and also by harnessing powerful
and a significant milestone in the clinical man- immune effector cell functions against cancer
agement of patients. Omalizumab has also cells through passive immunotherapy with
contributed new understanding of the interac- monoclonal antibodies (138, 139, 140, 142, 146,
tions between IgE, its receptors, and allergens 147, 148, 149, 150, 151, 152, 153). A growing
and novel insights into the mechanisms by number of studies, including those from our
which these interactions impact on the devel- group, demonstrate the promise of activating
opment of allergic inflammation. These allergic responses against cancer and have
developments have also placed these interac- defined the emerging field of AllergoOncology
tions at the forefront of translational efforts to (138, 154, 155). Passive immunotherapy with
design novel treatments. Furthermore, they engineered antibodies has, in our hands, dem-
have prompted increased focus on the intelli- onstrated considerable efficacy with a number
CHAPTER 5 Immunoglobulin E and Allergy 95

of antibody therapeutic candidates, in different 2. Oppenheimer J, Nelson HS. 2006. Skin test-
oncological disease settings, and in a number of ing. Ann Allergy Asthma Immunol 96:S6S12.
3. Bernstein IL, Li JT, Bernstein DI, Hamilton R,
disease-relevant models (138). Ultimately, the Spector SL, Tan R, Sicherer S, Golden DB,
utility of an antibody class such as IgE, Khan DA, Nicklas RA, Portnoy JM, Blessing-
commonly associated with the pathogenesis of Moore J, Cox L, Lang DM, Oppenheimer J,
allergies, may find a new purpose in cancer Randolph CC, Schuller DE, Tilles SA, Wallace
DV, Levetin E, Weber R. 2008. Allergy diag-
therapy. Our planned first-in-class clinical
nostic testing: an updated practice parameter.
study of our anti-FR IgE antibody will un- Ann Allergy Asthma Immunol 100:S1S148.
doubtedly provide new insights into the hy- 4. Akhabir L, Sandford AJ. 2011. Genome-wide
pothesis that the potent effector activatory association studies for discovery of genes
functions of IgE antibodies could be directed involved in asthma. Respirology 16:396406.
against malignancy. 5. Valenta R, Ball T, Focke M, Linhart B,
Mothes N, Niederberger V, Spitzauer S,
Swoboda I, Vrtala S, Westritschnig K, Kraft
ACKNOWLEDGMENTS D. 2004. Immunotherapy of allergic disease.
Adv Immunol 82:105153.
We acknowledge support from Cancer Re- 6. Barnes PJ. 2011. Pathophysiology of allergic
search UK (C30122/A11527); CR UK/EPSRC/ inflammation. Immunol Rev 242:3150.
7. Johansson SG, Bennich H. 1967. Immunolog-
MRC/NIHR KCL/UCL Comprehensive Can-
ical studies of an atypical (myeloma) immuno-
cer Imaging Centre (C1519/A10331); KCL Ex- globulin. Immunology 13:381394.
perimental Cancer Medicine Centre, jointly 8. Ishizaka K, Ishizaka T, Hornbrook MM. 1966.
funded by Cancer Research UK, the National Physico-chemical properties of human reaginic
Institute for Health Research (NIHR), Welsh antibody. IV. Presence of a unique immunoglobu-
lin as a carrier of reaginic activity. J Immunol 97:
Assembly Government, HSC R&D Office for
7585.
Northern Ireland, and Chief Scientist Office, 9. Geha RS, Jabara HH, Brodeur SR. 2003. The
Scotland. The research was supported by the regulation of immunoglobulin E class-switch
NIHR Biomedical Research Centre based at recombination. Nat Rev Immunol 3:721732.
Guys and St. Thomas National Health Service 10. Gould HJ, Sutton BJ. 2008. IgE in allergy and
(NHS) Foundation Trust and Kings College asthma today. Nat Rev Immunol 8:205217.
11. Galli SJ, Tsai M. 2012. IgE and mast cells in
London.
allergic disease. Nat Med 18:693704.
Conflicts of interest: We disclose no con- 12. Dhaliwal B, Yuan D, Pang MO, Henry AJ,
flicts. Cain K, Oxbrow A, Fabiane SM, Beavil AJ,
The views expressed are those of the McDonnell JM, Gould HJ, Sutton BJ. 2012.
authors and not necessarily those of the NHS, Crystal structure of IgE bound to its B-cell
receptor CD23 reveals a mechanism of recip-
the NIHR, or the Department of Health.
rocal allosteric inhibition with high affinity
receptor FcepsilonRI. Proc Natl Acad Sci USA
109:1268612691.
CITATION 13. Kraft S, Kinet JP. 2007. New developments
in FcepsilonRI regulation, function and inhibi-
Karagiannis SN, Karagiannis P, Josephs DH,
tion. Nat Rev Immunol 7:365378.
Saul L, Gilbert AE, Upton N, Gould HJ. 2013. 14. Dullaers M, De Bruyne R, Ramadani F, Gould
IgE and allergy: antibodies in immune in- HJ, Gevaert P, Lambrecht BN. 2012. The who,
flammation and treatment. Microbiol Spec- where, and when of IgE in allergic airway
trum 1(1):AID-0006-2012. disease. J Allergy Clin Immunol 129:635645.
15. Acharya M, Borland G, Edkins AL, Maclellan
LM, Matheson J, Ozanne BW, Cushley W. 2010.
CD23/FcepsilonRII: molecular multi-tasking.
REFERENCES
Clin Exp Immunol 162:1223.
1. Galli SJ, Tsai M, Piliponsky AM. 2008. The 16. Dierks SE, Bartlett WC, Edmeades RL, Gould
development of allergic inflammation. Nature HJ, Rao M, Conrad DH. 1993. The oligomeric
454:445454. nature of the murine Fc epsilon RII/CD23.
96 KARAGIANNIS ET AL.

Implications for function. J Immunol 150: Comeau MR, Pearce EJ, Laufer TM, Artis D.
23722382. 2009. MHC class II-dependent basophil-CD4+
17. Vouldoukis I, Mazier D, Moynet D, Thiolat D, T cell interactions promote T(H)2 cytokine-
Malvy D, Mossalayi MD. 2011. IgE mediates dependent immunity. Nat Immunol 10:697
killing of intracellular Toxoplasma gondii by 705.
human macrophages through CD23-dependent, 28. Saenz SA, Siracusa MC, Perrigoue JG, Spen-
interleukin-10 sensitive pathway. PLoS One 6: cer SP, Urban JF Jr, Tocker JE, Budelsky AL,
e18289. Kleinschek MA, Kastelein RA, Kambayashi T,
18. Vouldoukis I, Riveros-Moreno V, Dugas B, Bhandoola A, Artis D. 2010. IL25 elicits a
Ouaaz F, Becherel P, Debre P, Moncada S, multipotent progenitor cell population that
Mossalayi MD. 1995. The killing of Leishman- promotes T(H)2 cytokine responses. Nature
ia major by human macrophages is mediated 464:13621366.
by nitric oxide induced after ligation of the Fc 29. Karagiannis SN, Warrack JK, Jennings KH,
epsilon RII/CD23 surface antigen. Proc Natl Murdock PR, Christie G, Moulder K, Sutton
Acad Sci USA 92:78047808. BJ, Gould HJ. 2001. Endocytosis and recycling
19. Chen HY, Liu FT, Yang RY. 2005. Roles of of the complex between CD23 and HLA-DR in
galectin-3 in immune responses. Arch Immunol human B cells. Immunology 103:319331.
Ther Exp (Warsaw) 53:497504. 30. Mudde GC, Bheekha R, Bruijnzeel-Koomen
20. Bax HJ, Keeble AH, Gould HJ. 2012. CA. 1995. Consequences of IgE/CD23-mediat-
Cytokinergic IgE action in mast cell activation. ed antigen presentation in allergy. Immunol
Front Immunol 3:229. Today 16:380383.
21. Ziegler SF, Artis D. 2010. Sensing the outside 31. Ying S, Humbert M, Meng Q, Pfister R, Menz
world: TSLP regulates barrier immunity. Nat G, Gould HJ, Kay AB, Durham SR. 2001.
Immunol 11:289293. Local expression of epsilon germline gene
22. Fort MM, Cheung J, Yen D, Li J, Zurawski transcripts and RNA for the epsilon heavy
SM, Lo S, Menon S, Clifford T, Hunte B, chain of IgE in the bronchial mucosa in atopic
Lesley R, Muchamuel T, Hurst SD, Zurawski and nonatopic asthma. J Allergy Clin Immunol
G, Leach MW, Gorman DM, Rennick DM. 107:686692.
2001. IL-25 induces IL-4, IL-5, and IL-13 and 32. Cameron L, Hamid Q, Wright E, Nakamura
Th2-associated pathologies in vivo. Immunity Y, Christodoulopoulos P, Muro S, Frenkiel S,
15:985995. Lavigne F, Durham S, Gould H. 2000. Local
23. Schmitz J, Owyang A, Oldham E, Song Y, synthesis of epsilon germline gene transcripts,
Murphy E, McClanahan TK, Zurawski G, IL-4, and IL-13 in allergic nasal mucosa after
Moshrefi M, Qin J, Li X, Gorman DM, Bazan ex vivo allergen exposure. J Allergy Clin
JF, Kastelein RA. 2005. IL-33, an interleukin- Immunol 106:4652.
1-like cytokine that signals via the IL-1 recep- 33. Fawaz LM, Sharif-Askari E, Hajoui O, Soussi-
tor-related protein ST2 and induces T helper Gounni A, Hamid Q, Mazer BD. 2007. Expres-
type 2-associated cytokines. Immunity 23:479 sion of IL-9 receptor alpha chain on human
490. germinal center B cells modulates IgE secre-
24. Lambrecht BN, Hammad H. 2010. The role of tion. J Allergy Clin Immunol 120:12081215.
dendritic and epithelial cells as master regulators 34. Karagiannis SN, Bracher MG, Beavil RL, Beavil
of allergic airway inflammation. Lancet 376:835 AJ, Hunt J, McCloskey N, Thompson RG, East
843. N, Burke F, Sutton BJ, Dombrowicz D, Balkwill
25. Sokol CL, Chu NQ, Yu S, Nish SA, Laufer FR, Gould HJ. 2008. Role of IgE receptors in IgE
TM, Medzhitov R. 2009. Basophils function antibody-dependent cytotoxicity and phagocytosis
as antigen-presenting cells for an allergen- of ovarian tumor cells by human monocytic cells.
induced T helper type 2 response. Nat Immunol Cancer Immunol Immunother 57:247263.
10:713720. 35. Karagiannis SN, Bracher MG, Hunt J,
26. Yoshimoto T, Yasuda K, Tanaka H, Nakahira McCloskey N, Beavil RL, Beavil AJ, Fear DJ,
M, Imai Y, Fujimori Y, Nakanishi K. 2009. Thompson RG, East N, Burke F, Moore RJ,
Basophils contribute to T(H)2-IgE responses Dombrowicz DD, Balkwill FR, Gould HJ. 2007.
in vivo via IL-4 production and presentation IgE-antibody-dependent immunotherapy of solid
of peptide-MHC class II complexes to CD4+ tumors: cytotoxic and phagocytic mechanisms of
T cells. Nat Immunol 10:706712. eradication of ovarian cancer cells. J Immunol
27. Perrigoue JG, Saenz SA, Siracusa MC, 179:28322843.
Allenspach EJ, Taylor BC, Giacomin PR, 36. Ishizaka T, Conrad DH, Schulman ES, Sterk
Nair MG, Du Y, Zaph C, van Rooijen N, AR, Ishizaka K. 1983. Biochemical analysis of
CHAPTER 5 Immunoglobulin E and Allergy 97

initial triggering events of IgE-mediated his- 48. Tsujimura Y, Obata K, Mukai K, Shindou H,
tamine release from human lung mast cells. Yoshida M, Nishikado H, Kawano Y, Minegishi
J Immunol 130:23572362. Y, Shimizu T, Karasuyama H. 2008. Basophils
37. Dombrowicz D, Flamand V, Brigman KK, Koller play a pivotal role in immunoglobulin-G-mediated
BH, Kinet JP. 1993. Abolition of anaphylaxis by but not immunoglobulin-E-mediated systemic
targeted disruption of the high affinity immuno- anaphylaxis. Immunity 28:581589.
globulin E receptor alpha chain gene. Cell 75:969 49. Finkelman FD, Urban JF Jr. 2001. The other
976. side of the coin: the protective role of the TH2
38. Davey AM, Krise KM, Sheets ED, Heikal cytokines. J Allergy Clin Immunol 107:772780.
AA. 2008. Molecular perspective of antigen- 50. Verwaerde C, Joseph M, Capron M, Pierce RJ,
mediated mast cell signaling. J Biol Chem 283: Damonneville M, Velge F, Auriault C, Capron A.
71177127. 1987. Functional properties of a rat monoclonal
39. MacNeil AJ, Yang YJ, Lin T-J. 2011. MAPK IgE antibody specific for Schistosoma mansoni.
kinase 3 specifically regulates Fc epsilonRI- J Immunol 138:44414446.
mediated IL-4 production by mast cells. J Immu- 51. Capron M, Capron A. 1994. Immunoglobulin
nol 187:33743382. E and effector cells in schistosomiasis. Science
40. Medina-Tamayo J, Sanchez-Miranda E, Balleza- 264:18761877.
Tapia H, Ambriz X, Cid ME, Gonzalez-Espinosa 52. Gounni AS, Lamkhioued B, Ochiai K, Tanaka Y,
D, Gutierrez AA, Gonzalez-Espinosa C. 2007. Delaporte E, Capron A, Kinet JP, Capron M.
Super-oxidized solution inhibits IgE-antigen- 1994. High-affinity IgE receptor on eosinophils
induced degranulation and cytokine release in mast is involved in defence against parasites. Nature
cells.IntImmunopharmacol7:10131024. 367:183186.
41. Nishida K, Yamasaki S, Ito Y, Kabu K, Hattori K, 53. Hagan P, Blumenthal UJ, Dunn D, Simpson
Tezuka T, Nishizumi H, Kitamura D, Goitsuka R, AJ, Wilkins HA. 1991. Human IgE, IgG4 and
Geha RS, Yamamoto T, Yagi T, Hirano T. 2005. resistance to reinfection with Schistosoma
Fc{epsilon}RI-mediated mast cell degranula- haematobium. Nature 349:243245.
tion requires calcium-independent microtubule- 54. Dunne DW, Butterworth AE, Fulford AJ,
dependent translocation of granules to the plasma Ouma JH, Sturrock RF. 1992. Human IgE
membrane. J Cell Biol 170:115126. (Erratum, responses to Schistosoma mansoni and resis-
171:177.) tance to reinfection. Mem Inst Oswaldo Cruz
42. Oka T, Sato K, Hori M, Ozaki H, Karaki H. 87(Suppl 4):99103.
2002. FcepsilonRI cross-linking-induced actin 55. Gurish MF, Bryce PJ, Tao H, Kisselgof AB,
assembly mediates calcium signalling in RBL- Thornton EM, Miller HR, Friend DS, Oettgen
2H3 mast cells. Br J Pharmacol 136:837846. HC. 2004. IgE enhances parasite clearance and
43. Oettgen HC, Martin TR, Wynshaw-Boris A, regulates mast cell responses in mice infected with
Deng C, Drazen JM, Leder P. 1994. Active Trichinella spiralis. J Immunol 172:11391145.
anaphylaxis in IgE-deficient mice. Nature 370: 56. Watanabe N, Bruschi F, Korenaga M. 2005.
367370. IgE: a question of protective immunity in Trich-
44. Miyajima I, Dombrowicz D, Martin TR, Ravetch inella spiralis infection. Trends Parasitol 21:175178.
JV, Kinet JP, Galli SJ. 1997. Systemic anaphylaxis 57. Davis SD, Schaller J, Wedgwood RJ. 1966.
in the mouse can be mediated largely through IgG1 Jobs Syndrome. Recurrent, cold, staphylo-
and Fc gammaRIII. Assessment of the cardiopul- coccal abscesses. Lancet i:10131015.
monary changes, mast cell degranulation, and 58. Freeman AF, Holland SM. 2009. Clinical
death associated with active or IgE- or IgG1- manifestations, etiology, and pathogenesis of
dependent passive anaphylaxis. J Clin Investig the hyper-IgE syndromes. Pediatr Res 65:32R
99:901914. 37R.
45. Jonsson F, Mancardi DA, Kita Y, Karasuyama H, 59. Woellner C, Gertz EM, Schaffer AA, Lagos M,
Iannascoli B, Van Rooijen N, Shimizu T, Daeron Perro M, Glocker EO, Pietrogrande MC, Cossu F,
M, Bruhns P. 2011. Mouse and human neutrophils Franco JL, Matamoros N, Pietrucha B,
induce anaphylaxis. J Clin Investig 121:14841496. Heropolitanska-Pliszka E, Yeganeh M, Moin M,
46. Daeron M. 1997. Fc receptor biology. Annu Espanol T, Ehl S, Gennery AR, Abinun M,
Rev Immunol 15:203234. Breborowicz A, Niehues T, Kilic SS, Junker A,
47. Jonsson F, Mancardi DA, Zhao W, Kita Y, Turvey SE, Plebani A, Sanchez B, Garty BZ,
Iannascoli B, Khun H, van Rooijen N, Shimizu Pignata C, Cancrini C, Litzman J, Sanal O,
T, Schwartz LB, Daeron M, Bruhns P. 2012. Baumann U, Bacchetta R, Hsu AP, Davis JN,
Human FcgammaRIIA induces anaphylactic and Hammarstrom L, Davies EG, Eren E, Arkwright
allergic reactions. Blood 119:25332544. PD, Moilanen JS, Viemann D, Khan S, Marodi L,
98 KARAGIANNIS ET AL.

Cant AJ, Freeman AF, Puck JM, Holland SM, hyper-IgE syndrome. J Allergy Clin Immunol
Grimbacher B. 2010. Mutations in STAT3 and 122:181187.
diagnostic guidelines for hyper-IgE syndrome. 69. Erazo A, Kutchukhidze N, Leung M, Christ
J Allergy Clin Immunol 125:424432. AP, Urban JF Jr, Curotto de Lafaille MA,
60. Su HC, Jing H, Zhang Q. 2011. DOCK8 Lafaille JJ. 2007. Unique maturation program
deficiency. Ann NY Acad Sci 1246:2633. of the IgE response in vivo. Immunity 26:191
61. Engelhardt KR, McGhee S, Winkler S, Sassi 203.
A, Woellner C, Lopez-Herrera G, Chen A, 70. Gevaert P, Nouri-Aria KT, Wu H, Harper CE,
Kim HS, Lloret MG, Schulze I, Ehl S, Thiel J, Takhar P, Fear DJ, Acke F, De Ruyck N,
Pfeifer D, Veelken H, Niehues T, Siepermann Banfield G, Kariyawasam HH, Bachert C,
K, Weinspach S, Reisli I, Keles S, Genel F, Durham SR, Gould HJ. 2013. Local receptor
Kutukculer N, Camcioglu Y, Somer A, revision and class switching to IgE in chronic
Karakoc-Aydiner E, Barlan I, Gennery A, rhinosinusitis with nasal polyps. Allergy 68:
Metin A, Degerliyurt A, Pietrogrande MC, 5563.
Yeganeh M, Baz Z, Al-Tamemi S, Klein C, 71. Chularojanamontri L, Wimoolchart S,
Puck JM, Holland SM, McCabe ER, Tuchinda P, Kulthanan K, Kiewjoy N. 2009.
Grimbacher B, Chatila TA. 2009. Large Role of omalizumab in a patient with hyper-
deletions and point mutations involving the IgE syndrome and review of dermatologic
dedicator of cytokinesis 8 (DOCK8) in the manifestations. Asian Pac J Allergy Immunol
autosomal-recessive form of hyper-IgE syn- 27:233236.
drome. J Allergy Clin Immunol 124:12891302. 72. Imani F, Proud D, Griffin DE. 1999. Measles
62. Freeman AF, Holland SM. 2010. Clinical virus infection synergizes with IL-4 in IgE
manifestations of hyper IgE syndromes. Dis class switching. J Immunol 162:15971602.
Markers 29:123130. 73. Rager KJ, Langland JO, Jacobs BL, Proud D,
63. Xiong H, Curotto de Lafaille MA, Lafaille JJ. Marsh DG, Imani F. 1998. Activation of anti-
2012. What is unique about the IgE response? viral protein kinase leads to immunoglobulin E
Adv Immunol 116:113141. class switching in human B cells. J Virol 72:
64. Minegishi Y, Saito M, Tsuchiya S, Tsuge I, 11711176.
Takada H, Hara T, Kawamura N, Ariga T, 74. Johnston SL, Pattemore PK, Sanderson G,
Pasic S, Stojkovic O, Metin A, Karasuyama Smith S, Lampe F, Josephs L, Symington P,
H. 2007. Dominant-negative mutations in the OToole S, Myint SH, Tyrrell DA, et al. 1995.
DNA-binding domain of STAT3 cause hyper- Community study of role of viral infections
IgE syndrome. Nature 448:10581062. in exacerbations of asthma in 911 year old
65. He J, Shi J, Xu X, Zhang W, Wang Y, Chen X, children. BMJ 310:12251229.
Du Y, Zhu N, Zhang J, Wang Q, Yang J. 2012. 75. Nicholson KG, Kent J, Ireland DC. 1993.
STAT3 mutations correlated with hyper-IgE Respiratory viruses and exacerbations of asthma
syndrome lead to blockage of IL-6/STAT3 in adults. BMJ 307:982986.
signalling pathway. J Biosci 37:243257. 76. Murray CS, Poletti G, Kebadze T, Morris J,
66. Milner JD, Brenchley JM, Laurence A, Woodcock A, Johnston SL, Custovic A. 2006.
Freeman AF, Hill BJ, Elias KM, Kanno Y, Study of modifiable risk factors for asthma
Spalding C, Elloumi HZ, Paulson ML, Davis exacerbations: virus infection and allergen
J, Hsu A, Asher AI, Oshea J, Holland SM, exposure increase the risk of asthma hospital
Paul WE, Douek DC. 2008. Impaired T(H)17 admissions in children. Thorax 61:376382.
cell differentiation in subjects with autosomal 77. Green RM, Custovic A, Sanderson G, Hunter
dominant hyper-IgE syndrome. Nature 452: J, Johnston SL, Woodcock A. 2002. Syner-
773776. gism between allergens and viruses and risk of
67. Dong C. 2008. TH17 cells in development: an hospital admission with asthma: case-control
updated view of their molecular identity and study. BMJ 324:763.
genetic programming. Nat Rev Immunol 8:337348. 78. Zambrano JC, Carper HT, Rakes GP, Patrie
68. Renner ED, Rylaarsdam S, Anover-Sombke S, J, Murphy DD, Platts-Mills TA, Hayden FG,
Rack AL, Reichenbach J, Carey JC, Zhu Q, Gwaltney JM Jr, Hatley TK, Owens AM,
Jansson AF, Barboza J, Schimke LF, Leppert Heymann PW. 2003. Experimental rhinovirus
MF, Getz MM, Seger RA, Hill HR, Belohradsky challenges in adults with mild asthma: re-
BH, Torgerson TR, Ochs HD. 2008. Novel signal sponse to infection in relation to IgE. J Allergy
transducer and activator of transcription 3 Clin Immunol 111:10081016.
(STAT3) mutations, reduced T(H)17 cell numbers, 79. Soto-Quiros M, Avila L, Platts-Mills TA,
and variably defective STAT3 phosphorylation in Hunt JF, Erdman DD, Carper H, Murphy
CHAPTER 5 Immunoglobulin E and Allergy 99

DD, Odio S, James HR, Patrie JT, Hunt W, fragment of human IgE bound to its high-affinity
ORourke AK, Davis MD, Steinke JW, Lu X, receptor Fc[epsi]RI[alpha]. Nature 406:259266.
Kennedy J, Heymann PW. 2012. High titers of 90. Beck LA, Marcotte GV, MacGlashan D,
IgE antibody to dust mite allergen and risk for Togias A, Saini S. 2004. Omalizumab-induced
wheezing among asthmatic children infected reductions in mast cell FcRI expression and
with rhinovirus. J Allergy Clin Immunol 129: function. J Allergy Clin Immunol 114:527530.
14991505. 91. Prussin C, Griffith DT, Boesel KM, Lin H,
80. Corne JM, Marshall C, Smith S, Schreiber J, Foster B, Casale TB. 2003. Omalizumab treat-
Sanderson G, Holgate ST, Johnston SL. 2002. ment downregulates dendritic cell FcepsilonRI
Frequency, severity, and duration of rhinovi- expression. J Allergy Clin Immunol 112:11471154.
rus infections in asthmatic and non-asthmatic 92. MacGlashan DW, Bochner BS, Adelman DC,
individuals: a longitudinal cohort study. Lancet Jardieu PM, Togias A, McKenzie-White J,
359:831834. Sterbinsky SA, Hamilton RG, Lichtenstein
81. Message SD, Laza-Stanca V, Mallia P, Parker LM. 1997. Down-regulation of Fc(epsilon)RI
HL, Zhu J, Kebadze T, Contoli M, Sanderson expression on human basophils during in vivo
G, Kon OM, Papi A, Jeffery PK, Stanciu LA, treatment of atopic patients with anti-IgE anti-
Johnston SL. 2008. Rhinovirus-induced lower body. J Immunol 158:14381445.
respiratory illness is increased in asthma and 93. Hunt J, Keeble AH, Dale RE, Corbett MK,
related to virus load and Th1/2 cytokine and Beavil RL, Levitt J, Swann MJ, Suhling K,
IL-10 production. Proc Natl Acad Sci USA 105: Ameer-Beg S, Sutton BJ, Beavil AJ. 2012. A
1356213567. fluorescent biosensor reveals conformational
82. Paul WE, Zhu J. 2010. How are T(H)2-type changes in human immunoglobulin E Fc: im-
immune responses initiated and amplified? plications for mechanisms of receptor binding,
Nat Rev Immunol 10:225235. inhibition, and allergen recognition. J Biol Chem
83. Papadopoulos NG, Stanciu LA, Papi A, Holgate 287:1745917470.
ST, Johnston SL. 2002. Rhinovirus-induced alter- 94. DAmato G. 2006. Role of anti-IgE monoclonal
ations on peripheral blood mononuclear cell antibody (omalizumab) in the treatment of bron-
phenotype and costimulatory molecule expression chial asthma and allergic respiratory diseases.
in normal and atopic asthmatic subjects. Clin Exp Eur J Pharmacol 533:302307.
Allergy 32:537542. 95. Leung DY, Sampson HA, Yunginger JW,
84. Perez-Yarza EG, Moreno A, Lazaro P, Mejias Burks AW Jr, Schneider LC, Wortel CH,
A, Ramilo O. 2007. The association between Davis FM, Hyun JD, Shanahan WR Jr. 2003.
respiratory syncytial virus infection and the Effect of anti-IgE therapy in patients with
development of childhood asthma: a systemat- peanut allergy. N Engl J Med 348:986993.
ic review of the literature. Pediatr Infect Dis J 96. van Neerven RJ, van Roomen CP, Thomas
26:733739. WR, de Boer M, Knol EF, Davis FM. 2001.
85. Sigurs N, Bjarnason R, Sigurbergsson F, Humanized anti-IgE mAb Hu-901 prevents the
Kjellman B, Bjorksten B. 1995. Asthma and activation of allergen-specific T cells. Int Arch
immunoglobulin E antibodies after respiratory Allergy Immunol 124:400402.
syncytial virus bronchiolitis: a prospective cohort 97. Poole JA, Meng J, Reff M, Spellman MC,
study with matched controls. Pediatrics 95:500 Rosenwasser LJ. 2005. Anti-CD23 monoclo-
505. nal antibody, lumiliximab, inhibited allergen-
86. Dakhama A, Lee YM, Ohnishi H, Jing X, induced responses in antigen-presenting cells
Balhorn A, Takeda K, Gelfand EW. 2009. and T cells from atopic subjects. J Allergy Clin
Virus-specific IgE enhances airway respon- Immunol 116:780788.
siveness on reinfection with respiratory syn- 98. Shiung YY, Chiang CY, Chen JB, Wu PC,
cytial virus in newborn mice. J Allergy Clin Hung AF, Lu DC, Pan RL, Chang TW. 2012.
Immunol 123:138145. An anti-IgE monoclonal antibody that binds
87. Chang TW. 2000. The pharmacological basis to IgE on CD23 but not on high-affinity IgE.
of anti-IgE therapy. Nat Biotechnol 18:157162. Fc receptors. Immunobiology 217:676683.
88. Adis International Ltd. 2002. Omalizumab: 99. Chu SY, Horton HM, Pong E, Leung IW,
anti-IgE monoclonal antibody E25, E25, Chen H, Nguyen DH, Bautista C, Muchhal
humanised anti-IgE MAb, IGE 025, monoclo- US, Bernett MJ, Moore GL, Szymkowski DE,
nal antibody E25, Olizumab, Xolair, rhuMAb- Desjarlais JR. 2012. Reduction of total IgE by
E25. BioDrugs 16:380386. targeted coengagement of IgE B-cell receptor
89. Garman SC, Wurzburg BA, Tarchevskaya SS, and FcgammaRIIb with Fc-engineered anti-
Kinet J-P, Jardetzky TS. 2000. Structure of the Fc body. J Allergy Clin Immunol 129:11021115.
100 KARAGIANNIS ET AL.

100. Abdulla NE, Ninan MJ, Markowitz AB. 2012. Safety profile, pharmacokinetics, and biologic
Rituximab: current status as therapy for malig- activity of MEDI-563, an anti-IL-5 receptor
nant and benign hematologic disorders. BioDrugs alpha antibody, in a phase I study of subjects
26:7182. with mild asthma. J Allergy Clin Immunol 125:
101. Simon D, Hosli S, Kostylina G, Yawalkar N, 12371244.
Simon HU. 2008. Anti-CD20 (rituximab) 113. Kolbeck R, Kozhich A, Koike M, Peng L,
treatment improves atopic eczema. J Allergy Andersson CK, Damschroder MM, Reed JL,
Clin Immunol 121:122128. Woods R, Dallacqua WW, Stephens GL,
102. Arkwright PD. 2009. Anti-CD20 or anti-IgE Erjefalt JS, Bjermer L, Humbles AA, Gossage
therapy for severe chronic autoimmune urti- D, Wu H, Kiener PA, Spitalny GL, Mackay
caria. J Allergy Clin Immunol 123:510511. CR, Molfino NA, Coyle AJ. 2010. MEDI-563, a
(Authors reply, 123:511.) humanized anti-IL-5 receptor alpha mAb with
103. Tanaka H, Nagai H, Maeda Y. 1998. Effect of enhanced antibody-dependent cell-mediated
anti-IL-4 and anti-IL-5 antibodies on allergic cytotoxicity function. J Allergy Clin Immunol
airway hyperresponsiveness in mice. Life Sci 125:13441353.
62:PL169PL174. 114. Kearley J, Erjefalt JS, Andersson C, Benjamin
104. Steinke JW. 2004. Anti-interleukin-4 therapy. E, Jones CP, Robichaud A, Pegorier S, Brewah
Immunol Allergy Clin N Am 24:599614. Y, Burwell TJ, Bjermer L, Kiener PA, Kolbeck
105. Piper E, Brightling C, Niven R, Oh C, R, Lloyd CM, Coyle AJ, Humbles AA. 2011.
Faggioni R, Poon K, She D, Kell C, May RD, IL-9 governs allergen-induced mast cell num-
Geba GP, Molfino NA. 2013. A phase 2 bers in the lung and chronic remodeling of the
placebo-controlled study of tralokinumab in airways. Am J Respir Crit Care Med 183:865
moderate-to-severe asthma. Eur Respir J 41: 875.
330338. 115. Parker JM, Oh CK, LaForce C, Miller SD,
106. Corren J, Lemanske RF, Hanania NA, Pearlman DS, Le C, Robbie GJ, White WI,
Korenblat PE, Parsey MV, Arron JR, Harris White B, Molfino NA. 2011. Safety profile and
JM, Scheerens H, Wu LC, Su Z, Mosesova S, clinical activity of multiple subcutaneous doses
Eisner MD, Bohen SP, Matthews JG. 2011. of MEDI-528, a humanized anti-interleukin-9
Lebrikizumab treatment in adults with asthma. monoclonal antibody, in two randomized phase
N Engl J Med 365:10881098. 2a studies in subjects with asthma. BMC Pulm
107. Lee JJ, McGarry MP, Farmer SC, Denzler Med 11:14.
KL, Larson KA, Carrigan PE, Brenneise IE, 116. Brightling C, Berry M, Amrani Y. 2008.
Horton MA, Haczku A, Gelfand EW, Leikauf Targeting TNF-alpha: a novel therapeutic
GD, Lee NA. 1997. Interleukin-5 expression in approach for asthma. J Allergy Clin Immunol
the lung epithelium of transgenic mice leads to 121:510.
pulmonary changes pathognomonic of asthma. 117. Wenzel SE, Barnes PJ, Bleecker ER, Bousquet
J Exp Med 185:21432156. J, Busse W, Dahlen SE, Holgate ST, Meyers
108. Stein ML, Villanueva JM, Buckmeier BK, DA, Rabe KF, Antczak A, Baker J, Horvath I,
Yamada Y, Filipovich AH, Assaad AH, Mark Z, Bernstein D, Kerwin E, Schlenker-
Rothenberg ME. 2008. Anti-IL-5 (mepolizumab) Herceg R, Lo KH, Watt R, Barnathan ES,
therapy reduces eosinophil activation ex vivo and Chanez P. 2009. A randomized, double-blind,
increases IL-5 and IL-5 receptor levels. J Allergy placebo-controlled study of tumor necrosis
Clin Immunol 121:14731483. factor-alpha blockade in severe persistent asth-
109. Adis International Ltd. 2008. Mepolizumab: ma. Am J Respir Crit Care Med 179:549558.
240563, anti-IL-5 monoclonal antibody - Glaxo- 118. Ivanciuc O, Schein CH, Braun W. 2002. Data
SmithKline, anti-interleukin-5 monoclonal anti- mining of sequences and 3D structures of aller-
body - GlaxoSmithKline, SB 240563. Drugs R D genic proteins. Bioinformatics 18:13581364.
9:125130. 119. Reisinger J, Horak F, Pauli G, van Hage M,
110. Robinson DS. 2013. Mepolizumab treatment Cromwell O, Konig F, Valenta R, Niederberger
for asthma. Expert Opin Biol Ther 13:295302. V. 2005. Allergen-specific nasal IgG antibodies
111. Walsh GM. 2009. Reslizumab, a humanized induced by vaccination with genetically modified
anti-IL-5 mAb for the treatment of eosinophil- allergens are associated with reduced nasal aller-
mediated inflammatory conditions. Curr Opin gen sensitivity. J Allergy Clin Immunol 116:347
Mol Ther 11:329336. 354.
112. Busse WW, Katial R, Gossage D, Sari S, Wang 120. Niederberger V, Horak F, Vrtala S, Spitzauer S,
B, Kolbeck R, Coyle AJ, Koike M, Spitalny Krauth MT, Valent P, Reisinger J, Pelzmann M,
GL, Kiener PA, Geba GP, Molfino NA. 2010. Hayek B, Kronqvist M, Gafvelin G, Gronlund H,
CHAPTER 5 Immunoglobulin E and Allergy 101

Purohit A, Suck R, Fiebig H, Cromwell O, Pauli Lund K, Wurtzen PA, Durham SR. 2012. Func-
G, van Hage-Hamsten M, Valenta R. 2004. tional rather than immunoreactive levels of IgG4
Vaccination with genetically engineered allergens correlate closely with clinical response to grass
prevents progression of allergic disease. Proc Natl pollen immunotherapy. Allergy 67:217226.
Acad Sci USA 101(Suppl 2):1467714682. 128. James LK, Bowen H, Calvert RA, Dodev TS,
121. Bordas-Le Floch V, Bussieres L, Airouche S, Shamji MH, Beavil AJ, McDonnell JM, Durham
Lautrette A, Bouley J, Berjont N, Horiot S, Huet SR, Gould HJ. 2012. Allergen specificity of IgG(4)-
A, Jain K, Lemoine P, Chabre H, Batard T, expressing B cells in patients with grass pollen
Mascarell L, Baron-Bodo V, Tourdot S, Nony E, allergy undergoing immunotherapy. J Allergy Clin
Moingeon P. 2012. Expression and characteriza- Immunol 130:663670.
tion of natural-like recombinant Der p 2 for 129. Pilette C, Nouri-Aria KT, Jacobson MR, Wilcock
sublingual immunotherapy. Int Arch Allergy LK, Detry B, Walker SM, Francis JN, Durham
Immunol 158:157167. SR. 2007. Grass pollen immunotherapy induces an
122. Chruszcz M, Pomes A, Glesner J, Vailes LD, allergen-specific IgA2 antibody response associat-
Osinski T, Porebski PJ, Majorek KA, Heymann ed with mucosal TGF-beta expression. J Immunol
PW, Platts-Mills TAE, Minor W, Chapman 178:46584666.
MD. 2012. Molecular determinants for antibody 130. Satoguina JS, Weyand E, Larbi J, Hoerauf A.
binding on group 1 house dust mite allergens. 2005. T regulatory-1 cells induce IgG4 production
J Biol Chem 287:73887398. by B cells: role of IL-10. J Immunol 174:47184726.
123. Marsh DG, Lichtenstein LM, Campbell DH. 1970. 131. Wood N, Bourque K, Donaldson DD, Collins M,
Studies on allergoids prepared from naturally Vercelli D, Goldman SJ, Kasaian MT. 2004. IL-21
occurring allergens. I. Assay of allergenicity and effects on human IgE production in response to
antigenicity of formalinized rye group I compo- IL-4 or IL-13. Cell Immunol 231:133145.
nent. Immunology 18:705722. 132. van der Neut Kolfschoten M, Schuurman J,
124. Ferrari E, Breda D, Longhi R, Vangelista L, Losen M, Bleeker WK, Martinez-Martinez P,
Nakaie CR, Elviri L, Casali E, Pertinhez TA, Vermeulen E, den Bleker TH, Wiegman L, Vink
Spisni A, Burastero SE. 2012. In search of a T, Aarden LA, De Baets MH, van de Winkel JG,
vaccine for mouse allergy: significant reduction of Aalberse RC, Parren PW. 2007. Anti-inflamma-
Mus m 1 allergenicity by structure-guided single- tory activity of human IgG4 antibodies by dynamic
point mutations. Int Arch Allergy Immunol 157: Fab arm exchange. Science 317:15541557.
226237. 133. Passalacqua G, Albano M, Riccio A, Fregonese L,
125. Valenta R, Linhart B, Swoboda I, Niederberger Puccinelli P, Parmiani S, Canonica GW. 1999.
V. 2011. Recombinant allergens for allergen- Clinical and immunologic effects of a rush sublin-
specific immunotherapy: 10 years anniversary gual immunotherapy to Parietaria species: a dou-
of immunotherapy with recombinant allergens. ble-blind, placebo-controlled trial. J Allergy Clin
Allergy 66:775783. Immunol 104:964968.
126. Canonica GW, Bousquet J, Casale T, Lockey RF, 134. Savilahti EM, Rantanen V, Lin JS, Karinen S,
Baena-Cagnani CE, Pawankar R, Potter PC, Saarinen KM, Goldis M, Makela MJ,
Bousquet PJ, Cox LS, Durham SR, Nelson HS, Hautaniemi S, Savilahti E, Sampson HA. 2010.
Passalacqua G, Ryan DP, Brozek JL, Compalati Early recovery from cows milk allergy is associ-
E, Dahl R, Delgado L, van Wijk RG, Gower RG, ated with decreasing IgE and increasing IgG4
Ledford DK, Filho NR, Valovirta EJ, Yusuf OM, binding to cows milk epitopes. J Allergy Clin
Zuberbier T, Akhanda W, Almarales RC, Immunol 125:13151321.
Ansotegui I, Bonifazi F, Ceuppens J, Chivato 135. Savilahti EM, Saarinen KM, Savilahti E. 2010.
T, Dimova D, Dumitrascu D, Fontana L, Duration of clinical reactivity in cows milk allergy
Katelaris CH, Kaulsay R, Kuna P, Larenas- is associated with levels of specific immunoglob-
Linnemann D, Manoussakis M, Nekam K, ulin G4 and immunoglobulin A antibodies to beta-
Nunes C, OHehir R, Olaguibel JM, Onder NB, lactoglobulin. Clin Exp Allergy 40:251256.
Park JW, Priftanji A, Puy R, Sarmiento L, 136. Harada K, Shimoda S, Kimura Y, Sato Y, Ikeda H,
Scadding G, Schmid-Grendelmeier P, Seberova Igarashi S, Ren XS, Sato H, Nakanuma Y. 2012.
E, Sepiashvili R, Sole D, Togias A, Tomino C, Significance of immunoglobulin G4 (IgG4)-posi-
Toskala E, Van Beever H, Vieths S. 2009. Sub- tive cells in extrahepatic cholangiocarcinoma:
lingual immunotherapy: World Allergy Organiza- molecular mechanism of IgG4 reaction in cancer
tion Position Paper 2009. Allergy 64(Suppl 91):1 tissue. Hepatology 56:157164.
59. 137. Karagiannis P, Gilbert AE, Josephs DH, Ali N,
127. Shamji MH, Ljorring C, Francis JN, Calderon Dodev T, Saul L, Correa I, Roberts L, Beddowes
MA, Larche M, Kimber I, Frew AJ, Ipsen H, E, Koers A, Hobbs C, Ferreira S, Geh JL, Healy C,
102 KARAGIANNIS ET AL.

Harries M, Acland KM, Blower PJ, Mitchell T, 144. Jensen-Jarolim E, Singer J. 2011. Why could
Fear DJ, Spicer JF, Lacy KE, Nestle FO, passive immunoglobulin E antibody therapy be
Karagiannis SN. 2013. IgG4 subclass antibodies safe in clinical oncology? Clin Exp Allergy 41:
impair antitumor immunity in melanoma. J Clin 13371340.
Investig 123:14571474. 145. CharacterisationSutton BJ, Beavil RL, Beavil
138. Karagiannis SN, Josephs DH, Karagiannis P, AJ. 2000. Inhibition of IgE-receptor interac-
Gilbert AE, Saul L, Rudman SM, Dodev T, tions. Br Med Bull 56:10041018.
Koers A, Blower PJ, Corrigan C, Beavil AJ, 146. Nigro EA, Brini AT, Soprana E, Ambrosi A,
Spicer JF, Nestle FO, Gould HJ. 2012. Recom- Dombrowicz D, Siccardi AG, Vangelista L.
binant IgE antibodies for passive immunotherapy 2009. Antitumor IgE adjuvanticity: key role of
of solid tumours: from concept towards clinical Fc epsilon RI. J Immunol 183:45304536.
application. Cancer Immunol Immunother 61: 147. Teng MW, Kershaw MH, Jackson JT, Smyth
15471564. MJ, Darcy PK. 2006. Adoptive transfer of chime-
139. Karagiannis SN, Wang Q, East N, Burke F, ric FcepsilonRI gene-modified human T cells for
Riffard S, Bracher MG, Thompson RG, Durham cancer immunotherapy. Hum Gene Ther 17:1134
SR, Schwartz LB, Balkwill FR, Gould HJ. 2003. 1143.
Activity of human monocytes in IgE antibody- 148. Kershaw MH, Darcy PK, Trapani JA,
dependent surveillance and killing of ovarian MacGregor D, Smyth MJ. 1998. Tumor-specific
tumor cells. Eur J Immunol 33:10301040. IgE-mediated inhibition of human colorectal car-
140. Gould HJ, Mackay GA, Karagiannis SN, cinoma xenograft growth. Oncol Res 10:133142.
OToole CM, Marsh PJ, Daniel BE, Coney 149. Teo PZ, Utz PJ, Mollick JA. 2012. Using the
LR, Zurawski VR Jr, Joseph M, Capron M, allergic immune system to target cancer: activity of
Gilbert M, Murphy GF, Korngold R. 1999. IgE antibodies specific for human CD20 and
Comparison of IgE and IgG antibody-dependent MUC1. Cancer Immunol Immunother 61:2295
cytotoxicity in vitro and in a SCID mouse xenograft 2309.
model of ovarian carcinoma. Eur J Immunol 29: 150. Spillner E, Plum M, Blank S, Miehe M, Singer
35273537. J, Braren I. 2012. Recombinant IgE antibody
141. Karagiannis SN, Nestle FO, Gould HJ. 2010. IgE engineering to target EGFR. Cancer Immunol
interacts with potent effector cells against tumors: Immunother 61:15651573.
ADCC and ADCP, p 185213. In Penichet ML, 151. Daniels TR, Leuchter RK, Quintero R, Helguera
Jensen-Jarolim E (ed), Cancer and IgE. Humana G, Rodriguez JA, Martinez-Maza O, Schultes
Press, Springer, New York, NY. BC, Nicodemus CF, Penichet ML. 2012. Targeting
142. Karagiannis P, Singer J, Hunt J, Gan SK, HER2/neu with a fully human IgE to harness the
Rudman SM, Mechtcheriakova D, Knittelfelder allergic reaction against cancer cells. Cancer
R, Daniels TR, Hobson PS, Beavil AJ, Spicer J, Immunol Immunother 61:9911003.
Nestle FO, Penichet ML, Gould HJ, Jensen- 152. Riemer AB, Untersmayr E, Knittelfelder R,
Jarolim E, Karagiannis SN. 2009. Charac- Duschl A, Pehamberger H, Zielinski CC,
terisation of an engineered trastuzumab IgE Scheiner O, Jensen-Jarolim E. 2007. Active
antibody and effector cell mechanisms targeting induction of tumor-specific IgE antibodies by
HER2/neu-positive tumour cells. Cancer Immunol oral mimotope vaccination. Cancer Res 67:3406
Immunother 58:915930. 3411.
143. Rudman SM, Josephs DH, Cambrook H, 153. Knittelfelder R, Riemer AB, Jensen-Jarolim E.
Karagiannis P, Gilbert AE, Dodev T, Hunt 2009. Mimotope vaccination--from allergy to
J, Koers A, Montes A, Taams L, Canevari S, cancer. Expert Opin Biol Ther 9:493506.
Figini M, Blower PJ, Beavil AJ, Nicodemus 154. Jensen-Jarolim E, Achatz G, Turner MC,
CF, Corrigan C, Kaye SB, Nestle FO, Gould Karagiannis S, Legrand F, Capron M, Penichet
HJ, Spicer JF, Karagiannis SN. 2011. ML, Rodriguez JA, Siccardi AG, Vangelista L,
Harnessing engineered antibodies of the IgE Riemer AB, Gould H. 2008. AllergoOncology: the
class to combat malignancy: initial assessment role of IgE-mediated allergy in cancer. Allergy
of FcRI-mediated basophil activation by a 63:12551266.
tumour-specific IgE antibody to evaluate the 155. Jensen-Jarolim E, Pawelec G. 2012. The nascent
risk of type I hypersensitivity. Clin Exp Allergy field of AllergoOncology. Cancer Immunol
41:14001413. Immunother 61:13551357.
ANTIBODY DISCOVERY
APPROACHES
Phage and Yeast Display

JARED SHEEHAN1 and WAYNE A. MARASCO1


6
DISCOVERY OF THERAPEUTIC ANTIBODIES USING
PHAGE DISPLAY TECHNOLOGY

Bacteriophage Biology and Antibody Display Method


Bacteriophage (phage) are viruses that infect and replicate within bacterial
cells. Filamentous phage particles inject single-stranded DNA into target
bacterial cells for subsequent replication and assembly of new virions
within the host cytoplasm. The filamentous phage species capable of
infecting Escherichia coli manifest as long, thin filaments that are secreted
from host bacteria without cell lysis. Due to their ease of manipulation and
stability in a range of temperatures and pH, F pilus-specific filamentous
phage species, including f1, fd, and M13, serve as reliable vehicles for combi-
natorial technologies, such as phage display (14).
A collection of the first human antibody libraries displayed on the phage
surface was published in the early 1990s (58). This antibody phage display
technology is based upon a large collection of human antibody genes subcloned
into an E. coli expression vector, which is packaged into filamentous phage. Upon
production from host bacterial cells, these phage particles display the antibody

1
Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School,
Boston, MA 02215.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0028-2014

105
106 SHEEHAN AND MARASCO

fragment on its surface as a fusion product with establish the library repertoire, the heteroge-
one of the viral coat proteins. This established neous pool of antibody genes is typically
library must be screened to isolate the phage derived from B cell mRNA from healthy
antibodies of interest. To ensure representation donors (nave library) or immunized donors
of all immunoglobulin variants within the or animals (immune library) (67). The vari-
library, a larger number of phage particles able fragments from the donated B cell cDNA
above the total number of unique antibody are amplified using germline gene family-
sequencesis typically screened. This fold- based primers (8). These heavy (VH) and
excess in selection experiments is easily light (VL) chain gene copies are randomly
achieved since filamentous phage can produce associated through cloning methods involving
titers up to 1013 particles per ml of culture (1). specific expression vectors depending upon
The purpose of antibody display library the antibody fragment format to be displayed
selections is to isolate an antibody variant with on the phage surface (discussed in Antibody
the desired functional properties. A purified, Phage Display Formats, below). In addition,
heterogeneous population of phage library synthetic libraries may be established through
members is screened to identify and enrich prearranged V, D, and J gene associations and
for the antibody sequences of interest. This VH/VL pairings, based upon desired immuno-
phage library selection typically involves the genetic profiles or biochemical properties.
capture of the target antigen, either by immo-
bilization on a plastic surface (solid phase) or Nave Phage Antibody Libraries
coupled to magnetic beads (solution phase). While nonimmunized human B cell donor
When incubated with this captured antigen, sources are typically peripheral blood mono-
the phage surfacespurified virions in solu- nuclear cells, other sources of a librarys
tionallow binding of specific antibody vari- antibody gene repertoire include spleen,
ants. Multiple washes remove nonbinding tonsils, bone marrow, and murine peripheral
phage particles, while attached phage are blood mononuclear cells (7, 912). The advan-
typically eluted under either low- or high-pH tage of the nave library lies in the diversity of
conditions to disrupt the antibody-antigen the antigen-unbiased variants, which can be
association. Following this elution step, these used for selections against a large panel of
phage antibodies are used to infect an E. coli targets, including self and toxic antigens (7, 10,
culture for amplification of the antigen-reac- 13). The quality of monoclonal antibodies
tive library subpopulation. The purified, am- (mAbs) isolated from this type of nave library
plified phage antibodies from this initial is dependent upon the total population num-
selection event are used for subsequent rounds ber, with large, diverse libraries of 1010
of panning. Due to the possibility of nonspe- unique variants permitting the discovery of
cific binding, at least two to three rounds of human antibodies with subnanomolar binding
this type of iterative library selection are affinities (10). The availability of large, pre-
performed to enrich for positive binders and established collections of nonimmune immu-
reduce background binding events. An over- noglobulin gene pools has made the creation of
view of this phage library selection procedure nave phage antibody libraries more conve-
is outlined in Fig. 1. nient than immune libraries (12).

Immune Phage Antibody Libraries


Sources of Antibody Genes for
In addition to these nave sources, a phage
Display Libraries
librarys antibody gene pool may be created
The quality and diversity of a phage librarys from human donors or animals immunized
total antibody repertoire is dependent upon with an antigen of interest. This type of
the source of immunoglobulin genes. To immune library is enriched for antigen-reactive
CHAPTER 6 Phage and Yeast Display 107

FIGURE 1 Overview of phage antibody library production and selections. (A) The phage antibody library
repertoire is derived from the B cells of nave or immune donors. The amplied IGHV and IGLV/IGKV genes
are subcloned into a phage display vector for E. coli expression and library production. (B) The phage
antibody library is selected against an immobilized target antigen. After washing to remove nonbinders,
the Ag-reactive phage antibodies are eluted, amplied, and reselected through subsequent rounds. ELISA
screens identify monoclonal, Ag-binding phage antibodies, whose heavy and light chain antibody genes
are subcloned into mammalian expression vectors. doi:10.1128/microbiolspec.AID-0028-2014.f1

variants with populations of affinity matured cantly more mAbs can be isolated from a
VH and VL gene fragments, due to the host single immunized donor through an immune
immune system machinery (6, 12, 14). Unlike phage antibody library (12).
nave gene pools, immune phage libraries are
comprised of antibody genes that are biased Synthetic Phage Antibody Libraries
toward the initial antigen. While this anti- An antibody gene repertoire is artificially
genic bias limits the scope of targets for this designed, prior to cloning into a phage expres-
type of library, immune gene pools are sion vector, to create a synthetic phage display
composed of a higher frequency of antigen- library. Usually, the in vitro assembly of novel
reactive library members at the onset of immunoglobulin genes involves a designed
selections. Further, immune phage antibody randomization of complementary determining
libraries permit the monitoring of natural regions (CDRs) into V, D, and J segments (17).
immunoglobulin responses, functioning as The heavy chain CDR3 is considered the most
diagnostics in patients with autoimmune hypervariable of all antigen-contacting loops,
disorders and viral infections (15, 16). Com- causing this region to be targeted for extensive
pared with hybridoma campaigns, signifi- manipulation (18).
108 SHEEHAN AND MARASCO

Although several optimized phage libraries


are established, two examples of modern,
synthetic, and semi-synthetic phage display
libraries are frequently described in the liter-
ature as discovery tools. One corporation
created a phage display library that combines
both DNA sequences from synthetic design
and nave human donors. For this semi-
synthetic antibody library, donor-sourced
fragments comprise the VH CDR3 and com-
plete VL, while in vitro synthesis of V segment FIGURE 2 Types of phage antibody display. (A)
DNA introduces designed diversity in VH Monovalent display with the scFv or Fab fusion
(green circle) to truncated pIII along with wild-
CDR1 and CDR2 (19). Another group devel- type copies of pIII (purple circles). This monova-
oped synthetic phage display libraries by lent mAb display format can also be used with
modifying the sequence and length of all six pVII (olive) or pIX (light blue) separately. (B)
CDRs based upon their context in natural Multivalent display with the scFv or Fab fusion
human antibody gene families (2022). to all copies of truncated pIII. Multivalent mAb
display is also possible with the major coat
protein pVIII (black border) separate from pIII.
Antibody Phage Display Formats The pVI (red circles) coat proteins are also present
in these diagrams. doi:10.1128/microbiolspec.
Previous reports describe phage display librar- AID-0028-2014.f2
ies as fusion products with all five of the pIII,
pVI, pVII, pVIII, and viral coat proteins (5, 13,
2326). Fusion products of the minor coat pIII domains, respectively (5). The establishment
either a truncated or full-length protein of modern, optimized, human antibody phage
represent the majority of human antibody display libraries supports the contemporary
display libraries. The surface protein pIII is role of this combinatorial technology in ongo-
an integral membrane protein that is necessary ing antibody discovery (1922).
for phage particle assembly and release from
host cells (2). Since only the pIII C-terminus is scFv-pIII Display
necessary for new virion construction, a Within this scFv-pIII display technology, the
human antibody or antibody fragment can be single-chain antibodies can be expressed as
expressed as a fusion product with this monovalent or multivalent fusion products on
truncated viral protein (3, 5, 13). Schematics the phage surface. The majority of pIII fusion
of these phage protein-scFv fusions are de- libraries are based on monovalent (phagemid)
tailed in Fig. 2. vectors compared to multivalent (phage)
Two of the primary pIII antibody display vectors (27). To rescue the viral particles
library types involve human single-chain var- from host bacterial cells, the phagemid vector
iable fragment (scFv) or fragment antigen- system requires the addition of other wild-type
binding (Fab) formats. The human scFv format phage genes and proteins via helper phage
consists of a heavy chain variable (VH) domain infection (28). This helper phage infection
and light chain variable (VL) domain ex- introduces a wild-type copy of pIII, which
pressed as a single polypeptide connected by outcompetes the scFv-pIII fusion during par-
a flexible linker region, with no associated ticle incorporation. This preference for wild-
constant domains. With Fab libraries, the type pIII results in a recovered phage particle
heavy and light chains are expressed as population whose majority displays no scFv-
separate gene products from the same vector pIII fusion on the viral surface, with the
with constant heavy 1 and constant light second most-frequent population bearing a
CHAPTER 6 Phage and Yeast Display 109

single copy of the fusion product (27). Despite light chain constant domainpreferably the
this wild-type dominance, the monovalent constant heavy 1 to avoid anchored VL dimers
scFv-pIII display technique allows for the is C-terminally fused to pIII. The non-
selection of higher-affinity antibodies due to anchored partner chain is secreted into the
single-copy competition among library vari- periplasmic region, where V H /V L chain
ants during iterative rounds of selection. This pairing occurs to form a complete Fab on the
monovalent competition requires at least two phage surface (12). While the reduced size of
to three successive rounds of panning to enrich the scFv fragment provides for increased
for scFv-pIII variants over wild-type pIII genetic stability over Fab libraries, many
phage particles. scFvs are observed to oligomerize, negatively
A multivalent scFv-pIII library can be impacting selection strategies (12, 13).
created through a helper phage population Structural properties of the Fab fragment
with a gene III deletion, delivery of a helper have desirable features for certain antibody
plasmid, or the subcloning of scFv genes into a discovery processes. Specifically, the presence
true phage vector (13, 27, 2931). The multiva- of the constant heavy 1 and constant light
lent display of scFv-pIII fusion proteins domains may influence conformational char-
permits for the selection of library isolates acteristics of the VH and VL fragments. Com-
based upon avidity, rather than affinity. In pared with the scFv format, certain reports
addition, these phage library-derived variants have described the increased capacity of
demonstrate a stronger functional affinity selected Fabs to retain antigen specificity
when the target antigen is multivalent, such when converted to a full-length IgG antibody
as the environment during solid phase selec- (34). In addition to favorable biochemical
tions (27). An advantage of this multivalent properties, in vivo studies demonstrated the
display strategy is the reduced number of biological potential of Fab antibody fragments.
iterative rounds of selection required to isolate Efficacy studies in a rheumatoid arthritis
antigen-reactive clones. Selection campaigns mouse model reported no significant thera-
using a multivalent scFv-pIII library yield peutic difference between Fab or IgG1
antigen-binding populations after only a single treatments (35). In terms of antibody develop-
round of panning, with antigen-reactive fre- ment, one study described the Fab format as a
quencies of >30% observed by the second reliable platform during affinity maturation
round (27). Similar selections with monovalent efforts. The improvements in binding charac-
phagemid libraries require at least three teristics observed in particular Fab conforma-
rounds of selection before reaching this fre- tions were retained following the conversion
quency of positive binding events. Despite this to full-length IgG. (36). These findings suggest
accelerated selection process, multivalent that the Fab format is a successful antibody
phage library vectors suffer poorer transfor- fragment surrogate for the selection, predic-
mation efficiencies, resulting in smaller, less tion, and optimization of IgG candidates.
diverse antibody gene pools (27).
Antibodies for Infectious Diseases
Fab-pIII Display
Discovered by Phage Display
Early pioneering work in establishing phage
display included libraries expressing Fab frag- The continuing emergence of novel strains
ments for selections (5, 9, 32, 33). Modern and drug-resistant microbial pathogens has
phage libraries displaying Fab fragments are created persistent threats to public health.
well established and engineered for improved The ongoing discovery of mAbs directed
biochemical and expression properties for against targets of infectious agents presents
therapeutic discovery (1922). For displaying clinical opportunities for patients. These anti-
Fabs on the phage surface, either the heavy or microbial mAbs are typically well tolerated
110 SHEEHAN AND MARASCO

in patients and demonstrate high specificity initially evaluated through designed in vitro
for the targeted pathogen, reducing the experiments. Following these primary bio-
opportunity for cross-reactivity with host assays, the in vivo protection of lead candidate
or nontarget agents. mAbs is determined through viral challenge
The in vitro discovery technology of studies in animals. Potential mechanisms of
phage display is one of the most promising viral neutralization include the inhibition of
vehicles for identifying antimicrobial, ther- virus attachment to host cell receptors, the
apeutic antibodies. This selection method blockage of membrane fusion events releasing
allows for the opportunity to isolate mAbs viral genetic material, and prevention of new
against toxic species or antigens, a threat- virion release from infected cells (42, 43).
ening characteristic not suitable for murine Outside of direct inactivation, nonneu-
hybridoma campaigns. Previous phage dis- tralizing antiviral mAbs may bind viral anti-
play approaches describe the discovery of gens on the cell surface during progeny release,
antiviral and antibacterial mAbs. recruiting cytotoxic cells or complement pro-
teins to offer in vivo protection. These findings
Antiviral mAbs Discovered by fueled the ongoing debate surrounding the
Phage Display correlation between in vitro neutralization and
The efforts of antiviral research are aimed at a in vivo protection for antibodies (41, 44). Since
fluid pathogenic landscape whose future the principal fraction of nonneutralizing mAbs
characteristics are difficult to predict. Among fail to clear viral infections in vivo, however,
all human pathogens, viruses are the best- the emphasis during antiviral mAb discovery
suited pathogens to evade host immune pres- should be placed on neutralizing immuno-
sure. Viral mutational robustness allows for globulins (41). A brief summary of antiviral
antigenic drift, where escape mutations within mAbs discovered using phage antibody librar-
targeted epitopes result in progeny with ies is listed in Table 1.
increased resistance to neutralizing mAbs or
small molecules (37). Furthermore, coin- Anti-Influenza mAbs Discovered by
fections with two or more viral strains allow Phage Display
for antigenic shift through gene transfer, One of the most popular targets for recent
establishing novel pathogens and complicating antiviral mAb discovery is the influenza virus.
vaccine and treatment strategies (3840). Due to the absence of a universal vaccine and
These continuous modes of viral evolution the ongoing emergence of new strains as
necessitate the development of new treatment human pathogens, the discovery of protective
options, which include the discovery of antiviral mAbs against influenza remains relevant.
mAbs. Influenza A virusesthe group of influenza
The targets of early antiviral mAb discovery viruses most closely associated with human
using phage display included human immuno- infectionsare classified within the classical
deficiency virus type 1 (HIV-1), respiratory subtypes of H1-H16 and N1-N9 based upon
syncytial virus, and herpes simplex virus (14, structural and antigenic characteristics of the
16). Surface viral glycoproteins are preferred hemagglutinin (HA) and neuraminidase viral
targets for phage selection campaigns, given glycoproteins (45). The HA molecule is the
their extracellular exposure, typically soluble major surface envelope protein and is respon-
character, and critical roles in the viral life sible for mediating attachment to sialic acid on
cycle (41). Viral neutralization is a significant the target cell surface (46). The globular head
trait under consideration during the charac- of HA, which contains the sialic acid binding
terization of mAbs isolated from selections domain, is amenable to mutations to avoid
against viral antigens. This capacity of antiviral immune pressure without significant costs to
mAbs to bind and inactivate viral particles is viral fitness (41, 47). The HA stem domain is
CHAPTER 6 Phage and Yeast Display 111

TABLE 1 Summary of antiviral antibodies discovered using phage displaya


Ab fragment
Viral family Virus Viral target displayed Ab source Reference

Coronaviridae MERS-CoV Spike glycoprotein scFv Nave 72


MERS-CoV Spike receptor binding Fab Nave 73
domain (RBD)
SARS-CoV Spike, S1 domain scFv Nave 65
SARS-CoV Spike RBD Fab Nave 66
Filoviridae Ebola virus Inactivated virus Fab Immune 78
Flaviviridae Dengue virus mAb-captured dengue scFv Nave 76
virus
Dengue virus Nonstructural protein 5 Fab Nave 77
(NS5)
West Nile virus E glycoprotein scFv Nave 74
West Nile virus Inactivated virus, VLP, scFv Immune 75
or E glycoprotein
Herpesviridae Herpes simplex virus mAb-captured gB and Fab Immune 81
gD glycoproteins
Orthomyxoviridae Inuenza A virus Hemagglutinin (HA) scFv Nave 4951
Inuenza A virus HA scFv Semi-synthetic 52
Inuenza A virus HA scFv Immune 53
Inuenza A and B HA scFv Immune 51
viruses
Paramyxoviridae Respiratory syncytial F glycoprotein Fab Immune 7980
virus
Retroviridae HIV-1 gp120 Fab Immune 59, 60,
63
HIV-1 gp41 Fab Immune 6163
a
Note: This list is a summary of selected publications and is not to be considered complete.

involved in the pH-dependent endosomal heterosubtypic neutralization for stem-directed


membrane fusion event and is a more con- anti-HA mAbs (52).
served region of the viral glycoprotein and is Phage display experiments using influenza
less tolerant of amino acid substitutions (48). HA as the selection antigen have also identified
Seminal phage display studies using puri- neutralizing antibodies directed against the
fied, recombinant HA protein as the selection globular head of the glycoprotein. While
antigen reported whole panels of broadly these antibodies are capable of binding HA
neutralizing human mAbs (4951). The broad- proteins across multiple viral subtypes, globu-
ly neutralizing mAbs isolated from these phage lar head-directed mAbs tend to be less hetero-
library selections are overwhelmingly biased reactive, given the lack of strict conservation
toward the conserved stem domain, pre- among these epitopes. Studies involving phage
venting endosomal membrane fusion but per- library selections yielded head-directed mAbs
mitting viral attachment. Given the stem capable of neutralizing single or multiple
region conservation and epitope proximity, subtypes, including H1N1, H2N2, and H3N2
the potent neutralization among these phage viruses (5354). Despite this limited scope of
display-derived mAbs is observed across mul- reactivity, mAbs that are neutralizing to the
tiple subtypes of influenza viruses. Following globular head are typically more potent, a trend
up on these initial studies, selections using a likely due to epitope availability on the viral
semi-synthetic phage library identified specif- surface (41). Neutralization escape variants are
ic immunoglobulin signatures associated with more easily generated by immune pressure on
112 SHEEHAN AND MARASCO

the globular head, however, as the loops and heavily on single B cell sorting or reverse
glycosylation sites allow this region to tolerate transcriptase PCR techniques and the screen-
mutations (55). ing of in vitro-activated B cells (41).

Anti-HIV-1 mAbs Discovered by Anticoronavirus mAbs Discovered by


Phage Display Phage Display
Another well-documented target for phage The severe acute respiratory syndrome
display library selections is HIV-1. The prima- coronavirus (SARS-CoV) predated its Middle
ry target of anti-HIV-1 humoral responses is East respiratory syndrome coronavirus
the trimeric Env protein of the viral surface. (MERS-CoV) relative as an emerging human
The gp120 protein of this Env complex binds pathogen during the early 2000s. A potent
the CD4 molecule and the CCR5 or CXCR5 neutralizing antibody was identified from
coreceptors on CD4+ T cells and macrophages. panning experiments with purified, truncated
This engagement causes a conformational S1 domain-only antigen (65). Other selection
change in the Env complex to induce the studies produced a similarly neutralizing mAb
membrane fusion activity of the gp41 compo- (66). These phage display studies using SARS-
nent (56). One of the primary challenges in CoV antigen fragments influenced the future
developing an HIV-1 vaccine strategy or strategies surrounding the current MERS-CoV
broadly neutralizing mAbs is the extensive emerging infectious disease.
genetic diversity, with up to 10% Env gene A human pathogen recently targeted by
variability within a single patient (57). Similar phage display studies is the emerging MERS-
to influenza HA, the HIV-1 Env trimer utilizes CoV. Similar to its SARS-CoV Coronaviridae
several mechanisms to avoid neutralizing family member, the MERS-CoV strains are
mAbs, including masking of critical epitopes, capable of residing in other mammalian spe-
amino acid mutations, and glycan shielding cies, including bats and camels (67, 68). The
without loss of protein activity (58). MERS-CoV uses the major envelope spike (S)
Among the collection of first-generation glycoprotein trimer for attachment and entry
anti-HIV-1 mAbs, several neutralizing library into target cells, with the S1 domain containing
variants, including mAb b12, were isolated the dipeptidyl peptidase 4 (DPP4) receptor
using phage display technology (59, 60). binding site and the S2 region mediating
Although these discoveries provided initial membrane fusion (69, 70). Previous reports
data related to the CD4 binding site epitope, involving infected patients describe the pro-
these mAbs are limited in their scope of duction of S protein-directed neutralizing
neutralization, with mAb b12 neutralizing antibodies (71).
only one third of total HIV-1 isolates (41). Recently published studies describe phage
More recent phage library selections against library selections to identify panels of neutral-
the viral gp120 and gp41 antigen yielded mAbs izing mAbs. Selections against full-length
neutralizing a diverse collection of primary MERS-CoV S protein using scFv-pIII display
isolates and subtypes (6163). In addition, the libraries yielded neutralizing antibodies local-
immunization schedule using HIV-1 antigens izing to the receptor binding domain. Despite
influences the selection quality of anti-HIV-1 the presence of the full-length protein during
mAbs from immune phage libraries, consistent selections, this campaign resulted in S1 do-
with earlier findings regarding immune librar- main-specific mAbs, which neutralized MERS-
ies capacity for tracking natural humoral CoV pseudotyped virus-like particles by
responses in humans (64). While these phage blocking the DPP4 binding site on the viral
display-related studies offered primary map- glycoprotein (72). Similarly, Fab-pIII library
ping data and early vaccine strategies, current selections against truncated DPP4 receptor
anti-HIV-1 mAb discovery efforts rely more binding domain-only antigen yielded mAbs
CHAPTER 6 Phage and Yeast Display 113

capable of neutralizing both pseudotyped and global threats to both healthy individuals and
live MERS-CoV (73). These studies provide infected patients. Specifically, the misuse of
examples of the ongoing relevance of phage antimicrobial drugs drives bacterial evolution
display technology in current, emerging infec- such that antibiotics are becoming increasingly
tious diseases. ineffective among Gram-negative and Gram-
positive strains (8285). Previous clinical
Other Antiviral mAbs Discovered by observations support the role of passive
Phage Display immunotherapy as prophylactic and therapeu-
In addition to the viruses discussed above, tic strategies against bacterial pathogens and
mAbs directed against other viral pathogens their toxins (86).
are detailed in the literature. Initial studies Phage display technologies allow for anti-
involving phage library selections against body library selections against bacterial anti-
purified West Nile Virus (WNV) envelope E gens, infectious agents which are impractical
glycoprotein produced neutralizing antibody for in vivo approaches such as hybridoma
sequences (74). Additional neutralizing mAbs campaigns. Unlike approaches with viral
were isolated from phage library selections pathogens, successful phage display efforts
using inactivated WNV or virus-like particles surrounding antibacterial mAb discovery
as well as immobilized E glycoprotein (75). A involve secreted toxins as selection antigens,
total of nine unique antibody sequences were rather than surface or structural proteins.
identified from phage library selections against These species-specific exotoxins cause pa-
Ab-captured dengue virus particles; all tient symptoms including diarrhea, renal
members of this mAb panel demonstrated failure, paralysis, and death (87).
some degree of cross-reactivity among the In terms of these secreted toxins, mAb-
four dengue virus serotypes (76). Recent based antibacterial therapies offer several
phage library panning experiments using pu- advantages over antibiotic compounds. These
rified dengue nonstructural protein 5 generat- antibiotic drugs do not demonstrate the ability
ed a collection of cross-reactive mAbs to eliminate pro-inflammatory bacterial fac-
intended for future mechanistic studies of the tors or cell fragments, which can be neutral-
viral replication machinery (77). ized or cleared through mAb binding. This
Outside of flaviviruses and coronaviruses, outcome may permit disease progression de-
other viral pathogens previously served as spite the removal of the bacterial pathogen.
targets of early phage display campaigns. Further, toxin neutralization mechanisms
Experiments involving inactivated Ebola create more feasible preclinical models for
virus isolates generated one antiglycoprotein evaluating novel mAbs, accelerating their
mAb capable of neutralizing the virus in vitro discovery (87). A brief summary of antibacte-
(78). Neutralizing mAbs showing in vivo rial mAbs discovered using phage antibody
protection against respiratory syncytial virus libraries is listed in Table 2.
were isolated from preliminary Fab library
selections against the viral F glycoprotein (79 Antibotulinum mAbs Discovered by
80). Similarly, neutralizing mAbs preventing Phage Display
the cell-to-cell transmission of herpes simplex The Clostridium genus encompasses a group
virus were discovered from pannings against of Gram-positive bacteria species capable of
glycoprotein D (81). secreting toxins. Clostridium botulinum pro-
duces botulinum neurotoxins (BoNTs) of
Antibacterial mAbs Discovered by serotypes A-G, with the A, B, and E serotypes
Phage Display most frequently causing human botulism (88).
The ongoing development of bacterial resis- These BoNTs act at cholinergic nerve endings
tance to antimicrobial agents continues to pose to prevent acetylcholine release, providing the
114 SHEEHAN AND MARASCO

TABLE 2 Summary of antibacterial antibodies discovered using phage displaya


Ab fragment
Bacterial family Bacterial species Bacterial target displayed Ab source Reference

Bacillaceae Bacillus anthracis Protective antigen scFv Nave 94


Bacillus anthracis Lethal factor scFv Immune 95
Bacillus anthracis Edema factor Fab Immune 96
Clostridiaceae Clostridium botulinum Botulinum neurotoxin scFv Immune 88
(BoNT) subtypes AE and nave
Clostridium botulinum BoNT/A heavy chain scFv Immune 90
Clostridium difcile Toxin B scFv Immune 97
Clostridium tetani Tetanus toxoid Fab Nave 98
Staphylococcaceae Staphylococcus aureus GrfA peptide scFv Immune 99
fragments
a
Note: This list is a summary of selected publications and is not to be considered complete.

paralytic effects of these exotoxins (89). Phage Phage library selections against recombi-
library selections against BoNT serotypes A-E nant PA resulted in the discovery of raxi-
produced neutralizing mAbs, with the suc- bacumab (ABthrax), a human IgG1 mAb that
cessful clones preferentially isolated from binds PA to inhibit cellular delivery of LF and
immune sources (88, 90). The most potent of EF to prevent and treat inhalational anthrax
these neutralizing mAbs were directed against (94). Prior to FDA approval, this anti-PA mAb
the BoNT heavy chain C-terminus. The ad- demonstrated in vivo protection in both
ministration of any mAb pair recognizing rabbits and monkeys. An anti-LF scFv pro-
nonoverlapping heavy chain epitopes provided duced from phage library panning demon-
complete protection during murine in vivo strated in vitro toxin neutralization and in vivo
toxin neutralization experiments, while single protection (95). Phage library selections
antibody delivery resulted in significant via- against recombinant EF yielded an anti-EF
bility loss (91). The ongoing mAb discovery mAb that protected mice from edema and
surrounding BoNTs remains important, given death during lethal dose administration (96).
their classification as high-risk agents as These phage display studies continue to be
bioweapons (91). valuable in defining protective, biodefense
strategies.
Antianthrax mAbs Discovered by Phage
Display Other Antibacterial mAbs Discovered by
Similar to the C. botulinum BoNTs, the Gram- Phage Display
positive Bacillus anthracis, the causative Bacterial pathogens other than C. botulinum
agent of anthrax, is considered a public safety and B. anthracis are described as targets of
threat because of its use as a biological phage display experiments. A relative of C.
weapon (92). Upon establishing an infection botulinum, the Clostridium difficile pathogen is
within lymph nodes, dividing bacilli release the leading cause of antibiotic-associated
three exotoxins: protective antigen (PA), diarrhea, with increasing severity due to the
lethal factor (LF), and edema factor (EF). ongoing emergence of hypervirulent strains
The PA toxin binds cellular receptors for (87). Disease onset occurs by production of
delivery of LF or EF into the host cytosol. The toxins A and B following intestinal coloniza-
PA toxin may combine with LF to form lethal tion. Phage library panning experiments
toxin or EF to create the edema toxin, both of against C. difficile toxin B generated a mAb,
which induce the toxic symptoms associated with improved affinity over the parent anti-
with anthrax disease (93). body, intended for use as a novel immunodi-
CHAPTER 6 Phage and Yeast Display 115

agnostic agent (97). Clostridium tetani, another yeast cell wall through two disulfide bonds to
member of this Clostridium genus, establishes Aga1p (100). The cell wall of each yeast cell can
infection through open wounds and secretes display 104 to 105 agglutinins (100, 102). Since
tetanus toxoid to induce painful muscle rigid- the development of the yeast display technol-
ity and spasms. A mAb isolated from library ogy, S. cerevisiae has served as the most
selections against tetanus toxoid demonstrat- commonly used species in this combinatorial
ed in vitro neutralization of this exotoxin (98). strategy.
While the majority of successful phage To establish a yeast antibody display li-
library panning campaigns involve exotoxins, brary, the heterogeneous immunoglobulin
less successful experiments involving bacterial gene pool is cloned into the yeast display
surface proteins are described in the literature. plasmid and expressed as fusion products with
Staphylococcus aureus continues to present the Aga2p protein. The human antibody
major challenges in clinical settings due to fragments are typically fused to the C-termi-
the tendency of this species to develop antibi- nus of the Aga2p subunit (100). While the
otic resistance, such as methicillin-resistant S. scFvs are the most frequently used format in
aureus (MRSA). Phage library selections yeast display, other antibody fragments may be
against peptide fragments of the GrfA surface displayed on the yeast surface, including Fabs,
macromolecule transporter from MRSA whole IgG1s, and camelid domain antibodies
resulted in a lead candidate scFv (Aurograb), (102). The Aga1p cell wall protein is stably
which eventually failed in clinical trials (87, expressed from a chromosomal locus. The
99). Given the spread of MRSA strains among expression of both Aga1p and the mAb-Aga2p
hospitals, its elevated resistance rate to antibi- product are under the control of the galactose-
otics, and the current failure of available inducible GAL1 promoter (102, 103). A diagram
human antibodies, the continued development detailing the yeast surface display system is
of new mAb selection strategies will create shown in Fig. 3.
improved therapeutic alternatives during pa- In the presence of galactose, the yeast cell
tient infections. displays the antibody fragment fused to the
C-terminus of the Aga2p subunit. Variations
in the surface density of this combinatorial
complex can be monitored and normalized
DISCOVERY OF THERAPEUTIC
using immunofluorescent labeling of either the
ANTIBODIES USING YEAST DISPLAY
HA or c-Myc tags bordering the scFv or Fab.
TECHNOLOGY
Since protein expression is induced following
cell growth, the GAL1 promoter protects
Yeast Biology and Antibody
potentially toxic antibody fragments from
Display System
negative selection. Following the subcloning
Yeast are eukaryotic, unicellular organisms of the antibody gene repertoire into the yeast
comprising a variety of strains belonging to the display vector, the typical established library
kingdom Fungi. The budding yeast Saccharo- consists of 107 to 109 unique variants (100, 103).
myces cerevisiae possesses a rigid cell wall Although the early studies with yeast
approximately 200 nm thick, primarily com- display predominantly involved affinity matu-
posed of mannoproteins and b-linked glucans ration of existing antibody sequences, recent
(100). The budding yeast cell wall surface discovery campaigns show the successful
agglutinins function as adhesion proteins, isolation of novel, de novo immunoglobulins.
promoting aggregation of opposite mating Both magnetic assisted cell sorting (MACS)
types during mating events (101). The Aga2p and fluorescence activated cell sorting (FACS)
subunit, which binds the agglutinin proteins procedures are employed during isolation of
of the opposite mating type, is tethered to the antibody clones from yeast libraries (103). Due
116 SHEEHAN AND MARASCO

FIGURE 3 Overview of yeast antibody display. The scFv (or Fab) is displayed as a fusion product with the
Saccharomyces Aga2p protein (light blue). This fusion product can be detected and normalized by
uorescent signaling through the HA tags (orange) and c-Myc tags (dark blue). During FACS selections,
the Ag-reactive library variants are detected through the uorescent avidin tag (pink) on the
biotinylated target antigen (red). doi:10.1128/microbiolspec.AID-0028-2014.f3

to the flow cytometry rate limitations of 107 to tion of scFv or Fab expression (using the c-
108 cells per hour, the MACS steps are used to Myc tag) and antigen binding (using the biotin
remove nonbinding mAb fragments to reduce label on this target). During this first round of
the cell input number for subsequent FACS FACS selection, the top 5% of this double-
selection. These MACS separations mimic the positive population is typically collected to
panning process from phage display to remove protect diversity, with the selection gate
nonbinding yeast cells. Even less successful narrowed to the top 0.1 to 1% in subsequent
MACS selections can reduce the nonreactive rounds of flow cytometry screening (103).
background by approximately 100 times, per- Following three to five rounds of these FACS
mitting reasonable yeast cell numbers for selections, the plasmid DNA is isolated from
FACS screening (102). this heterogeneous, sorted population and
Following this MACS-based depletion, the used to transform competent E. coli cells.
antigen-reactive population is screened by Plasmid DNA is isolated from a selection of
FACS to enrich for candidate clones. The these E. coli transformants for antibody se-
antigen concentration during yeast library quence analysis.
screening is kept at 10-fold excess above the
desired dissociation constant (KD) of individ-
Yeast Display versus Phage
ual clones to allow the majority of the
Display Technologies
displayed mAb fragments to be engaged at
equilibrium. Further, this antigen excess Both advantages and drawbacks exist when
prevents the titration of target by antibody comparing yeast and phage antibody library
binding (102). These MACS-selected yeast technologies, although their gene repertoires
cells are labeled for the simultaneous detec- may arise from the same sources (see Sources
CHAPTER 6 Phage and Yeast Display 117

of Antibody Genes for Display Libraries, Another disadvantage of the yeast display
above). The most immediate advantage of system involves the number of antibody copies
yeast display is the accurate control over displayed per cell. Since each yeast cell
selection parameters during FACS screening. displays 1 104 to 1 105 scFv or Fab copies,
The collected population percentage, signal the selection events are based upon antibody
normalization, and desired binding affinities avidity, rather than affinity. This property is
can be fixed by the flow cytometry boundaries. emphasized during selections against oligo-
This ability to define binding criteria during meric antigens, where the multicopy yeast
the selection process represents an advantage display profile allows for multivalent binding
over phage display platforms, where variant (102). This antibody density on the yeast cell
discrimination is dependent on washing steps surface may result in the isolation of variants
rather than real-time kinetic observations with lower affinity than those discovered from
(102). In other words, this systematic bias monovalent phage display methods.
toward desired binding properties available A modern selection strategy makes use of
during yeast library selections is not available both phage and yeast selections. In this study,
during phage library campaigns. two rounds of phage selections against Myco-
Other advantages surrounding yeast dis- bacterium tuberculosis Ag85 were performed.
play technologies are rooted in the quality of Following this nave phage library selection,
antibody fragment displayed. Yeast libraries this antigen-enriched population was sub-
make use of the eukaryotic, posttranslational cloned into a yeast display system for mini-
modifications to display scFvs or Fabs in a library construction, which yielded over 100
similar fashion as mammalian cells. Yeast unique mAbs (105). This combined selection
cells employ similar glycosylation patterns as approach takes advantage of the strengths of
mammalian systems, improving antibody both technologies: the large library size and
solubility as well as removing a future un- affinity-based selections of phage display and
known attribute during full-length IgG ex- the controlled, defined collection of variants
pression in mammalian systems. In addition, from yeast display. This powerful, two-system
proper folding events within the yeast endo- strategy should be considered in future dis-
plasmic reticulum, in the presence of chap- covery efforts, given the larger panel of valu-
erone proteins, protect library diversity by able mAbs isolated through the strengths of
permitting expression of clones too complex both technologies.
for prokaryotic processing used by E. coli cells
during phage display (102).
Antibodies for Infectious Diseases
While this eukaryotic translation machin-
Discovered by Yeast Display
ery offers expression advantages over phage
display libraries, certain drawbacks exist in Although the previously discussed aspects
yeast display approaches. The total number of of yeast display highlight the capacity of this
unique antibody clones in yeast libraries is combinatorial technology as an effective se-
always several orders of magnitude lower than lection system, the use of yeast libraries for
phage libraries, due to the lower transforma- novel antibody discovery is relatively new
tion efficiency observed in yeast cells. Yeast compared with the history of phage display
antibody libraries possess a theoretical limit of libraries. Since the initial application of yeast
1 107 to 1 109 total clones, with phage display display technologies focused on epitope map-
libraries capable of representing 1 106 to 1 ping and affinity maturation, the current
1011 variants (103). However, recent advances employment of yeast surface display in the
in transformation optimization allow for the field of antibody discovery for infectious dis-
routine generation of yeast antibody display ease remains biased toward these structural
libraries with 1 1010 total unique clones (104). and functional characterization studies (102).
118 SHEEHAN AND MARASCO

Compared to phage display approaches, the most common viral targets for yeast display
published list of antiviral and antibacterial studies include HIV-1 and flaviviruses. As this
mAbs isolated from yeast libraries is limited. yeast display platform continues to develop,
additional viral pathogens should be targeted
Antiviral mAb Discovery Involving using yeast antibody libraries. A brief sum-
Yeast Display mary of antiviral mAb discovery efforts using
Ongoing research surrounding the discovery yeast antibody libraries is listed in Table 3.
of novel antiviral mAbs is supported by the
yeast display technology. Unlike phage dis- Anti-HIV-1 mAbs Discovered by Yeast
play, epitope mapping serves as the most Display
popular application of yeast libraries in the Separate selection campaigns against recombi-
development of antiviral mAbs. Typically, nant HIV-1 gp120 using yeast and phage display
mAbs isolated from phage libraries or murine systems provided a comparative study exam-
hybridoma campaigns are mapped to specific ining the collection of selected antibodies.
epitopes on the target antigen by yeast display These libraries were created from the same
techniques. However, certain reports describe immune donor pool and screened using the
the use of yeast library selections for the same antigen, limiting the investigation to
discovery of novel antiviral mAbs. Despite this display system differences (106). These yeast
efficacy, fewer antiviral mAb discovery efforts library selections produced at least twice as
using yeast display have been published. The many unique antibody variants as the phage

TABLE 3 Summary of antiviral antibodies discovered using yeast displaya


Ab fragment or Ab Library
Viral family Virus Viral target Ag displayed source application Reference

Flaviviridae Dengue virus DENV-1 E DENV-1 E N/A Epitope 108


(DENV) glycoprotein DIII mutants mapping
Dengue virus DENV-2 E DENV-2 E N/A Epitope 109
glycoprotein DI-DIII mutants mapping
Dengue virus DENV-3 E DENV-3 E N/A Epitope 110
glycoprotein wild-type and mapping
mutant DIII
fragments
Dengue virus DENV-4 E DENV-4 E N/A Epitope 111
glycoprotein ectodomain and mapping
DI-DIII fragments
Dengue virus DENV DIII Fab Nave mAb 112
fragments selections
West Nile virus E glycoprotein WNV E N/A Epitope 72, 107
ectodomain and mapping
mutant DIII
fragments
Orthomyxoviridae Inuenza A virus HA Wild-type HA0, N/A Epitope 113
HA1, and HA2 mapping
subunits; HA1
subunit mutants
Inuenza A virus HA Wild-type HA N/A Epitope 114
fragments and mapping
HA1 subunit
fragments
Retroviridae HIV-1 gp120 scFv Immune mAb 106
selections
a
Note: This list is a summary of selected publications and is not to be considered complete.
CHAPTER 6 Phage and Yeast Display 119

selections, including all the mAbs identified display libraries (112). These novel, competi-
from the phage library. The authors attributed tive yeast library selections were based upon
this difference in antibody recovery to the cross-reactive, neutralizing epitope deter-
eukaryotic expression of the scFv fragment as minants provided from yeast mapping studies
the primary factor between yeast and phage (109). Competitive FACS sorting in the pre-
display. This increased sensitivity in selecting sence of a mutant DIII fragment expressed as
against gp120 supports the demand for future an Fc fusion protein produced several wild-
yeast library technology in antiviral antibody type DIII-Fc-reactive populations. The obser-
isolation. vation of these wild-type antigen-binding
subpopulations suggests the mutant protein
Anti-Flavivirus mAb Discovery competition influenced yeast library selec-
Involving Yeast Display tions. This unique strategy of competing
Among the flaviviruses, WNV is targeted for antigens during selections offers a novel
protective antibody discovery. Antibody-me- method for isolating epitope-defined variants
diated neutralization of WNV involves recog- from yeast antibody display libraries.
nition of the E glycoprotein. This WNV surface
E protein is characterized by three separate Anti-Influenza mAb Discovery Involving
structural domains. Yeast display experiments Yeast Display
using hybridoma-derived mAbs provided the Previous fine epitope mapping studies for
molecular basis for their neutralization activ- anti-influenza mAbs describe the use of yeast
ity (107). The majority of the mAbs character- display libraries. Broad domain mapping for
ized in this study localized to domain III (DIII) independent panels of anti-influenza mAbs are
of the viral E antigen. In addition, yeast surface described from yeast libraries displaying full-
display mapped a panel of E-selected neutral- length, precursor influenza HA (HA0) or the
izing mAbs from a phage library to specific HA1 and HA2 subunits separately (113, 114).
domains of the viral glycoprotein (72). These After confirming binding and domain locali-
phage library-derived scFvs selectively bound zation, sublibraries of mutant HA fragments
DI/DII, with no interaction with DIII. These were generated by error-prone PCR for fine
yeast-library mapped variations in E glycopro- epitope mapping. Loss of binding to particular
tein epitope localization may be rooted in the mutant HA subunits indicated particular res-
different antibody selection methods. These idues on the viral envelope protein required
yeast display-detailed protective epitopes are for mAb binding. These investigations provid-
projected to influence ongoing vaccine strate- ed structural neutralization determinants for
gies against WNV. separate mAb panels against surface HA from
Similar to WNV, the four serotypes of high-pathogenic influenza strains.
dengue virus (DENV1-4) possess a surface E
glycoprotein composed of three distinct do- Antibacterial mAb Discovery Involving
mains. Yeast display strategies using a series of Yeast display
mutant DIII fragments from the DENV-1 E The advantages surrounding yeast surface
protein revealed the single or combinations of display for antibacterial mAb discovery offer
residues for the binding of seven mAbs isolated the same strengths as phage libraries, namely
from murine hybridomas (108). Likewise, the elimination of in vivo obstacles imposed
panels of neutralizing mAbs against DENV-2, by hybridoma campaigns and the rapid, high
DENV-3, and DENV-4 were finely mapped throughput selection of whole antibody librar-
across all three domains of the E antigen using ies. Similar to the discovery work with phage
yeast libraries (109111). libraries, the primary success with yeast
Recent selections isolated human anti- library selections involves soluble, secreted
bodies against dengue E protein from yeast exotoxins, rather than bacterial cell wall or
120 SHEEHAN AND MARASCO

membrane-associated antigens. In keeping two previously identified scFvs were sorted on


with the trends discussed concerning antiviral decreasing concentrations of BoNT/B, E, or F
mAb discovery, the majority of published yeast and produced improved mAbs with broadened
library studies describe epitope mapping or high affinity binding to additional subtypes
affinity maturation. While recent studies de- beyond the parent clone (116). Similarly, VL
scribe success with novel antibacterial mAb shuffling through haploid yeast mating meth-
isolation from yeast libraries, the shorter ods generated affinity-matured Fabs from the
history of these technologies results in a shor- parent scFvs isolated from an immune yeast
ter catalog of published discovery data. A library (117).
brief summary of bacterial mAb discovery
efforts using yeast antibody libraries is listed Antianthrax mAb Discovery Involving
in Table 4. Yeast Display
Fine epitope mapping studies using yeast
Antibotulinum mAb Discovery Involving surface display provided the structural re-
Yeast Display quirements for mAbs neutralizing B. anthracis
Recent selection efforts using yeast display toxin components. The screening of a yeast
libraries report the successful discovery of display library composed of mutant edema
novel mAbs binding to C. botulinum neurotox- factor DIII fragments identified critical resi-
in subtype A (BoNT/A). This campaign yielded dues for neutralization of an anthrax toxin
three lead candidate scFv clones demonstrat- (118). These yeast display efforts revealed that
ing high-affinity binding to full-length BoNT/ the heavy chain CDR3 of an EF-neutralizing
A (115). To identify novel epitopes, yeast clones mAb competes for an epitope shared with
were screened against BoNT/A after labeling calmodulin, which serves as an enzymatic
with previously characterized mAbs recogniz- activator of B. anthracis edema factor. In the
ing the neutrotoxin heavy chain to mask this same fashion, a yeast library displaying mu-
domain during library selection. tants of the N-terminal region of the protective
In addition to novel mAb discovery, yeast antigen (PA20) defined the contact residues of
surface display approaches are used for affin- a known, neutralizing mAb (119). These map-
ity maturation of known anti-BoNT anti- ping data indicated that the published mAb
bodies. Yeast display VL shuffled libraries of prevents the furin-mediated activation of PA,

TABLE 4 Summary of antibacterial antibodies discovered using yeast displaya


Ab fragment or Ab Library application Reference
Bacterial family Bacterial species Bacterial target Ag displayed source

Bacillaceae Bacillus Edema factor Mutant EF N/A Epitope mapping 118


anthracis DIII fragments
Bacillus Protective Mutant N/A Epitope mapping 119
anthracis antigen (PA) N-terminal PA
fragments
(PA20)
Clostridiaceae Clostridium Botulinum scFv Nave mAb selections 115
botulinum neurotoxin
(BoNT)
subtype A
Clostridium BoNT subtypes Fab Immune Afnity maturation 116
botulinum B, E, and F
Clostridium BoNT subtypes scFv and Fab Immune mAb selections 117
botulinum A and B and afnity
maturation
a
Note: This list is a summary of selected publications and is not to be considered complete.
CHAPTER 6 Phage and Yeast Display 121

despite the contact residues being distant from REFERENCES


the furin recognition sequence. 1. Rakonjac J, Bennett NJ, Spagnuolo J, Gagic
D, Russel M. 2011. Filamentous bacteriophage:
biology, phage display and nanotechnology
CONCLUSIONS applications. Curr Issues Mol Biol 13:5176.
2. Boeke JD, Model P. 1982. A prokaryotic mem-
brane anchor sequence: carboxyl terminus of
This article details the development and use of
bacteriophage f1 gene III protein retains it in
the phage and yeast display technologies for the membrane. Proc Natl Acad Sci USA 79:
the discovery of novel mAbs for infectious 52005204.
diseases. Through the application of these 3. Rakonjac J, Feng J, Model P. 1999. Filamen-
combinatorial methods, the successful isola- tous phage are released from the bacterial
membrane by a two-step mechanism involving
tion of unique antibodies as treatment options
a short C-terminal fragment of pIII. J Mol Biol
and vaccine development tools continues to be 289:12531265.
reported. The discovery of pathogen-specific 4. Loeb T. 1960. Isolation of a bacteriophage for
mAbs alleviates the clinical challenges posed the F plus and Hfr mating types of Escherichia
by the enduring development of microbial coli K-12. Science 131:932933.
resistance, which is provided through escape 5. Barbas CF III, Kang AS, Lerner RA, Benkovic
SJ. 1991. Assembly of combinatorial antibody
mutations and evolution of resistant strains.
libraries on phage surfaces: the gene III site.
These isolated mAbs offer nontoxic, target- Proc Natl Acad Sci USA 88:79787982.
specific treatment options and vaccine strategy 6. Clackson T, Hoogenboom HR, Griffiths AD,
determinants for viral and bacterial pathogens. Winter G. 1991. Making antibody fragments
The selection of high-affinity (within and using phage display libraries. Nature 352:624
628.
below the nanomolar range) antibodies across
7. Marks JD, Hoogenboom HR, Bonnert TP,
formats (scFv and Fab) and degrees of valency McCafferty J, Griffiths AD, Winter G. 1991.
(mono- and multivalent) describes the flexi- By-passing immunization. Human antibodies
bility of the phage and yeast display ap- from V-gene libraries displayed on phage. J
proaches. The continued improvement of Mol Biol 222:581597.
phage library quality has maintained the 8. Marks JD, Tristem M, Karpas A, Winter G.
1991. Oligonucleotide primer for polymerase
relevance of this discovery platform for
chain reaction amplification of human immu-
todays emerging infectious diseases. In addi- noglobulin variable genes and design of family-
tion, the ongoing success of antibody specific oligonucleotide probes. Eur J Immunol
selections using yeast display libraries drives 21:985991.
the expansion of this approach beyond its early 9. de Haard HJ, van Neer N, Reurs A, Hufton SE,
Roovers RC, Henderikx P, de Brune AP,
epitope mapping and affinity maturation
Arends JW, Hoogenboom HR. 1999. A large
applications. These microbial surface display non-immunized human Fab phage library that
technologies offer powerful platforms for permits rapid isolation and kinetic analysis of high
antibody drug discovery and vaccine develop- affinity antibodies. J Biol Chem 274:1821818230.
ment within and beyond infectious diseases. 10. Vaughan TJ, Williams AJ, Pritchard K, Osbourn
JK, Pope AR, Earnshaw JC, McCafferty J,
Hodits RA, Wilton J, Johnson KS. 1996.
ACKNOWLEDGMENT Human antibodies with sub-nanomolar affinities
isolated from a large non-immunized phage
Conflicts of interest: We declare no conflicts. display library. Nat Biotechnol 14:309314.
11. Gram H, Marconi LA, Barbas CF 3rd, Collet
TA, Lerner RA, Kang AS. 1992. In vitro
CITATION selection and affinity maturation of antibodies
from a nave combinatorial immunoglobulin
Sheehan J, Marasco WA. 2015. Phage and library. Proc Natl Acad Sci USA 89:35763580.
yeast display. Microbiol Spectrum 3(1):AID- 12. Hoogenboom HR. 2002. Overview of antibody
0028-2014. phage-display technology and its applications,
122 SHEEHAN AND MARASCO

p 138. In OBrien PM, Aitken R (ed), Antibody Plckthun A, Virneks B. 2000. Fully syn-
Phage Display, Methods and Protocols. Methods thetic human combinatorial antibody libraries
in Molecular Biology, Vol. 178. Humana Press, (HuCAL) based on modular consensus frame-
Totowa, NJ. works and CDRs randomized with trinucleo-
13. Griffiths AD, Malmqvist M, Marks JD, Bye tides. J Mol Biol 11:5786.
JM, Embleton MJ, McCafferty J, Baier M, 22. Tiller T, Schuster I, Deppe D, Siegers K,
Holliger KP, Gorick BD, Hughes-Jones NC. Strohner R, Herrmann T, Berenguer M,
1993. Human anti-self antibodies with high Poujol D, Stehle J, Stark Y, Heling M,
specificity from phage display libraries. EMBO Daubert D, Felderer K, Kaden S, Klln J,
J 12:725734. Enzelberger M, Urlinger S. 2013. A fully
14. Burton DR, Barbas CF 3rd, Persson MA, synthetic human Fab antibody library fixed
Koenig S, Chanock RM, Lerner RA. 1991. A on VH/VL framework pairings with favorable
large array of human monoclonal antibodies to biophysical properties. MAbs 5:445470.
type 1 human immunodeficiency virus from 23. Gao C, Mao S, Lo CH, Wirsching P, Lerner
combinatorial libraries of asymptomatic sero- RA, Janda KD. 1999. Making artificial anti-
positive individuals. Proc Natl Acad Sci USA bodies: a format for phage display of combi-
88:1013410137. natorial heterodimeric arrays. Proc Natl Acad
15. de Wildt RM, Finnern R, Ouwehand WH, Sci USA 96:60256030.
Griffiths AD, van Venrooij WJ, Hoet RM. 24. Jespers LS, De Keyser A, Stanssens PE. 1996.
1996. Characterization of human variable do- LambdaZLG6: a phage lambda vector for high-
main antibody fragments against the U1 RNA- efficiency cloning and surface expression of
associated A protein, selected from a synthetic cDNA libraries on filamentous phage. Gene
and a patient-derived combinatorial V gene 173:179181.
library. Eur J Immunol 26:629639. 25. Iannolo G, Minenkova O, Petruzzelli R,
16. Barbas CF 3rd, Burton DR. 1996. Selection Cesareni G. 1995. Modifying filamentous
and evolution of high-affinity human anti-viral phage capsid: limits in the size of the major
antibodies. Trends Biotechnol 14:230234. capsid protein. J Mol Biol 248:835844.
17. Hoogenboom HR, Winter G. 1992. By-passing 26. Zwick MB, Shen J, Scott JK. 2000. Homodimeric
immunization. Human antibodies from syn- peptides displayed by the major coat protein of
thetic repertoires of germline VH segments filamentous phage. J Mol Biol 300:307320.
rearranged in vitro. J Mol Biol 227:381388. 27. OConnell D, Becerril B, Roy-Burman A,
18. Zhu K, Day T. 2013. Ab initio structure pre- Daws M, Marks JD. 2002. Phage versus
diction of the antibody hypervariable H3 loop. phagemid libraries for generation of human
Proteins 81:10811089. monoclonal antibodies. J Mol Biol 312:4956.
19. Hoet RM, Cohen EH, Kent RB, Rookey K, 28. Marks JD, Hoogenboom HR, Griffiths AD,
Schoonbroodt S, Hogan S, Rem L, Frans N, Winter G. 1992. Molecular evolution of pro-
Daukandt M, Pieters H, van Hegelsom R, teins on filamentous phage. Mimicking the
Neer NC, Nastri HG, Rondon IJ, Leeds JA, strategy of the immune system. J Biol Chem
Hufton SE, Huang L, Kashin I, Devlin M, 267:1600716010.
Kuang G, Steukers M, Viswanathan M, Nixon 29. de Wildt RM, Tomlinson IM, Ong JL, Holliger
AE, Sexton DJ, Hoogenboom HR, Ladner RC. P. 2002. Isolation of receptor-ligand pairs by
2005. Generation of high-affinity human anti- capture of long-lived multivalent interaction
bodies by combining donor-derived and syn- complexes. Proc Natl Acad Sci USA 99:85308535.
thetic complementarity-determining-region 30. Rakonjac J, Jovanovic G, Model P. 1997. Fila-
diversity. Nat Biotechnol 23:344348. mentous phage infection-mediated gene expres-
20. Rothe C, Urlinger S, Lhning C, Prassler J, sion: construction and propagation of the gIII
Stark Y, Jger U, Hubner B, Bardroff M, deletion mutant helper phage R408d3. Gene
Pradel I, Boss M, Bittlingmaier R, Bataa T, 198:99103.
Frisch C, Brocks B, Honegger A, Urban M. 31. Chasteen L, Ayriss J, Pavlik P, Bradbury AR.
2008. The human combinatorial antibody 2006. Eliminating helper phage from phage
library HuCAL GOLD combines diversification display. Nucleic Acid Res 34:e145.
of all six CDRs according to the natural 32. Hoogenboom HR, Griffiths AD, Johnson KS,
immune system with a novel display method Chiswell DJ, Hudson P, Winter G. 1991.
for efficient selection of high-affinity anti- Multi-subunit proteins on the surface of fila-
bodies. J Mol Biol 376:11821200. mentous phage: methodologies for displaying
21. Knappik A, Ge L, Honegger A, Pack P, antibody (Fab) heavy and light chains. Nucleic
Fischer M, Wellnhofer G, Hoess A, Wlle J, Acids Res 19:41334137.
CHAPTER 6 Phage and Yeast Display 123

33. Garrard LJ, Yang M, OConnell MP, Kelley 46. Skehal JJ, Wiley DC. 2000. Receptor binding
RF, Henner DJ. 1991. Fab assembly and en- and membrane fusion in virus entry: the
richment in a monovalent phage system. Bio- influenza hemagglutinin. Annu Rev Biochem
technology 9:13731377. 69:531569.
34. Chan CE, Chan AH, Lim AP, Hanson BJ. 2011. 47. Wiley DC, Wilson IA, Skehel JJ. 1981. Struc-
Comparison of the efficiency of antibody tural identification of the antibody-binding
selection from semi-synthetic scFv and non- sites of Hong Kong influenza haemagglutinin
immune Fab phage display libraries against and their involvement in antigenic variation.
protein targets for rapid development of diag- Nature 289:373378.
nostic immunoassays. J Immunol Methods 373: 48. Both GW, Sleigh MJ, Cox NJ, Kendal AP.
7988. 1983. Antigenic drift in influenza virus H3
35. Qi J, Ye X, Ren G, Kan F, Zhang Y, Guo M, hemagglutinin from 1968 to 1980: multiple
Zhang Z, Li D. 2014. Pharmacological efficacy evolutionary pathways and sequential amino
of anti-IL-1b scFv, Fab, and full-length anti- acid changes at key antigenic sites. J Virol 48:
bodies in treatment of rheumatoid arthritis. 5260.
Mol Immunol 57:5965. 49. Sui J, Hwang WC, Perez S, Wei G, Aird D,
36. Steinwand M, Droste P, Frenzel A, Hust M, Chen LM, Santelli E, Stec B, Cadwell G, Ali
Dbel S, Schirrmann T. 2014. The influence of M, Wan H, Murakami A, Yammanuru A, Han
antibody fragment format on phage display T, Cox NJ, Bankston LA, Donis RO,
based affinity maturation of IgG. MAbs 6:204 Liddington RC, Marasco WA. 2009. Structur-
218. al and functional bases for broad-spectrum
37. Lauring AS, Frydman J, Andino R. 2013. The neutralization of avian and human influenza A
role of mutational robustness in RNA virus viruses. Nat Struct Mol Biol 16:265273.
evolution. Nat Rev Microbiol 11:327336. 50. Throsby M, van den Brink E, Jongeneelen M,
38. Balmer O, Tanner M. 2011. Prevalence and Poon LL, Alard P, Cornelissen L, Bakker A,
implications of multiple-strain infections. Lan- Cox F, van Deventer E, Guan Y, Cinatl J, ter
cet Infect Dis 11:868878. Meulen J, Lasters I, Carsetti R, Peiris M, de
39. Leye N, Vidal N, Ndiaye O, Diop-Ndiaye H, Kruif J, Goudsmit J. 2008. Heterosubtypic
Wade AS, Mboup S, Delaporte E, Toure- neutralizing monoclonal antibodies cross-pro-
Kane C, Peeters M. 2013. High frequency of tective against H5N1 and H1N1 recovered from
HIV-1 infections with multiple HIV-1 strains human IgM+ memory B cells. PLoS One 3:
in men having sex with men (MSM) in Senegal. e3942. doi:10.1371/journal.pone.0003942.
Infect Genet Evol 20:206214. 51. Dreyfus C, Laursen NS, Kwaks T, Zuijdgeest
40. Yewdell JW, Spiro DJ, Golding H, Quill H, D, Khayat R, Ekiert DC, Lee JH, Metlagel Z,
Mittelman A, Nabel GJ. 2013. Getting to the Bujny MV, Jongeneelen M, van der Vlugt R,
heart of influenza. Sci Transl Med 5:e191ed8. Lamrani M, Korse HJ, Geelen E, Sahin ,
41. Corti D, Lanzavecchia A. 2013. Broadly neutral- Sieuwerts M, Brakenhoff JP, Vogels R, Li
izing antiviral antibodies. Annu Rev Immunol OT, Poon LL, Peiris M, Koudstaal W, Ward
31:705742. AB, Wilson IA, Goudsmit J, Friesen RH.
42. Marasco WA, Sui J. 2007. The growth and 2012. Highly conserved protective epitopes
potential of human antiviral monoclonal anti- on influenza B viruses. Science 337:13431348.
body therapeutics. Nat Biotechnol 25:1421 52. Avnir Y, Tallarico AS, Zhu Q, Bennett AS,
1434. Connelly G, Sheehan J, Sui J, Fahmy A,
43. Thie H, Meyer T, Schirrmann T, Hust M, Huang CY, Cadwell G, Bankston LA,
Dbel S. 2008. Phage display derived thera- McGuire AT, Stamatatos L, Wagner G,
peutic antibodies. Curr Pharm Biotechnol 9: Liddington RC, Marasco WA. 2014. Molecu-
439446. lar signatures of hemagglutinin stem-directed
44. Jegaskanda S, Job ER, Kramski M, Laurie K, heterosubtypic human neutralizing antibodies
Isitman G, de Rose R, Winnall WR, Stratov I, against influenza A viruses. PLoS Pathog 10:
Brooks AG, Reading PC, Kent SJ. 2013. Cross- e1004103. doi:10.1371/journal.ppat.1004103.
reactive influenza-specific antibody-dependent 53. Ekiert DC, Kashyap AK, Steel J, Rubrum A,
cellular cytotoxicity antibodies in the absence of Bhabha G, Khayat R, Lee JH, Dillon MA,
neutralizing antibodies. J Immunol 190:1837 ONeil RE, Faynboym AM, Horowitz M,
1848. Horowitz L, Ward AB, Palese P, Webby R,
45. Gamblin SJ, Skehel JJ. 2010. Influenza hem- Lerner RA, Bhatt RR, Wilson IA. 2012. Cross-
agglutinin and neuraminidase membrane gly- neutralization of influenza A viruses mediated
coproteins. J Biol Chem 285:3840338409. by a single antibody loop. Nature 489:526532.
124 SHEEHAN AND MARASCO

54. Iba Y, Fujii Y, Ohshima N, Sumida T, Choudhary A, Rybak SM, Fouts T, Montefiori
Kubota-Koketsu R, Ikeda M, Wakiyama M, DC, Broder CC, Quinnan GV Jr, Dimitrov DS.
Shirouzu M, Okada J, Okuno Y, Kurosawa Y, 2007. Cross-reactive HIV-1 neutralizing mono-
Yokoyama S. Conserved neutralizing epitope clonal antibodies selected by screening of an
at globular head of hemagglutinin in H3N2 immune human phage library against an envelope
influenza viruses. J Virol 88:71307144. glycoprotein (gp140) isolated from a patient (R2)
55. Caton AJ, Brownlee GG, Yewdell JW, with broadly HIV-1 neutralizing antibodies.
Gerhard W. 1982. The antigenic structure of Virology 363:7990.
the influenza virus A/PR/8/34 hemagglutinin 64. Yoshikawa M, Mukai Y, Tsunoda S, Tsutsumi Y,
(H1 subtype). Cell 31:417427. Yoshioka Y, Okada N, Nakagawa S. 2011.
56. Bartesaghi A, Merk A, Borgnia MJ, Milne JL, Modifying the antigen-immunization schedule
Subramaniam S. 2013. Prefusion structure of improves the variety of monoclonal antibodies
trimeric HIV-1 envelope glycoprotein deter- obtained from immune-phage antibody libraries
mined by cryo-electron microscopy. Nat Struct against HIV-1 Nef and Vif. J Biosci Bioeng 111:597
Mol Biol 20:13521357. 599.
57. Taylor BS, Sobieszczyk ME, McCutchan FE, 65. Sui J, Li W, Murakami A, Tamin A, Matthews
Hammer SM. The challenge of HIV-1 subtype LJ, Wong SK, Moore MJ, Tallarico AS,
diversity. N Engl J Med 358:15901602. Olurinde M, Choe H, Anderson LJ, Bellini WJ,
58. Quakkelaar ED, Bunnik EM, van Alphen Farzan M, Marasco WA. 2004. Potent neutrali-
FP, Boeser-Nunnink BD, van Nuenen AC, zation of severe acute respiratory syndrome
Schuitemaker H. 2007. Escape of human (SARS) coronavirus by a human mAb to S1 protein
immunodeficiency virus type 1 from broadly that blocks receptor association. Proc Natl Acad
neutralizing antibodies is not associated with a Sci USA 101:25362541.
reduction of viral replicative capacity in vitro. 66. Prabakaran P, Gan J, Feng Y, Zhu Z,
Virology 363:447453. Choudhry V, Xiao X, Ji X, Dimitrov DS.
59. Burton DR, Barbas CF 3rd, Persson MA, Koenig 2006. Structure of severe acute respiratory
S, Chanock RM, Lerner RA. 1991. A large array of syndrome coronavirus receptor-binding domain
human monoclonal antibodies to type 1 human complexed with neutralizing antibody. J Biol
immunodeficiency virus from combinatorial li- Chem 281:1582915836.
braries of asymptomatic seropositive individuals. 67. Ferguson NM, Van Kerkhove MD. 2014.
Proc Natl Acad Sci USA 88:1013410137. Identification of MERS-CoV in dromedary
60. Burton DR, Pyati J, Kodrui R, Sharp SJ, camels. Lancet Infect Dis 14:9394.
Thornton GB, Parren PW, Sawyer LA, 68. Memish ZA, Mishra N, Olival KJ, Fagbo SF,
Hendry RM, Dunlop N, Nara PL, Lamacchia Ka p o o r V , Ep s t e i n J H , A l ha k e e m R ,
M, Garratty E, Stiehm ER, Bryson YJ, Cao Y, Durosinloun A, Al Asmari M, Islam A, Kapoor
Moore JP, Ho DD, Barbas CF 3rd. 1994. A, Briese T, Daszak P, Al Rabeeah AA, Lipkin
Efficient neutralization of primary isolates of WI. 2013. Middle East respiratory syndrome
HIV-1 by a recombinant human monoclonal coronavirus in bats, Saudi Arabia. Emerg Infect
antibody. Science 266:10241027. Dis 19:18191823.
61. Zwick MB, Labrijn AF, Wang M, Spenlehauer 69. Raj VS, Mou H, Smits SL, Dekkers DH, Mller
C, Saphire EO, Binley JM, Moore JP, Stiegler MA, Dijkman R, Muth D, Demmers JA, Zaki A,
G, Katinger H, Burton DR, Parren PW. 2001. Fouchier RA, Thiel V, Drosten C, Rottier PJ,
Broadly neutralizing antibodies targeted to the Osterhaus AD, Bosch BJ, Haagmans BL.
membrane-proximal external region of human Dipeptidyl pepetidase 4 is a functional receptor
immunodeficiency virus type 1 glycoprotein for the emerging human coronavirus-EMC.
gp41. J Virol 75:1089210905. Nature 495:251254.
62. Zhu Z, Qin HR, Chen W, Zhao Q, Shen X, 70. Graham RL, Donaldson EF, Baric RS. A decade
Schutte R, Wang Y, Ofek G, Streaker E, after SARS: strategies for controlling emerging
Prabakaran P, Fouda GG, Liao HX, Owens J, coronaviruses. Nat Rev Microbiol 11:836848.
Louder M, Yang Y, Klaric KA, Moody MA, 71. Gierer S, Bertram S, Kaup F, Wrensch F,
Mascola JR, Scott JK, Kwong PD, Montefiori D, Heurich A, Krmer-Khl A, Welsch K, Winkler
Haynes BF, Tomaras GD, Dimitrov DS. 2011. M, Meyer B, Drosten C, Dittmer U, von Hahn T,
Cross-reactive HIV-1-neutralizing human mono- Simmons G, Hofmann H, Phlmann S. 2013. The
clonal antibodies identified from a patient with spike protein of the emerging coronavirus EMC
2F5-like antibodies. J Virol 85:1140111408. uses a novel coronavirus receptor for entry, can be
63. Choudhry V, Zhang MY, Sidorov IA, Louis JM, activated by TMPRSS2, and is targeted by neu-
Harris I, Dimitrov AS, Bouma P, Cham F, tralizing antibodies. J Virol 87:55025511.
CHAPTER 6 Phage and Yeast Display 125

72. Tang XC, Agnihothram SS, Jiao Y, Stanhope effective therapeutically when introduced
J, Graham RL, Peterson EC, Avnir Y, Tallarico directly into the lungs of RSV-infected mice.
AS, Sheehan J, Zhu Q, Baric RS, Marasco Proc Natl Acad Sci USA 91:13861390.
WA. 2014. Identification of human neutraliz- 81. Burioni R, Williamson RA, Sanna PP, Bloom
ing antibodies against MERS-CoV and their FE, Burton DR. 1994. Recombinant human
role in virus adaptive evolution. Proc Natl Acad Fab to glycoprotein D neutralizes infectivity
Sci USA 111:E2018E2026. and prevents cell-to-cell transmission of herpes
73. Ying T, Du L, Ju TW, Prabakaran P, Lau CC, Lu simplex viruses 1 and 2 in vitro. Proc Natl Acad
L, Liu Q, Wang L, Feng Y, Wang Y, Zheng BJ, Sci USA 91:355359.
Yuen KY, Jiang S, Dimitrov DS. 2014. Excep- 82. Christensen DJ, Gottlin EB, Benson RE,
tionally potent neutralization of Middle East Hamilton PT. 2001. Phage display for target-
respiratory syndrome coronavirus by human based antibacterial drug discovery. Drug Discov
monoclonal antibodies. J Virol 88:77967805. Today 6:721727.
74. Gould LH, Sui J, Foellmer H, Oliphant T, 83. Lee HS, Loh YX, Lee JJ, Liu CS, Chu C. 2014.
Wang T, Ledizet M, Murakami A, Noonan K, Antimicrobial consumption and resistance in
Lambeth C, Kar K, Anderson JF, de Silva five Gram-negative bacterial species in a hos-
AM, Diamond MS, Koski RA, Marasco WA, pital from 2003 to 2011. J Microbiol Immunol
Fikrig E. 2005. Protective and therapeutic Infect S1684:1182(14)00074-7. doi:10.1016/j.
capacity of human single-chain Fv-Fc fusion jmii.2014.04.009.
proteins against West Nile Virus. J Virol 79: 84. Velayati AA, Masjedi MR, Farnia P, Tabarsi
1460614613. P, Ghanavi J, Ziazarifi AH, Hoffner SE. 2009.
75. Throsby M, Geuijen C, Goudsmit J, Bakker Emergence of new forms of totally drug-
AQ, Korimbocus J, Kramer RA, Clijsters-van resistant tuberculosis bacilli: super extensively
der Horst M, de Jong M, Jongeneelen M, drug-resistant tuberculosis or totally drug-
Thijsse S, Smit R, Visser TJ, Bijl N, Marissen resistant strains in Iran. Chest 136:420425.
WE, Loeb M, Kelvin DJ, Preiser W, ter 85. Ford C, Yusim K, Ioerger T, Feng S, Chase
Meulen J, de Kruif J. 2006. Isolation and M, Greene M, Korber B, Fortune S. 2012.
charactertization of human monoclonal anti- Mycobacterium tuberculosisheterogeneity
bodies from individuals infected with West revealed through whole genome sequencing.
Nile Virus. J Virol 80:69826992. Tuberculosis (Edinb) 92:194201.
76. Cabezas S, Rojas G, Pavon A, Alvarez M, Pupo 86. Gronski P, Seiler FR, Schwick HG. 1991.
M, Guillen G, Guzman MG. 2008. Selection of Discovery of antitoxins and development of
phage-displayed human antibody fragments on antibody preparations for clinical uses from
dengue virus particles captured by a monoclonal 1890 to 1990. Mol Immunol 28:13211332.
antibody: application to the four serotypes. J Virol 87. Oleksiewicz MB, Nagy G, Nagy E. 2012. Anti-
Methods 147:235243. bacterial monoclonal antibodies: back to the
77. Zhao Y, Moreland NJ, Tay MY, Lee CC, future? Arch Biochem Biophys 526:124131.
Swaminathan K, Vasudevan SG. 2014. Iden- 88. Amersdorfer P, Wong C, Smith T, Chen S,
tification and molecular characterization of Deshpande S, Sheridan R, Marks JD. 2002.
human antibody fragments specific for dengue Genetic and immunological comparison of anti-
NS5 protein. Virus Res 179:225230. botulinum type A antibodies from immune and
78. Maruyama T, Rodriguez LL, Jahrling PB, non-immune human phage libraries. Vaccine
Sanchez A, Khan AS, Nichol ST, Peters CJ, 20:16401648.
Parren PW, Burton DR. 1999. Ebola virus can be 89. Rossetto O, Pirazzini M, Bolognese P, Rigoni
effectively neutralized by antibody produced in M, Montecucco C. 2011. An update of the
natural human infection. J Virol 73:60246030. mechanism of action of tetanus and botulinum
79. Barbas CF 3rd, Crowe JE Jr, Cababa D, Jones neurotoxins. Acta Chim Slov 58:702707.
TM, Zebedee SL, Murphy BR, Chanock RM, 90. Amersdorfer P, Wong C, Chen S, Smith T,
Burton DR. 1992. Human monoclonal Fab frag- Deshpande S, Sheridan R, Finnern R, Marks
ments derived from a combinatorial library bind JD. 1997. Molecular charactertization of murine
to respiratory syncytial virus F glycoprotein and humoral immune response to botulinum neuro-
neutralize infectivity. Proc Natl Acad Sci USA toxin type A binding domain as assessed by using
89:1016410168. phage antibody libraries. Infect Immun 65:3743
80. Crowe JE Jr, Murphy BR, Chanock RM, 3752.
Williamson RA, Barbas CF 3rd, Burton DR. 91. Marks JD. 2004. Deciphering antibody properties
1994. Recombinant human respiratory syncy- that lead to potent botulinum neurotoxin neutral-
tial virus (RSV) monoclonal antibody Fab is ization. Mov Disord 19(Suppl 8):S101S108.
126 SHEEHAN AND MARASCO

92. Jernigan DB, Raghunathan PL, Bell BP, 101. Lipke PN, Kurjan J. 2000. Sexual agglutina-
Brechner R, Bresnitz EA, Butler JC, Cetron M, tion in budding yeasts: structure, function, and
CohenM,DoyleT,FischerM,GreeneC,Griffith regulation of adhesion glycoproteins. Micro-
KS,GuarnerJ,HadlerJL,HayslettJA,MeyerR, biol Rev 56:180194.
Petersen LR, Phillips M, Pinner R, Popovic T, 102. Boder ET, Raeeszadeh-Sarmazdeh M, Price
Quinn CP, Reefhuis J, Reissman D, Rosenstein JV. 2012. Engineering antibodies by yeast
N,SchuchatA,ShiehWJ,SiegalL,SwerdlowDL, display. Arch Biochem Biophys 526:99106.
Tenover FC, Traeger M, Ward JW, Weisfuse I, 103. Chao G, Lau WL, Hackel BJ, Sazinsky SL,
Wiersma S, Yeskey K, Zaki S, Ashford DA, Lippow SM, Wittrup KD. 2006. Isolating and
Perkins BA, Ostroff S, Hughes J, Fleming D, engineering human antibodies using yeast
KoplanJP,GerberdingJL.2002.Investigationof surface display. Nat Protoc 1:755768.
bioterrorism-related anthrax, United States, 104. Benatuil L, Perez JM, Belk J, Hsieh CM. 2010.
2001: epidemiologic findings. Emerg Infect Dis An improved yeast transformation method for the
8:10191028. generation of very large human antibody libraries.
93. Chen Z, Moayeri M, Purcell R. 2011. Mono- ProteinEngDesSel23:155159.
clonal antibody therapies against anthrax. Toxins 105. Ferrara F, Naranjo LA, Kumar S, Gaiotto T,
(Basel) 3:10041019. Mukundan H, Swanson B, Bradbury AR. 2012.
94. Migone TS, Subramanian GM, Zhong J, Using phage and yeast display to select hundreds of
Healey LM, Corey A, Devalaraja M, Lo L, monoclonal antibodies: application to antigen 85, a
Ullrich S, Zimmerman J, Chen A, Lewis M, tuberculosismarker.PLoSOne7:e49535.doi:10.1371/
Meister G, Gillum K, Sanford D, Mott J, journal.pone.0049535.
Bolmer SD. 2009. Raxibacumab for the treat- 106. Bowley DR, Labrijn AF, Zwick MB, Burton DR.
ment of inhalational anthrax. N Engl J Med 361: 2007. Antigen selection from an HIV-1 immune
135144. antibody library displayed on yeast yields many
95. Pelat T, Hust M, Laffly E, Condemine F, novel antibodies compared to selection from the
Bottex C, Vidal D, Lefranc MP, Dbel S, same library displayed on phage. Protein Eng Des
Thullier P. 2007. High-affinity, human anti- Sel 20:8190.
body-like antibody fragment (single-chain var- 107. Oliphant T, Engle M, Nybakken GE, Doane C,
iable fragment) neutralizing the lethal factor Johnson S, Huang L, Gorlatov S, Mehlhop E,
(LF) of Bacillus anthracis by inhibiting protec- Marri A, Chung KM, Ebel GD, Kramer LD,
tive antigen-LF complex formation. Anti- Fremont DH, Diamond MS. 2005. Development
microb Agents Chemother 51:27582764. of a humanized monoclonal antibody with ther-
96. Chen Z, Moayeri M, Zhao H, Crown D, apeutic potential against West Nile virus. Nat Med
Leppla SH, Purcell RH. 2009. Potent neutral- 11:522530.
ization of anthrax edema toxin by a humanized 108. Shrestha B, Brien JD, Sukupolvi-Petty S, Austin
monoclonal antibody that competes with cal- SK, Edeling MA, Kim T, OBrien KM, Nelson CA,
modulin for edema factor binding. Proc Natl Johnson S, Fremont DH, Diamond MS. 2010.
Acad Sci USA 106:1348713492. The development of therapeutic antibodies that
97. Deng XK, Nesbit LA, Morrow KJ Jr. 2003. neutralize homologous and heterologous geno-
Recombinant single-chain variable fragment anti- types of dengue virus type 1. PLoS Pathog 6:
bodies directed against Clostridium difficile toxin B e1000823. doi:10.1371/journal.ppat.1000823.
produced by use of an optimized phage display 109. Sukupolvi-Petty S, Austin SK, Engle M, Brien
system. Clin Diagn Lab Immunol 10:587595. JD, Dowd KA, Williams KL, Johnson S, Rico-
98. Neelakantam B, Sridevi NV, Shukra AM, Hesse R, Harris E, Pierson TC, Fremont DH,
Sugumar P, Samuel S, Rajendra L. 2014. Diamond MS. 2010. Structure and function
Recombinant human antibody fragment against analysis of therapeutic monoclonal antibodies
tetanus toxoid produced by phage display. Eur J againstdenguevirustype2.JVirol84:92279239.
Microbiol Immunol (Bp) 4:4555. 110. Brien JD, Austin SK, Sukupolvi-Petty S, OBrien
99. Burnie JP, Matthews RC, Carter T, Beaulieu E, KM, Johnson S, Fremont DH, Diamond MS.
Donohoe M, Chapman C, Williamson P, 2010. Genotype-specific neutralization and protec-
Hodgetts SJ. 2000. Identification of an immuno- tion by antibodies against dengue virus type 3. J
dominant ABC transporter in methicillin-resis- Virol84:1063010643.
tant Staphylococcus aureus infections. Infect 111. Sukupolvi-Petty S, Brien JD, Austin SK,
Immun 68:32003209. Shrestha B, Swayne S, Kahle K, Doranz BJ,
100. Gera N, Hussain M, Rao BM. 2013. Protein Johnson S, Pierson TC, Fremont DH, Diamond
selection using yeast surface display. Methods MS. 2013. Functional analysis of antibodies
60:1526. against dengue virus type 4 reveals strain-
CHAPTER 6 Phage and Yeast Display 127

dependent epitope exposure that impacts neutral- 116. Garcia-Rodriguez C, Geren IN, Lou J,
ization and protection. J Virol 87:88268842. Conrad F, Forsyth C, Wen W, Chakraborti
112. Puri V, Streaker E, Prabakaran P, Zhu Z, S, Zao H, Manzanarez G, Smith TJ, Brown J,
Dimitrov DS. 2013. Highly efficient selection of Tepp WH, Liu N, Wijesuriya S, Tomic MT,
epitope specific antibody through competitive Johnson EA, Smith LA, Marks JD. 2011.
yeast display library sorting. MAbs 5:533539. Neutralizing human monoclonal antibodies
113. Han T, Sui J, Bennett AS, Liddington RC, Donis binding multiple serotypes of botulinum neutro-
RO, Zhu Q, Marasco WA. 2011. Fine epitope toxin. Protein Eng Des Sel 24:321331.
mapping of monoclonal antibodies against hem- 117. Lou J, Geren I, Garcia-Rodriguez C, Forsyth
agglutinin of a highly pathogenic H5N1 influenza CM, Wen W, Knopp K, Brown J, Smith T,
virus using yeast surface display. Biochem Biophys Smith LA, Marks JD. 2010. Affinity maturation of
Res Commun 409:253259. human botulinum neurotoxin antibodies by light
114. Hu H, Voss J, Zhang G, Buchy P, Zuo T, Wang chain shuffling via yeast mating. Protein Eng Des
L, Wang F, Zhou F, Wang G, Tsai C, Calder L, Sel 23:311319.
Gamblin SJ, Zhang L, Deubel V, Zhou B, 118. Makiya M, Dolan M, Agulto L, Purcell R,
Skehel JJ, Zhou P. 2012. A human antibody Chen Z. 2012. Structural basis of anthrax
recognizing a conserved epitope of H5 hemag- edema factor neutralization by a neutralizing
glutinin broadly neutralizes highly pathogenic antibody. Biochem Biophys Res Commun 417:
avian influenza. J Virol 86:29782989. 324329.
115. Gray SA, Barr JR, Kalb SR, Marks JD, Baird 119. Reason D, Liberato J, Sun J, Camacho J,
CL, Cangelosi GA, Miller KD, Feldhaus MJ. Zhou J. 2011. Mechanism of lethal toxin
2011. Synergistic capture of Clostridium botu- neutralization by a human monoclonal anti-
linum type A neurotoxin by scFv antibodies to body specific for the PA(20) region of Bacillus
novel epitopes. Biotechnol Bioeng 108:2456 anthracis protective antigen. Toxins (Basel)
2467. 3:979990.
Efcient Methods To Isolate Human
Monoclonal Antibodies from Memory B
Cells and Plasma Cells

DAVIDE CORTI1,2 and ANTONIO LANZAVECCHIA2


7
MANY WAYS TO MAKE HUMAN MONOCLONAL ANTIBODIES

Today, several methods are available to isolate human monoclonal anti-


bodies. The first efficient method described was the panning of phage
display libraries constructed from the Ig variable genes of immunized or
infected individuals (1) or from random synthetic libraries (2). While this
method has led to the isolation of several neutralizing antibodies against
multiple pathogens, the resulting antibodies do not represent necessarily
the natural antibody repertoire, since the antibody fragments are generated
from the random pairing of immunoglobulin VH and VL variable regions.
Thus, in the case of phage libraries, it is unlikely that a given VH/VL pair
went through a selection process, including the negative selection for self-
reactivity. Another significant drawback is that target antigens must be
known a priori, since the selection is based on binding to a purified antigen,
rather than for instance neutralization. Consequently, this system is not
suitable to identify new neutralizing targets within complex pathogens. In
addition, selection for high-affinity binding does not necessarily translate
into higher protection if the epitope recognized is not readily available on
the viral spikes. An additional problem of this approach that was frequently

1
Humabs BioMed SA, 6500 Bellinzona, Switzerland; 2Institute for Research in Biomedicine, 6500 Bellinzona,
Switzerland.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0018-2014

129
130 CORTI AND LANZAVECCHIA

encountered is that the antibodies isolated collected after antigenic boost. A significant
in Escherichia coli or yeasts may be expressed limitation of this approach is that the cloning
suboptimally in mammalian cells. and expression of individual antibodies is
Mice that carry human immunoglobulin very labor intensive, and, consequently, the
loci produce fully human antibodies in re- throughput has been generally limited to a few
sponse to immunization with the significant hundred clones. However, recent technologi-
advantage that, being mice, they can be cal advances using nanofluidic devices have
immunized and make antibodies to human considerably increased the throughput of this
antigens (3). This system has become the approach (7).
method of choice for the isolation of anti- A simple and effective alternative to the
bodies specific for human antigens such as single-cell RT-PCR and expression method
cytokines or cell surface molecule. However, it is the direct screening of the antibodies
is less suitable to make antibodies against produced by plasma cells or by activated or
human pathogens such as HIV, hepatitis C immortalized memory B cells. This can be
virus (HCV), hepatitis B virus (HBV), cyto- achieved by using methods based on cell
megalovirus (CMV), that do not infect mice. In cultures, such as those described in this
addition, the immune response of mice is often article, that allow instead the interrogation
suboptimal, possibly owing to the mismatch of the cell-derived monoclonal antibodies in
between human Ig and mouse Fc receptors. their natural secreted form (Fig. 1). In 2004,
Thus, the isolation of antibodies from immune we reported an efficient method of memory
donors offers the advantage of fully exploiting B-cell immortalization using EBV and CpG
the strength of the human antibody response (8), and, in 2011, we demonstrated the
to a human pathogen. feasibility of culturing single plasma cells
An important advance in this field was and identifying those producing antibodies
represented by the use of single-cell reverse with special properties (9). These methods
transcription (RT)-PCR to isolate the immu- will be described in detail below. In 2008,
noglobulin heavy and light chain variable gene Spits and coworkers developed an alterna-
pairs from single B cells, followed by expres- tive method to immortalize human memory
sion of the full antibody by transfection of H B cells by using retroviruses encoding the
and L chain genes in 293 cells (4). The single- antiapoptotic factors Bcl-6 and Bcl-xL (10).
cell RT-PCR and expression approach has Memory B cells are transduced under poly-
been also used to isolate antibodies from clonal conditions (typically 100 cells per
plasma cells isolated soon after a booster well) in the presence of IL-21, and CD40L-
vaccination (5), and from memory B cells, expressing cells differentiate into long-lived
which can be isolated according to their antibody-secreting cells that still expressed
capacity to bind fluorescently labeled antigens their B-cell receptor on the cell surface.
(6). The latter approach has been particularly These cells also maintained a low-level
successful in the isolation of broadly neutral- activation-induced cytidine deaminase-me-
izing antibodies against the CD4 binding site diated mutation activity that represents a
of gp120. However, staining and sorting B cells possible source of antibody clone instabil-
with labeled antigens is not generally feasible ity, but which can also be used to increase
whenever the target antigen is not available or antibody affinity through an in vitro itera-
unknown. In conclusion, the single-cell RT- tive selection process. In 2009, Walker et al.
PCR and expression approach has been very described the isolation of an HIV-1 broadly
useful, especially when antigen binding can be neutralizing antibody able to recognize the
used to enrich for specific cells or when Env protein only in the trimeric native form
specific cells are highly represented in the by using another approach that relied on
population, as is the case for plasma cells the in vitro activation and expansion of
CHAPTER 7 Efcient Methods To Isolate Human mAbs 131

FIGURE 1 Interrogation of human memory B cells and plasma cell repertoires using high-throughput
cellular screens. Shown is a timeline of the process that goes from the selection of donors to the
isolation, characterization, and development of the antibodies. doi:10.1128/microbiolspec.AID-0018-
2014.f1

human memory B cells for approximately ml, respectively. The low antibody levels might
10 days, followed by the screening of culture limit the testing and identification of anti-
supernatant and lysis of all expanded cells bodies in several types of functional assays that
(11). The details of this method have not could require higher levels of antibody.
been disclosed, but Connors and colleagues
have recently described and published in
detail a similar approach (12). The antibody- EFFICIENT IMMORTALIZATION
containing supernatants are used in func- OF MEMORY B CELLS BY USING
tional sensitive assays to identify the wells from EBV AND CPG
which the antibody genes can be retrieved for
cloning and expression in 293 cells. A signifi- Memory B cells are readily accessible in
cant drawback of the latter two methods is peripheral blood and persist for the lifetime of
related to the low levels of antibodies produced an individual and are therefore an excellent
in the culture supernatants. In the Bcl-6/Bcl- source to isolate monoclonal antibodies. Work
xL transduction and the polyclonal stimulation in the late 1970s showed that human memory
technologies, the average productivities are B cells could be immortalized with EBV and
claimed to be 0.1 to 1 g/ml and 0.01 to 0.1 g/ that this method could be used to isolate
132 CORTI AND LANZAVECCHIA

human monoclonal antibodies (13). However, efficiency immortalization of 20 to 30%, we


the efficiency of B-cell immortalization was typically seed 3 cells/well in order to have
low, typically in the order of 0.1%, and clonal growth. This procedure allows the
therefore did not allow an efficient interro- avoidance or minimization of the need to
gation of the human memory repertoire. In subclone positive cultures and the easy rescue
2004, we reported that the efficiency of EBV of the few clones that may show poor growth
immortalization could be dramatically in- in vitro. Using this method and taking advan-
creased by the addition of a toll-like receptor tage of an automated liquid-handling system
(TLR) agonist, in particular, CpG or R848 that and fully integrated immunoassays, we can
trigger TLR9 and TLR7, which are expressed typically screen repertoires of >104 IgG mem-
at high levels on human B cells (8, 14). ory B cells. An even higher number of cells can
The basis for the powerful synergy be screened by plating higher numbers of
between EBV and TLR stimulation was cells per culture (up to 30 to 100). In the latter
clarified in a subsequent study where we case, subcloning of positive cultures will be
examined the requirements for the activa- required in order to isolate a clone making the
tion of human B cells (15). In this study, desired antibody. Importantly, in all cases, the
we reported that maximal stimulation of B antibody is present at high concentrations in
cells requires three distinct signals: signal 1 the culture supernatant so that multiple assays
delivered by B-cell receptor (BCR) stimula- can be performed in parallel, including func-
tion, signal 2 delivered by T-helper cells tional assays such as virus neutralization or
primarily via CD40L/CD40 interaction, and bacterial opsonophagocytosis killing assays.
signal 3 delivered by a TLR agonist (Fig. 2A). An example of a screening by neutralization
In particular, while BCR cross-linking and is provided in Fig. 2D. Of almost 50,000
cognate interaction with activated T cells culture supernatants of immortalized B cells
provided a suboptimal stimulus, addition of from three donors, several were found to
CpG or R848 potently boosted B-cell pro- protect cells from the cytopathic effect of
liferation and differentiation to antibody- metapneumovirus (MPV), and one clone,
producing cells. Interestingly, EBV is known named MPE8, was found in a parallel assay
to activate B cells by expressing two pro- to neutralize respiratory syncytial virus (RSV)
teins, LMP2A and LMP1, that mimic activated as well. Another example (Fig. 2E) shows how
BCR and CD40, respectively, but there is no it is possible to rapidly identify in B cells
evidence that EBV may activate TLR as well obtained from an individual immunized with
(16). Thus, the three-signal model of B-cell the pandemic H1N1 strain so-called hetero-
activation is consistent with the potent syner- subtypic antibodies that bind to H1 and cross-
gy that TLR agonists have on B-cell immor- react with the heterologous H5 hemagglutinin.
talization (Fig. 2B). A significant advantage of the EBV-based
The method has been adapted to high- method is that the immortalized B cells
throughput cultures performed in 384 secrete high amounts of antibodies but also
plates. In a typical experiment, memory B maintain expression of BCR, although at a low
cells are isolated by positive or negative and variable level. Consequently, antigen-
selection according to the isotype expressed specific cells can be selected from a polyclonal
(IgG or IgA), incubated with EBV and CpG, population by panning or staining with
and seeded in multiple 384 plates in the isotype- or light chain-specific antibodies
presence of irradiated allogeneic peripheral or with the specific antigen. In addition, we
blood mononuclear cells (PBMCs) as feeder never found evidence of ongoing somatic
cells. The efficiency of immortalization, as mutations in EBV-immortalized clones. EBV-
demonstrated by the production of high levels immortalized B-cell clones maintain const-
of IgG is close to 30% (Fig. 2C). Given an ant productivity with antibodies typically
CHAPTER 7 Efcient Methods To Isolate Human mAbs 133

recovered in the culture supernatants at con-


centrations ranging from 5 to 50 mg/ml in
static cultures. Thus, antibodies can be easily
prepared from the culture supernatant in
milligram amounts. In the past 10 years, the
EBV+CpG immortalization method has been
used in our laboratory to interrogate memory
B and isolate monoclonal antibodies specific
for a variety of human pathogens, toxins, and
self-antigens (Table 1).

LONG-TERM CULTURE OF SINGLE


PLASMA CELLS AND THEIR
INTERROGATION USING
MULTIPLE ASSAYS

We originally reported that antigen-specific


plasma cells appear in high numbers in
peripheral blood 1 week after booster im-
munization (17) when they may account for
up to 40 to 90% of total Ig-secreting cells
(17). While other laboratories have used this
source of specific plasma cells to clone VH/VL
pairs by RT-PCR and produce the antibodies

1 cell/well in 384 culture plates containing irradi-


ated allogeneic PBMC. Shown is the concentration
of IgG in culture supernatants on day 10. Positive
values above 2.5 OD correspond to IgG concen-
trations >1 g/ml. The efciency was calculated
according to the Poisson distribution. (D) Example
of a primary screening for MPV-neutralizing
antibodies. In preliminary experiments, culture
conditions were dened to achieve cytopathic
effect by using primary MPV isolates. Culture
supernatants were incubated with MPV followed
by addition of LLC-MK2 cells. Living cells were
FIGURE 2 Efcient immortalization of human measured by using a colorimetric assay on day 8.
memory B cells by combination of EBV and CpG Several antibodies were isolated, including one
and isolation of rare neutralizing antibodies. (A) (MPE8) that neutralizes four different para-
BCR stimulation mediated by antigen, T-cell help myxoviruses. Shown is the total numbers of
mediated by CD40L/CD40 interaction, and TLR cultures screened. (E) Blood was collected from
stimulation provide three synergistic stimuli for an immune donor 2 weeks after vaccination with a
activation of human B cells (15). (B) EBV encodes seasonal vaccine, and IgG memory cells were
LMP2a and LMP1 that mimic constitutively acti- immortalized and plated at 3 cells/well in 384-
vated BCR and CD40, thus providing signal 1 and well plates. The supernatants were screened for
signal 2. Addition of TLR agonists potently the presence of antibodies that bind to either H1
synergizes with the viral genes leading to efcient HA (CA09) present in the vaccine or to H5 HA
activation and immortalization (8). (C) Immortal- (VN04) that represents a heterologous group 1
ization efciency of IgG+ memory B cells plated at HA. doi:10.1128/microbiolspec.AID-0018-2014.f2
134 CORTI AND LANZAVECCHIA

TABLE 1 Examples of human monoclonal antibodies isolated by using high-throughput cellular screens
Target Findings Reference
a
SARS-CoV Potent and broadly neutralizing antibodies 8, 22
Diphtheria toxin Substoichiometric neutralization of the toxin Unpublished
Anthrax protective antigen Substoichiometric neutralization of the toxin Unpublished
HIV-1 Neutralizing antibodies targeting different sites including 23, 24
heptad repeat 1
Inuenza A virus Broadly neutralizing and pan-inuenza-A-neutralizing antibodies 9, 25
HCMV Extremely potent neutralizing antibodies targeting the gHgLpUL 19
pentameric complex
RSV/MPV/PVM/BRSVb An antibody that neutralizes 4 paramyxoviruses 20
Plasmodium falciparum Antibodies to VAR2 CSA, MSP2, and p27. Fc-dependent killing 26, 27
of parasites
Norovirus Analysis of the GII.4 norovirus evolution 28
Dengue virus Fc-engineered antibodies neutralizing all 4 serotypes and 29, 30
effective in an ADE mouse model
Rabies virus Potent and broadly neutralizing antibodies covering all 7 Unpublished
lyssavirus genotypes
Staphylcoccus aureus Anti-MSCRAMMs and other target molecule antibodies with Unpublished
therapeutic potential
Avian u (H5N1) Potent and broadly neutralizing antibodies 31
Ebola virus Potent antibody neutralizing all Ebola species Unpublished
Pemphigus Fine specicity, mechanism of action, and role of somatic mutations 32
GM-CSFc Fine specicity, mechanism of action, and role of somatic mutations Unpublished
Citrullinated proteins Specicity and role of somatic mutations Unpublished
HBV Potent neutralizing antibodies against HBsAg covering all genotypes Unpublished
and drug selected mutants
HCV Potent and broadly neutralizing antibodies Unpublished
MERS-CoV d Potent and broadly neutralizing antibody Unpublished
a
SARS, severe acute respiratory syndrome.
b
PVM, pneumonia virus of mice; BRSV, bovine-RSV.
c
GM-CSF, granulocyte-macrophage colony-stimulating factor.
d
MERS, Middle East respiratory syndrome.

recombinantly (5), we were interested to dev- The plating efficiency in IL-6-dependent


elop a method that would allow us to rapidly cultures is high, ranging from 50 to 100%
focus on rare antibodies, for instance, capable (Fig. 3B). In a typical experiment, we isolate
of binding two different viruses. We reasoned CD138+ CD27hi cells by cell sorting and seed
that if single plasma cells could be maintained them in 384 well plates at 1 cell/well in 50 ml
in culture for enough time in a small volume, of complete medium supplemented with IL-6.
the antibody released in the supernatant could After 3 to 4 days, the supernatants are re-
reach concentrations sufficient to perform moved and tested in multiple parallel assays
several parallel assays. We found that single by using an automated liquid-handling system.
plasma cells isolated from blood or bone The throughput and efficiency of the method
marrow could be maintained in culture for is illustrated by the finding that the FI6 pan-
several weeks when cultured on a monolayer influenza-neutralizing antibody was isolated
of immortalized stromal cells. In these condi- every year for 5 consecutive years from the
tions, single plasma cells secreted Ig at a same donor from plasma cells collected after
constant rate of approximately 100 pg/cell/ infection or vaccination (D. Corti, unpublished
day (Fig. 3A). We also found that, in the data). An example of the high throughput of
absence of stromal cells, recombinant IL-6 this method is shown in Fig. 3C and D, where
was sufficient to maintain plasma cell viability 13,000 plasma cell cultures were screened to
at least for 3 to 4 days (9). determine the fraction of hemagglutinin (HA)-
CHAPTER 7 Efcient Methods To Isolate Human mAbs 135

FIGURE 3 Cultures of single plasma cells are instrumental for the rapid identication of rare antibodies.
(A) Survival of single CD138+ plasma cells isolated from peripheral blood (open circles) or bone marrow
(lled circles) in the presence of IL-6 and stromal cell monolayers. Shown is the cumulative production of
IgG in cultures containing single plasma cells. (B) Plasma cells were isolated from peripheral blood as
CD38+ CD138+ and plated at 0.5 cell/well in the presence of IL-6. IgG, IgA, IgM, and IgE levels were
measured in each culture supernatant. The efciency of cloning as estimated from the frequency of Ig-
containing cultures was estimated to be approximately 70%. (C), (D) HA-specic antibodies produced by
plasma cells isolated in 2009 following infection with H1N1 CA09 cross-react extensively with H5-HA
(VN04). (C) In contrast, plasma cells isolated from the same donor in 2010 following vaccination with the
seasonal trivalent vaccine are largely vaccine specic (D). doi:10.1128/microbiolspec.AID-0018-2014.f3

specific antibodies that cross-react with the recombinant antibodies are expressed by using
heterologous H5 HA. This same method was appropriate vectors. This high frequency of
used to clone rare IgE antibodies from an somatic mutations might represent a potential
allergic individual. problem to antibody sequencing, because the
Once identified, the positive cultures are mutated sequences may no longer be comple-
subjected to single-cell RT-PCR and the mentary to the oligos used for VH/VL gene
specific VH/VL pairs are isolated, and the amplification. To overcome this potential
136 CORTI AND LANZAVECCHIA

problem, we use, in a stepwise fashion, multi- and, through the analysis of their specificity, of
ple sets of primers spanning different regions the most promising targets for vaccine design.
of the variable region leader sequence that This approach was originally proposed by
allow us to obtain, with almost 100% efficiency, Dennis Burton as a way to identify conserved
VH and VL sequences from all isolated plasma epitopes in the highly variable viral glycopro-
cell clones. teins, such as the Env of HIV-1 (18), but in a
broader sense can be also used to identify in
complex pathogens the molecules that induce
A TARGET-AGNOSTIC APPROACH the most potent neutralizing response, with-
FOR ANTIGEN DISCOVERY FOR out prior knowledge of their nature.
VACCINE DESIGN A relevant example is provided by human
cytomegalovirus (HCMV), a herpes virus
The high throughput of the cell-based screens that uses several glycoproteins to infect
and the possibility of directly assessing the human cells. Previous studies focused on
function of the antibody (rather than just gB or gHgL, which are abundant proteins
binding) are essential elements to implement that were known to be targeted by neutral-
a target-agnostic approach that aims at the izing antibodies. Using a target-agnostic
identification of the most effective antibodies approach, we isolated from immune donors

FIGURE 4 A target-agnostic approach to antibody discovery and vaccine design. In analytic vaccinology,
donors that have developed a protective response are identied and memory B cells/plasma cells are
interrogated to identify the most effective neutralizing antibodies in terms of potency and breadth. The
antibodies are then used to identify the target antigen and to probe its correct conformation when the
latter is produced as a recombinant vaccine. The vaccine is expected to elicit antibodies of the same
quality as those originally isolated. In addition, the recombinant molecules can be used to identify
cellular receptors. doi:10.1128/microbiolspec.AID-0018-2014.f4
CHAPTER 7 Efcient Methods To Isolate Human mAbs 137

a large panel of antibodies based on their ACKNOWLEDGMENT


capacity to neutralize infection of fibroblasts
Conflicts of interest: We disclose no conflicts.
or epithelial cells by a primary HCMV isolate
and identified a group of unusually potent
antibodies that selectively neutralized infec-
CITATION
tion of epithelial, endothelial, and myeloid
cells at concentrations 1,000-fold lower than Corti D, Lanzavecchia A. 2014. Efficient
antibodies specific for gB or gHgL (19). We methods to isolate human monoclonal anti-
found that these antibodies recognize multiple bodies from memory B cells and plasma cells.
antigenic sites on a pentameric complex of Microbiol Spectrum 2(5):AID-0018-2014.
gH/gL/pUL128/pUL130/pUL131A, which
was previously described to be required
REFERENCES
for infection of endothelial epithelial and
myeloid cells but was not known to be the 1. Burton DR, Barbas CF. 1994. Human antibodies
target of neutralizing antibodies. We then from combinatorial libraries. Adv Immunol 57:
191280.
produced a soluble pentameric complex that 2. Hoogenboom HR. 2002. Overview of antibody
correctly displays all the neutralizing epitopes phage-display technology and its applications.
and elicits in mice very high titers of neutral- Methods Mol Biol 178:137.
izing antibodies. In addition, the soluble recom- 3. Green LL. 1999. Antibody engineering via
binant pentamer produced is used to identify genetic engineering of the mouse: XenoMouse
strains are a vehicle for the facile generation
the cellular receptors and dissect the mecha- of therapeutic human monoclonal antibodies.
nisms of virus entry in different cell types. We J Immunol Methods 231:1123.
refer to this process as analytic vaccinology, 4. Wardemann H, Yurasov S, Schaefer A, Young J,
since it is essentially based on the analysis of the Meffre E, Nussenzweig M. 2003. Predominant
autoantibody production by early human B cell
antibody response to the pathogen (Fig. 4).
precursors. Science 301:13741377.
Another example of analytic vaccinology 5. Wrammert J, Smith K, Miller J, Langley WA,
is provided by the analysis of the neutralizing Kokko K, Larsen C, Zheng N-Y, Mays I, Garman
antibody response to the F protein of RSV (20). L, Helms C, James J, Air GM, Capra JD, Ahmed
Previous studies suggested that the postfusion R, Wilson PC. 2008. Rapid cloning of high-
F protein could be developed as a vaccine, affinity human monoclonal antibodies against
influenza virus. Nature 453:667671.
since it was recognized by palivizumab, a 6. Scheid JF, Mouquet H, Feldhahn N, Seaman
neutralizing monoclonal antibody that is MS, Velinzon K, Pietzsch J, Ott RG, Anthony
used to prevent RSV infection in newborns RM, Zebroski H, Hurley A, Phogat A, Chakrabarti
(21). In contrast, we found that, of 30 neu- B, Li Y, Connors M, Pereyra F, Walker BD,
tralizing antibodies tested, only 4 bound to Wardemann H, Ho D, Wyatt RT, Mascola JR,
Ravetch JV, Nussenzweig MC. 2009. Broad diver-
the postfusion protein while the remaining, sity of neutralizing antibodies isolated from mem-
including the MPE8 antibody that cross- ory B cells in HIV-infected individuals. Nature
neutralizes four paramyxoviruses, were specific 458:636640.
for the prefusion F conformation. These find- 7. Dekosky BJ, Ippolito GC, Deschner RP, Lavinder
JJ, Wine Y, Rawlings BM, Varadarajan N,
ings suggest that the prefusion protein should
Giesecke C, Drner T, Andrews SF, Wilson
be considered as the most effective vaccine PC, Hunicke-Smith SP, Willson CG, Ellington
capable of inducing a broad spectrum of anti- AD, Georgiou G. 2013. High-throughput se-
bodies that neutralize but fail to react with quencing of the paired human immunoglobulin
the abundant postfusion F protein. Efforts to heavy and light chain repertoire. Nat Biotechnol
produce a stabilized prefusion form will be 31:166169.
8. Traggiai E, Becker S, Subbarao K, Kolesnikova
facilitated by the use of the antibodies that can L, Uematsu Y, Gismondo MR, Murphy BR,
be used as probes to test the conformation and Rappuoli R, Lanzavecchia A. 2004. An efficient
stability of different constructs. method to make human monoclonal antibodies
138 CORTI AND LANZAVECCHIA

from memory B cells: potent neutralization of human monoclonal antibodies that potently
SARS coronavirus. Nat Med 10:871875. neutralize human cytomegalovirus infection by
9. Corti D, Voss J, Gamblin SJ, Codoni G, Macagno targeting different epitopes on the gH/gL/
A, Jarrossay D, Vachieri SG, Pinna D, Minola A, UL128-131A complex. J Virol 84:10051013.
Vanzetta F, Silacci C, Fernandez-Rodriguez 20. Corti D, Bianchi S, Vanzetta F, Minola A, Perez
BM, Agatic G, Bianchi S, Giacchetto-Sasselli I, L, Agatic G, Guarino B, Silacci C, Marcandalli
Calder L, Sallusto F, Collins P, Haire LF, J, Marsland BJ, Piralla A, Percivalle E, Sallusto
Temperton N, Langedijk JPM, Skehel JJ, F, Baldanti F, Lanzavecchia A. 2013. Cross-
Lanzavecchia A. 2011. A neutralizing antibody neutralization of four paramyxoviruses by a human
selected from plasma cells that binds to group 1 monoclonal antibody. Nature 501:439443.
and group 2 influenza A hemagglutinins. Science 21. The-IMPACT-RSV-Study-Group. 1998.
333:850856. Palivizumab, a humanized respiratory syncy-
10. Kwakkenbos MJ, Diehl SA, Yasuda E, Bakker tial virus monoclonal antibody, reduces hospital-
AQ, van Geelen CMM, Lukens MV, van Bleek ization from respiratory syncytial virus infection
GM, Widjojoatmodjo MN, Bogers WMJM, Mei in high-risk infants. Pediatrics 102:531537.
H, Radbruch A, Scheeren FA, Spits H, Beaumont 22. Rockx B, Corti D, Donaldson E, Sheahan T,
T. 2010. Generation of stable monoclonal anti- Stadler K, Lanzavecchia A, Baric R. 2008.
body-producing B cell receptor-positive human Structural basis for potent cross-neutralizing
memory B cells by genetic programming. Nat Med human monoclonal antibody protection against
16:123128. lethal human and zoonotic severe acute respira-
11. Walker LM, Phogat SK, Chan-Hui P-Y, Wagner tory syndrome coronavirus challenge. J Virol
D, Phung P, Goss JL, Wrin T, Simek MD, Fling 82:32203235.
S, Mitcham JL, Lehrman JK, Priddy FH, Olsen 23. Corti D, Langedijk JPM, Hinz A, Seaman
OA, Frey SM, Hammond PW, Kaminsky S, MS, Vanzetta F, Fernandez-Rodriguez BM,
Zamb T, Moyle M, Koff WC, Poignard P, Silacci C, Pinna D, Jarrossay D, Balla-
Burton DR. 2009. Broad and potent neutralizing Jhagjhoorsingh S, Willems B, Zekveld MJ,
antibodies from an African donor reveal a new Dreja H, OSullivan E, Pade C, Orkin C, Jeffs
HIV-1 vaccine target. Science 326:285289. SA, Montefiori DC, Davis D, Weissenhorn
12. Huang J, Doria-Rose NA, Longo NS, Laub L, W, McKnight A, Heeney JL, Sallusto F,
Lin C-L, Turk E, Kang BH, Migueles SA, Sattentau QJ, Weiss RA, Lanzavecchia A.
Bailer RT, Mascola JR, Connors M. 2013. 2010. Analysis of memory B cell responses and
Isolation of human monoclonal antibodies from isolation of novel monoclonal antibodies with
peripheral blood B cells. Nat Protoc 8:19071915. neutralizing breadth from HIV-1-infected indi-
13. Steinitz M, Klein G, Koskimies S, Makel O. viduals. PLoS One 5:e8805. doi:10.1371/journal.
1977. EB virus-induced B lymphocyte cell lines pone.0008805.
producing specific antibody. Nature 269:420422. 24. Sabin C, Corti D, Buzon V, Seaman MS, Lutje
14. Bernasconi N, Onai N. 2003. A role for Toll-like Hulsik D, Hinz A, Vanzetta F, Agatic G,
receptors in acquired immunity: up-regulation Silacci C, Mainetti L, Scarlatti G, Sallusto F,
of TLR9 by BCR triggering in naive B cells and Weiss R, Lanzavecchia A, Weissenhorn W.
constitutive expression in memory B cells. Blood 2010. Crystal structure and size-dependent
101:45004504. neutralization properties of HK20, a human
15. Ruprecht CR, Lanzavecchia A. 2006. Toll-like monoclonal antibody binding to the highly con-
receptor stimulation as a third signal required served heptad repeat 1 of gp41. PLoS Pathog 6:
for activation of human naive B cells. Eur J e1001195. doi:10.1371/journal.ppat.1001195.
Immunol 36:810816. 25. Corti D, Suguitan AL, Pinna D, Silacci C,
16. Thorley-Lawson DA. 2001. Epstein-Barr Fernandez-Rodriguez BM, Vanzetta F, Santos
virus: exploiting the immune system. Nat Rev C, Luke CJ, Torres-Velez FJ, Temperton NJ,
Immunol 1:7582. Weiss RA, Sallusto F, Subbarao K, Lanzavecchia
17. Bernasconi NL, Traggiai E, Lanzavecchia A. A. 2010. Heterosubtypic neutralizing antibodies
2002. Maintenance of serological memory by poly- are produced by individuals immunized with
clonal activation of human memory B cells. a seasonal influenza vaccine. J Clin Invest 120:
Science 298:21992202. 16631673.
18. Burton D. 2002. Antibodies, viruses and 26. Barfod L, Bernasconi NL, Dahlbck M, Jarrossay
vaccines. Nat Rev Immunol 2:706713. D, Andersen PH, Salanti A, Ofori MF, Turner
19. Macagno A, Bernasconi NL, Vanzetta F, L, Resende M, Nielsen MA, Theander TG,
Dander E, Sarasini A, Revello MG, Gerna G, Sallusto F, Lanzavecchia A, Hviid L. 2007.
Sallusto F, Lanzavecchia A. 2010. Isolation of Human pregnancy-associated malaria-specific B
CHAPTER 7 Efcient Methods To Isolate Human mAbs 139

cells target polymorphic, conformational epitopes 30. Williams KL, Sukupolvi-Petty S, Beltramello
in VAR2CSA. Mol Microbiol 63:335347. M, Johnson S, Sallusto F, Lanzavecchia A,
27. Stubbs J, Olugbile S, Saidou B, Simpore J, Diamond MS, Harris E. 2013. Therapeutic
Corradin G, Lanzavecchia A. 2011. Strain- efficacy of antibodies lacking FcgR against lethal
transcending Fc-dependent killing of Plasmo- dengue virus infection is due to neutralizing
dium falciparum by merozoite surface protein potency and blocking of enhancing antibodies.
2 allele-specific human antibodies. Infect PLoS Pathog 9:e1003157. doi:10.1371/journal.
Immun 79:11431152. ppat.1003157.
28. Lindesmith LC, Beltramello M, Donaldson 31. Simmons CP, Bernasconi NL, Suguitan AL,
EF, Corti D, Swanstrom J, Debbink K, Mills K, Ward JM, Chau NVV, Hien TT,
Lanzavecchia A, Baric RS. 2012. Immunoge- Sallusto F, Ha DQ, Farrar J, de Jong MD,
netic mechanisms driving norovirus GII.4 Lanzavecchia A, Subbarao K. 2007. Prophy-
antigenic variation. PLoS Pathog 8:e1002705. lactic and therapeutic efficacy of human mono-
doi:10.1371/journal.ppat.1002705. clonal antibodies against H5N1 influenza. PLoS
29. Beltramello M, Williams KL, Simmons CP, Med 4:e178. doi:10.1371/journal.pmed.0040178.
Macagno A, Simonelli L, Quyen NTH, Sukupolvi- 32. Di Zenzo G, Di Lullo G, Corti D, Calabresi V,
Petty S, Navarro-Sanchez E, Young PR, de Silva Sinistro A, Vanzetta F, Didona B, Cianchini
AM, Rey FA, Varani L, Whitehead SS, Diamond G, Hertl M, Eming R, Amagai M, Ohyama B,
MS, Harris E, Lanzavecchia A, Sallusto F. 2010. Hashimoto T, Sloostra J, Sallusto F, Zambruno
The human immune response to Dengue virus is G, Lanzavecchia A. 2012. Pemphigus auto-
dominated by highly cross-reactive antibodies antibodies generated through somatic mutations
endowed with neutralizing and enhancing activ- target the desmoglein-3 cis-interface. J Clin Invest
ity. Cell Host Microbe 8:271283. 122:37813790.
Use of Human Hybridoma Technology
To Isolate Human Monoclonal
Antibodies

SCOTT A. SMITH1 and JAMES E. CROWE, JR.2


8
HISTORY OF HYBRIDOMAS

Monoclonal antibodies (mAbs) have revolutionized the conduct of science


since their first description in 1975 (1). The use of these specific reagents also
has made possible improved clinical diagnostics in the medical arena, and many
antibodies have found their way to clinical use as prophylactic or therapeutic
agents. Nevertheless, the potential of mAbs derived specifically from technology
based on human hybridomas remains largely unfulfilled. The principal reason
for the lack of a large number of hybridoma-derived mAb therapeutics
has simply been the technical difficulty in generating stable hybridomas that
secrete human mAbs of high affinity and functional activity. This chapter
reviews recent efforts to develop and employ novel methods for the efficient
generation of human hybridomas secreting human mAbs for clinical use.
The principal advantage of the use of human hybridoma technology for
mAb generation is that this approach preserves the authentic sequence and
pairing of antibody DNA from a natural B cell for the expression of a
naturally occurring full-length human mAb. There are significant theoret-
ical advantages for expressing cDNAs encoding authentic heavy and light

1
Vanderbilt Vaccine Center and Department of Medicine, Vanderbilt University Medical Center, Nashville,
TN 37232; 2 Vanderbilt Vaccine Center and Departments of Pediatrics and Pathology, Microbiology,
and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0027-2014

141
142 SMITH AND CROWE

chains with chains that are paired using the was evident early on and was the driving
coding sequence as it was generated natu- force behind intense effort put into the
rally through B cell selection, class switch, and development of human hybridoma methods.
affinity maturation. No genetic modification of Initial studies were done in the early 1970s
these sequences is required. Since antibody using mouse myeloma cells, fusing them
expression is typically very stable in hybrid- with primary human B cells (2). A human
omas, sequence amplification of antibody tetraploid hybridoma also was made through
variable genes is achieved easily if recombi- the fusion of two human lymphocyte lines (3).
nant production or manipulation is desired. It was not until 1980, however, that the first
The resulting recombinant mAb retains most successful human mAb was produced (4). This
features of naturally occurring human anti- feat was achieved by fusing lymphoid cells
bodies, as the clone retains the native amino harvested from spleens obtained during stag-
acid sequence and heavy/light chain pairing. ing laparotomy from patients with Hodgkins
Since the native constant region of the anti- lymphoma with the human myeloma cell line
body in the original human B cell is retained in U266. This accomplishment was a large step
the mAb expressed by the resulting hybrid- toward the use of human hybridomas to make
oma, the functional properties of the particu- mAbs, as it proved the feasibility of the
lar Fc region can be studied for Fc-mediated method. The large numbers of lymphoid cells
activities, such as antibody-dependent cel- used in that fusion process were sufficient to
lular cytotoxicity. overcome the low fusion efficiency at the time.
Despite the advantages inherent to mak- The challenge of identifying antigen-specific
ing natural human mAbs, the low efficiency cells and expanding them to numbers that
of the hybridoma isolation process histori- enabled researchers to overcome the barrier of
cally was a technical drawback that was too low fusion efficiency would, however, require
great to overcome, and many other methods several more decades of investigation.
of producing human mAbs have been used One significant obstacle to the genera-
instead in recent years to meet the demand for tion of human hybridomas over the years
generation of therapeutic antibodies. The was the inability to consistently expand
principal disadvantage of the hybridoma desired populations of antigen-specific B
method is low fusion efficiency. Over the last cells. Antigen-specific memory B cells gen-
several decades, however, this problem has erally circulate at low frequencies in the
been overcome slowly through improvements peripheral blood, typically in a range that
in several technical features of the process. centers around 1 in 10,000 B cells or lower.
These improvements will be discussed in For example, precursor frequencies of anti-
more detail throughout this article. Currently, tetanus toxoid-specific B cells were initially
human hybridomas can be generated with reported following immunization as being
great efficiency and throughput. Panels of approximately 1 per 10,000 peripheral blood
antigen-specific human hybridomas secreting mononuclear cells (PBMCs) (5). Fusion effi-
full-length naturally occurring mAbs to a ciencies typically have not been sufficient to
large number of targets have been developed immortalize enough cells from the number of
recently using human peripheral blood mono- B cells that can be obtained from humans by
nuclear cells as the starting material. routine phlebotomy. When combined with a
fusion efficiency of 1 in 100,000 B cells in
the suspension, as is typical of polyethylene
FIRST HUMAN HYBRIDOMAS glycol (PEG)-mediated cell fusion, one would
need approximately 1 liter of blood to obtain a
The prospect of using human mAbs for the single tetanus toxoid-specific human hybrid-
prevention or treatment of human diseases oma under these circumstances. Consequently,
CHAPTER 8 Use of Hybridoma Technology To Isolate Human mAbs 143

the generation of human hybridoma cells specific EBV-immortalized B cell line with a
secreting desirable human mAbs without hypoxanthine phosphoribosyl transferase-
expanding the frequency or number of anti- deficient human lymphoblastoid partner
gen-specific B cell populations was difficult. created previously. Many groups have now
Human B cells can be immortalized by used variations of these techniques to employ
methods using Epstein-Barr virus (EBV) EBV transformation to expand populations of
transformation (6, 7). EBV transformation antigen-specific B cells. These methods have
is often carried out on samples containing improved mAb yield and hybridoma gen-
large numbers of B cells, either from PBMCs eration throughput considerably. In fact, EBV
or cells obtained from lymphatic tissues such transformation continues to be used to this
as tonsil cells. These EBV-transformed B cell day to expand B cells prior to human hybrid-
lines, which contain many clones of differing oma generation.
specificities, must be grown for a considerable
period of time to allow for the emergence of
immortalized lines. Since immortalization B CELL SOURCE
does not occur with every clone of trans-
formed B cells, production of antigen-specific One of the critical factors underlying the
clones was historically very difficult. More- difficulty in generating human hybridomas
over, EBV-transformed B cells generally grow is the low frequency of antigen-specific B
poorly, often secrete low amounts of anti- cells in peripheral blood under normal
bodies, and can exhibit chromosomal instab- circumstances. Typically, antigen-specific
ility (6, 810). Recently, the addition of CpG B cells are present in peripheral blood at
oligonucleotide a toll-like receptor (TLR) 9 a frequency of 0.1%. Therefore, large num-
agonist during B cell transformation was bers of PBMCs are required from individuals to
shown to greatly facilitate the transformation obtain a significant number of antigen-specific B
process (1113). This stimulation significantly cells. For example, if a desired antigen-specific
improved the efficiency of transformation and B cell occurs at a frequency of 1 in 1,000, and
has been used to expand antigen-specific B PBMCs contain 10% B cells, then 1 million
cells for the clonal amplification of their PBMCs contain only 100 antigen-specific B
antibody variable sequences by reverse tran- cells. Several strategies have been devised to
scriptase PCR (13). enrich and/or expand antigen-specific B cell
The prospect of using EBV-transformed populations prior to fusion.
B cells for the generation of human hybrid- The most readily available source of
omas was first realized in the early 1980s. antigen-specific B cells is peripheral blood.
This method was achieved initially by fusing For the most part, directly fusing PBMCs
a human EBV-transformed B cell line pro- with a myeloma partner is not productive in
ducing antigen-specific mAb with a murine generating a desired antigen-specific human
myeloma to produce a stable heterohybrid- hybridoma, due to the limited number of
oma secreting mAb (14, 15). This procedure antigen-specific cells in circulation. Enrich-
resulted in a more stable, higher-producing ment of antigen-specific B cells from large
clone. Hypoxanthine phosphoribosyl transfer- samples of PBMCs using either fluorescence
ase-deficient variant EBV-transformed B cell activated cell sorting or other cell isolation
lines also were developed for use as human techniques (such as magnetic bead separation)
fusion partners (16). The use of an immortal- often results in only modest improvement in
ized EBV-transformed B cell clone to create a the yield of functional cells.
stable fully human hybridoma was described The use of various cytokines, or cell lines
in 1984 (17). A fully human hybridoma was gen- made to express B cell cytokines, to amplify
erated successfully by fusing a cytomegalovirus- single B cells in culture has been described
144 SMITH AND CROWE

in the B cell field, principally using CD40- using PBMCs is to use EBV transformation
ligand (CD40L or CD154) expressing cells (25). Freshly isolated or cryopreserved PBMCs,
and recombinant human cytokines (inter- obtained from an individual or individuals
leukin 2 [IL-2], IL-4, and IL-10) (18). who have been identified as possessing B cells
Synergy was also noted with the combina- of the desired antigen specificity, can be
tion of CD40L and IL-21 (19). Feeder cell expanded in an oligoclonal manner using
lines have also been used, based on their EBV transformation. Using culture superna-
ability to support primary human B cell tant of the marmoset cell line B95.8, which
growth, frequently following gamma irradi- contains high titers of EBV, B cells can be
ation to prevent them from dividing. Several infected via complement receptor 2 (CD21),
feeder cell lines have been described for this and transformants emerge over the course
purpose: a fibroblast cell line transfected with of 1 to 2 weeks, forming large colonies of
the human CD40L molecule (20), an immor- cells known as lymphoblastoid cells (often
talized cell line FDC-H1 with features of abbreviated LCLs) (26). Cultures can be
human follicular dendritic cells (21), the U- supplemented with cyclosporin A to inhibit
937 human macrophage-like cell line (ATCC- EBV-specific T cells, which are present in
CRL-1593.2) (22), and the EL4-B5 mutant many individuals, from killing B cell trans-
thymoma cell line (23). formants (27). The overall efficiency of EBV
In addition to human CD40L, B lympho- transformation was improved dramatically
cyte stimulating factor (BLyS), also known through the use of CpG oligonucleotide,
as BAFF (for B cell activating factor belong- increasing transformation of B cell popula-
ing to the tumor necrosis factor family) tions from <10% to >30% (13) (see Fig. 1).
(24), has been used to support primary B Recently it was shown that by adding an
cells in culture providing costimulatory inhibitor of EBV-associated apoptosis, a
signals. BAFF is a relatively new member pharmacologic inhibitor of the serine/thre-
of the tumor necrosis factor family that was onine-protein kinase 2, which is required
simultaneously identified by four different for checkpoint-mediated cell cycle arrest,
laboratories and named BAFF, BLyS, TALL-1, results in further improvement in lympho-
THANK, and zTNF4. The cytokine is ex- blastoid cell survival and expansion (28, 29).
pressed abundantly in monocytes and macro- In the end, increased numbers of lympho-
phages and is upregulated by interferon blastoid cells result in a greater probability of
gamma. Endogenous BAFF is processed successfully producing hybridomas secreting
intracellulary by a protease of the furin family antibodies of interest. Next, EBV-transformed
of proprotein convertases. Secreted BAFF acts
as a potent B cell growth factor. BAFF plays an
important role as costimulator of B cell
proliferation and function. Using soluble
cytokines or feeder cells able to provide cost-
imulator function and/or cytokines, single B
cells can be expanded to clones of a few hun-
dred cells in several weeks. Antigen-specific B
cell clones can be confirmed by testing cell
supernatant specificity using enzyme-linked
immunosorbent assay. The expanded antigen-
specific cells then can be immortalized by
fusion with a myeloma to form a hybridoma.
The simplest and most common strategy FIGURE 1 Lymphoblastoid cell formation from
for the amplification of B cell populations PBMCs. doi:10.1128/microbiolspec.AID-0027-2014.f1
CHAPTER 8 Use of Hybridoma Technology To Isolate Human mAbs 145

B cell cultures are screened for antigen-specific membrane, allowing for introduction of the
antibody production using enzyme-linked viral nucleocapsid into the cytoplasm of the
immunosorbent assay or other multiwall plate host cell. The use of Sendai virus for cell fusion
assays. Cultures producing the desired anti- was described thoroughly by Okada in 1993
body then are fused with a myeloma cell line in (32). One advantage of using Sendai virus is its
an oligoclonal manner. The resulting oligo- broad tropism, which is mediated by the
clonal hybridoma lines are then biologically hemagglutinin-neuraminidase protein that
cloned by a physical method to isolate single recognizes sialic acid as a receptor on the
cells, using one of the methods discussed later host cell membrane. Kits containing inacti-
in this article, to produce a human hybridoma vated Sendai virus are commercially available
secreting an antigen-specific mAb. for the generation of murine hybridomas. The
second virus that has been used for the fusion
FUSION METHODS of cells to generate hybridomas for mAb
secretion is vesicular stomatitis virus (33).
The most difficult and critical step in the In a study performed by Nagata et al., the
production of human hybridomas is the efficiencies of hybridoma generation were
ability to efficiently fuse desired populations compared using Sendai virus, vesicular stoma-
of lymphocytes with a myeloma partner. titis virus, and PEG-mediated cell fusion (34).
There are three basic techniques used to Interestingly, their results suggested that the
generate hybridomas for the purpose of mAb isotypes of antibodies obtained were influ-
production: (i) the use of chemical agents such enced by the fusion method, as vesicular
as PEG, (ii) fusogenic viruses, and (iii) electri- stomatitis virus-mediated fusion resulted in
cal cytofusion. The first method used to study greater production of IgG mAbs (34). A related
cell fusion took advantage of viruses, principally method that has been used is to transduce cells
Sendai virus, capable of fusing together the with viral fusogenic proteins. Unfortunately,
membranes of two cells. The most common the resulting cells often continue to fuse for
technique, however, employed in the 1975 as long as the proteins are expressed, which
seminal paper by Kohler and Milstein (1), makes this method less optimal.
with variations of the theme still commonly The most common method used to pro-
used today, involves chemical fusion using duce hybridomas takes advantage of the
PEG. Finally, protocols based on the use of fusogenic properties of PEG, discovered by
electrical currents to align and fuse the Kao and Michayluk in 1974 (35). The exact
membrane of cells for the efficient generation mechanism by which PEG-mediated cell fu-
of hybridomas were developed and optimized. sion occurs is not known. Most experts believe
Viruses were first used as a means to fuse that the mechanism primarily involves volume
cells in the 1960s and early 1970s (30). exclusion, driving adjacent cell membranes
There are several viruses, or their fusion together (36). Unfortunately PEG is quite toxic
proteins, which have been used successfully to cells, as it can fuse multiple cells, resulting
for the purpose of hybridoma generation. The in giant polykaryons, and can even result in
two most commonly used are Sendai virus the fusion of intracellular membrane struc-
and vesicular stomatitis virus. Sendai virus, tures (37). The fusion process, however, is
also referred to as murine parainfluenza virus relatively simple and cheap. B cells are mixed
type 1 or hemagglutinating virus of Japan, is a with a myeloma fusion partner and suspended
paramyxovirus which was the first animal in a solution containing PEG in a drop-wise
virus whose mode of infection was elucidated fashion. After a short incubation, fusion solu-
(31). The viral fusion protein, or F protein, tion is removed and the cells are washed and
makes up part of the exposed spikes on the resuspended in selection medium. Different
envelope of the virus and inserts into the cell PEG preparations have been shown to result
146 SMITH AND CROWE

in different efficiencies of cell fusion (38). meabilization may be under the control of
The main problem with using PEG for the the electric field parameters. The rate of
generation of human hybridomas is its resealing of the pore in the membrane may
low efficiency of fusion. Estimates of aver- be influenced by both pulse duration and
age fusion efficiencies are in the order of 1 number but is independent of the electric
hybridoma generated per 100,000 starting B field intensity that creates the permeabiliza-
cells. When fusing murine splenocytes from tion (40). Therefore, optimal evaluation of
hyper-immunized animals (which can possess critical parameters for membrane perme-
about 108 B cells), an extremely low ineffi- abilization requires flexible control over each
ciency can be tolerated if one simply aims aspect of the electrical field. One of the more
to find one or several mAbs that bind to an recent innovations in the electroporation
antigen. However, when using human samples hardware used for this purpose is that many
such as blood (yielding about 1 x 106 PBMCs of these parameters can be controlled and
or about 50,000 B cells per ml of blood), or varied independently. Previously designed
more precious samples such as tumor infil- electroporators did not have such precise
trating lymphocytes, such inefficiencies make control of all variables, thereby likely intro-
success unlikely. ducing uncontrolled variation in experiments.
The most efficient method of cell fusion Membrane effects of applied electrical
and hybridoma generation is electrical fields in electrofusion are similar to those in
cytofusion. This method of cell fusion is electroporation except that membranes in
an essential step in the most innovative close contact can fuse together during the
techniques in modern human hybridoma process of pore formation. Therefore, elec-
protocols and has been optimized for the tric field intensities used in electrofusion
purpose of human hybridoma generation are similar to those used in electroporation.
(39). At the center of this membrane fusion Electrofusion is performed in a sequence of
technique is the concept of electroporation. steps. First, cells are brought into contact
The application of high-intensity electric with other cells. Cell-cell contact can be
field pulses to cells causes transient mem- achieved by several methods, although none
brane permeabilization. The extent of per- has been sufficiently optimized in our
meabilization is thought to depend on several estimation. Chemical methods such as avi-
physical parameters associated with the tech- din-biotin bridging have been used to bring
nique such as pulse intensity, number, dura- together two cell types specifically (4144).
tion, shape, and interval. Electric field Chemical methods require more manipul-
intensity is the most critical parameter in the ation than other methods but can be useful
induction of permeabilization. The intensity in certain circumstances. Physical methods
must exceed a critical threshold for membrane such as centrifugation can bring cells into
permeabilization to be induced. The electric contact prior to (or after) the fusion pulse
field intensity delivered depends on the cell (45). Electroacoustic fusion of cells in sugar
size. The extent of permeabilization (which solutions and of cells brought into contact
correlates experimentally with the flow rate in an ultrasonic standing wave field has been
across the membrane) is controlled by both described (46). In fact, simple centrifugation
pulse number and duration (40). Increasing followed by electroporation can be used to create
the electric field intensity above the critical cell fusion. The most commonly described
threshold needed for permeabilization results effective method for bringing cells into contact
in an increase in the area of the membrane that prior to electrofusion, however, is termed
is involved. Permeabilization is transient and dielectrophoresis. Dielectrophoresis is achieved
disappears with time after delivery of the within a suspension of cells using an alternat-
electric field pulses. The half-life of per- ing current electrical field.
CHAPTER 8 Use of Hybridoma Technology To Isolate Human mAbs 147

In any fusion method, sufficient force the same formula used for electroporation.
must be applied to each cell to overcome the Multiple fusion pulses may be more effi-
negative surface charge. If electricity is cient than a single pulse.
used, merely applying a uniform electric The last step in the electrofusion process
field will not move a cell because the net (when using dielectrophoresis as an align-
charge of the cell is zero. Thus, from the ment tool) is postfusion alignment using alter-
definition of electric field there is no force nating current fields. Electrofusion is a process
applied. However, a nonuniform field moves that continues to occur over some time after
the positive ions inside each cell to one side the fusion pulse is applied. Reapplying di-
and the negative ions to the opposite side, electrophoresis after the fusion pulse allows
producing a dipole. Once the dipole is induced, maturation of the fusion process by holding
a net force is exerted on the cell because cells in optimal alignment and contact.
the intensity of the field is greater on one side Optimizing of dielectrophoresis and
than the other. The movement of cells in one electrofusion parameters has been achieved
direction causes the cells to concentrate in partly via observable events through the use
an area. Since the cells are now dipoles, of a microscope slide or coaxial electrode.
the negative side of one cell will attract the Pearl chain length (length of aligned cells)
positive side of another cell, overcoming the can be increased by increasing the voltage
negative surface charge. A photomicrograph or increasing the time of prefusion sine
illustrating pearl chain formation is shown wave application. Increased pearl chain
in Fig. 2. length may not always be advantageous,
The second step in electrofusion is to however. It is unclear at this point whether
apply one or more high-voltage pulses to long pearl chains are advantageous or not.
the cells, inducing membrane fusion. The According to Zimmermann (47), the fusion
voltage required must be above a threshold of adjacent cell membranes of cells in a
to induce membrane breakdown and below pearl chain is a stochastic process. Thus, the
a maximum voltage that causes cell death. electroporation conditions generate a probabil-
The threshold voltage is approximately one ity of fusion of adjacent cell membranes. This
volt across the cell membrane or two volts means that it is possible to select electrofu-
across two membranes (47). The voltage sion conditions that generate predominantly
across a cell is equal to 1.5 times the cell two cell fusions, even if pearl chain length is
radius times the electric field strength times long in contrast to two-cell-only pearl chains.
the cosine of the angle of the membrane in The goal of electrofusion, at least as ap-
relation of the direction of the field. This is plied to hybridoma formation, is to generate

FIGURE 2 (A) Pre- and (B) post-pearl chain formation. doi:10.1128/microbiolspec.AID-0027-2014.f2


148 SMITH AND CROWE

the most B cell-myeloma hybridomas. for fusion with human B cells are available at
Through the use of selective medium pre- the American Type Culture Collection.
parations, fusions of like cells (either B cells or A number of fully human myeloma cell
myeloma cells) are not productive. Likewise, lines have been used successfully in fusion
unfused B cells and myeloma cells are also for the generation of human hybridomas.
unable to survive. This selection is achieved The most studied of these is the human
through the use of medium containing HAT myeloma U266 and its derived lines. U266
(hypoxanthine-aminopterin-thymidine) and was originally isolated from a patient with
ouabain. Unfused myeloma cells or fused multiple myeloma and found to secrete
myeloma cells that do not contain a B cell IgE (48). This myeloma was made HAT-
nucleus are killed by aminopterin. Without sensitive and used as the first human fusion
being fused to a B cell, the myeloma fusion partner to generate fully human hybridomas
partner is not able to perform de novo (4, 49). The U266-derived SKO-007 line,
synthesis of DNA and is forced to undergo made to be 8-azaguanine sensitive, was also
apoptosis. The B cell possesses the ability to used to generate some of the first fully
overcome this blockade by performing the human hybridomas (5052). The human
salvage pathway, for which hypoxanthine and line LICR-LON-HMy2, also derived from
thymidine are used as raw material for malignant patient cells, was compared to
building DNA. Unfused B cells or fused B SKO-007 and found to be superior (51, 53).
cells that do not contain a myeloma cell LICR-LON-HMy2 was used to generate a
nucleus are killed by ouabain. Without being hybridoma secreting mAb through the
fused to the ouabain-resistant myeloma cell, fusion of lymph node lymphocytes from a
the B cell (particularly transformed B cells) is patient with breast cancer (54). The human
highly susceptible to the plasma membrane myeloma line RPMI 8226, which expressed
sodium pump inhibiting effects of ouabain, free light chain, was also used in the initial
and apoptosis is induced. The only products of attempts to make fully human hybridomas
fusion to survive in the selective medium are (55, 56). The human myeloma-like cell
the B cell-myeloma hybridomas. line KR12 was generated by fusion of the
human plasmacytoma line RPMI8226 with
the human lymphoblastoid cell line KR-4
FUSION PARTNERS (14, 57). A more recently isolated human
myeloma fusion partner, designated Karpas
Murine myeloma cells, originally developed 707H, was developed for the purpose of
for the production of mouse mAbs, were not making human hybridomas (58). Like U266,
suitable as fusion partners for human B cells, Karpas 707H was established from a patient
as they produced unstable heterospecific with multiple myeloma. It was made to be both
hybrids, often resulting in rejection of the HAT-sensitive and ouabain-resistant, so it
relevant human chromosomes. The develop- could be used with EBV-transformed B cells.
ment and refinement of suitable fusion Unfortunately, many laboratories attempting
partners has been an ongoing and instrumen- to make human hybridomas using these
tal part of the human hybridoma technology. myeloma lines met with only limited success.
Investigators have been developing myeloma A larger group of fusion partners, them-
cell lines suitable for fusion with human B cells selves being heterohybridomas, have been
since the early 1980s. In general, these lines developed by fusing murine myeloma cell
can be divided into two major types: fully lines with human cells. These fusion partners
human and heterohybridomas constructed by are nonsecreting mouse-human hybrids that
fusing murine myeloma cells with human are often made resistant to ouabain so they can
cells. Several of the myeloma cell lines suitable produce stable hybridomas upon fusion with
CHAPTER 8 Use of Hybridoma Technology To Isolate Human mAbs 149

human EBV-transformed B cells. Since the hybridoma fusion partner denoted SPYMEG
heterohybridoma partner does not secrete an was developed by fusion of Sp2 murine
antibody, antibody secreted from the resulting myeloma cells and MEG-01 human mega-
hybridoma is encoded by the nucleus of the karyoblastic leukemia cells (74). Recently,
fused B cell. Ostberg and Pursch described the SPYMEG was employed for the production
production of heterohybridomas made resis- of anti-influenza mAbs, generated by fusing
tant to 8-azaguanine and used to generate PBMCs from influenza-vaccinated and natu-
human hybridomas secreting anti-influenza rally infected volunteers (75, 76). It was also
mAbs (59). One of these heteromyeloma used successfully to make human hybridomas
fusion lines, SPAZ-4, was used later to gener- secreting anti-HIV neutralizing mAbs by
ate human mAbs with specificities to various fusion with PBMCs obtained from HIV-1-
malignancies by fusion with lymph node infected individuals (77). A more recently
lymphocytes from patients with carcinoma created line, MFP-2, is a trioma that was
(60, 61). Two heterohybridoma cell lines, generated by fusing a murine myeloma cell
SHM-D33 and HMMA 2.5, continue to be line with a human myeloma cell line, yielding
used for production of human mAbs. SHM- the intermediate heteromyeloma B6B11.
D33 was generated by fusing the mouse mye- This heterohybridoma was then fused with a
loma cell line P3X63Ag8 with human myeloma human lymphocyte to generate the MFP-2
cell line FU-266 (62). This fusion partner has trioma line (78). The authors used this partner
been used very successfully to generate large to develop mAbs with specificity to breast
panels of naturally occurring human anti-HIV cancer tissue and cell lines (78, 79). The MFP-
mAbs (6365). Another heterohybridoma 2 fusion partner was also used to produce
fusion partner that has been very produc- several anti-West Nile virus mAbs (80).
tive is HMMA 2.5 (66), which was generated Recently, a heterohybridoma fusion partner
using a multistep process. The parental line, cell line was derived from the murine myelo-
HMMA2.11TG/O, was made by first fusing ma cell line Sp2 and modified to coexpress
bone marrow mononuclear cells from a pa- genes encoding murine interleukin-6 and
tient with IgA myeloma with the mouse human telomerase catalytic subunit (TERT)
myeloma cell line P3X63Ag8.653. HMMA (81). The expression of murine IL-6 directly
2.5 is a subclone that then was selected for stimulates proliferation as well as immuno-
optimal fusion efficiency after passaging in 6- globulin production from the resulting hybrid-
thioguanosine. Cell line HMMA 2.5 was found oma. Human TERT can lengthen telomeres,
to achieve the highest fusion efficiency when it thereby providing cells with unlimited repli-
was compared to six other myeloma cell lines cation capability and promoting karyotypic
using electrofusion (39). This heterohybrid- stability. The Sp2/mIL-6/hTERT heterohy-
oma has been used to generate large panels bridoma line then was demonstrated to pro-
of naturally occurring fully human mAbs to duce stable hybridomas able to secrete fully
many viruses, including influenza and dengue human mAbs by fusing splenic B cells from a
viruses (29, 6769). patient immunized with a Streptococcus pneu-
Another heterohybridoma line used to moniae vaccine. The authors succeeded in
successfully generate human hybridomas is creating hybridomas that secrete human mAbs
K6H6/B5 (7072). This line was developed specific for S. pneumoniae antigens (81).
by fusing malignant lymphoid cells isolated
from a patient with nodular lymphoma with
the mouse myeloma cell line NS-1-Ag4. BIOLOGICAL CLONING METHODS
K6H6/B5 was used to successfully create
human hybridomas that secrete mAbs to The final step in the generation of a human
hepatitis C virus (73). A very unique hetero- hybridoma is to isolate successful fusion
150 SMITH AND CROWE

products as clones derived from single by counting the total number of cells to be
cells, a process often referred to as biolo- added to a given number of wells, such as
gical cloning. There are several advantages adding approximately 77 cells to a 96-well
to isolating individual hybridomas early and culture plate. By using the Poisson distri-
growing them in a clonal manner. Frequent- bution, this dilution provides that approxi-
ly, mixed populations of mAb-producing mately 36% of wells will contain 1 cell/well. By
hybridomas are created following fusion. the same probability distribution, approxi-
Depending on the experiment, there may be mately 45% will contain 0 cells, 14% will
hundreds or even thousands of different contain 2 cells, 4% will contain 3 cells, and
clones produced. Cell cloning is an essential approximately 1% of wells will contain 4 cells.
step in ensuring that a monoclonal antibody Thus, by using this method, one would expect
is ultimately generated. Also, hybridoma that 19% of wells would contain >1 cell. Since
clones that produce the greatest quantity hybridomas vary greatly in their ability to
of mAbs are relatively rare following fusion survive and make antibody, this calculation is
and consume a greater amount of energy and often an overestimation of final growth and
nutrients. Because of this, high-producing mAb expression. Limiting dilution plating is
clones often grow slower and can be over- often performed using multiple plates and
grown easily by low- or nonproducing hybrid- repeated for several rounds to improve the
oma clones (82). There are now several likelihood of isolating a true clone that is
methods that can be used to perform biological both stable and produces a high level of mAb.
cloning of human hybridomas. Traditionally, If greater cloning stringency is desired,
this was accomplished by limiting dilution hybridomas are plated at 0.3 cells/well,
plating. More recently, advances in flow resulting in the percent of wells with >1 cell
cytometric automated single-cell sorting, being only 3.3%, and this process can be
with indexing capabilities, have allowed for repeated in a serial fashion two or three
fast, accurate, and versatile single-cell plating. times. This method has the advantage of
Finally, semisolid medium preparations can be being simple and relatively inexpensive in
used to grow single hybridoma cells as isolat- terms of equipment. However, it is very time
ed, suspended colonies. With special clone- consuming and labor intensive, it has low
picking devices, this process can be highly throughput, and obtaining clonality is never
automated and can also be performed in an guaranteed (85, 86). One cannot be certain
antigen-specific and semi-quantitative fashion that the starting population of hybridomas
for selection and biological cloning of high- is in a homogenous single-cell suspension,
producing human hybridomas (83) (Fig. 3). as some clones may stick together. Addition-
The most common method to generate ally, since only a few hundred to a few
biological clones of hybridomas is limiting thousand clones can be interrogated, the pro-
dilution cloning. This technique, and spect of identifying a high-producing clone
variations thereof, is based on the Poisson is low.
distribution (84). The Poisson distribution An alternative related approach used by
is a discrete frequency distribution that many laboratories is to perform serial
describes the probability of a number of dilutions such that the cell concentration
independent events occurring in a fixed is diluted down and across the cell culture
interval of time. If a known number of plate. By adding a known number of cells to
viable cells are added to a plate with a given the A1 position well, diluting down the A
number of wells, this model can be used to column, then diluting the A column across
provide the probability that any given well the plate with a multichannel pipette, one
would contain a given number of cells, for can achieve adequate separation of cells.
example, if one were to plate 0.8 cells/well This method has the advantage of near
CHAPTER 8 Use of Hybridoma Technology To Isolate Human mAbs 151

FIGURE 3 Cloning in (A) semi-solid medium and (B) nal human hybridoma. doi:10.1128/microbiolspec.
AID-0027-2014.f3

infinite variability, which may be important be effectively interrogated is dramatically


in isolating poor-growing or low-viability amplified. Ideally, single-cell cloning could
clones. The clear disadvantage is that only a be coupled to selection of hybridomas based
handful of wells that are likely to contain on antigen specificity and antibody quantity.
isolated clones can be assessed in each Methods for using flow cytometry to
dilution plate. Multiple rounds of dilution single-cell sort hybridomas on the basis of
plating are often used as an attempt to their antigen specificity have been devel-
ensure clonality. oped and in some cases are commercially
Increasingly, laboratories have turned to available. The use of flow cytometric sorting
sterile single-cell sorting of hybridomas to select high-producing Chinese hamster
using a flow cytometer outfitted with an ovary (CHO) cell clones was described
automated single-cell deposition unit to initially (87) and then applied shortly there-
accomplish the task of biological cloning. after to hybridomas (88). Cell sorting to
Methods that use flow cytometry and cell identify and isolate clones also has been
sorting greatly increase the number of cells used for bispecific hybridomas and isotype-
that can be screened and can almost ensure switch variants, and even for identifying
that clonality is achieved. This technique of variants with higher antibody avidity (89
biological cloning is in many ways superior 91). One important limitation in the ability
to limiting dilution methods but requires to use flow cytometric sorting for isolating
access to an expensive instrument, which antigen-specific hybridomas is the differential
may not be available. For those with access expression levels of native surface antibodies.
to cell sorting, the added expense of using The B cell receptor is not expressed at high
the instrument is often offset by saving time levels on the surface of all hybridomas, making
and resources needed to perform multiple it difficult to selectively sort antigen-specific
rounds of limiting dilution cloning. By stain- hybridomas after fusion. For this reason, sev-
ing for viability and using forward and side eral strategies have been developed to sort
scatter to eliminate doublets and clumps, a cells for antigen specificity or level of produc-
single cell can be accurately and consistently tion by employing the secreted antibody.
placed into culture medium, even within a One method uses an artificial affinity matrix
384-well plate platform. This approach results to capture antibody, which has been secreted
in a considerable increase in throughput, from the cell and is directly retained on
and thus the quantity of hybridomas that can the surface of the cell (92). The secreted
152 SMITH AND CROWE

molecules are bound to the secreting cell and fully automated manner, high-producing clones
can be subsequently labeled for flow can be obtained in many cases by means of a
cytometric analysis. This method is accom- single culture step following fusion. A com-
plished by first directly biotinylating the plete hybridoma generation procedure that
secreting cells, adding an avidin conjugated incorporates methylcellulose embedding,
capture antibody, and then isolating cells fluorescent antigen labeling, and clone pick-
within a protein-impermeable matrix to pre- ing was described recently (99).
vent cross-contamination of the secreted
protein. The captured antibody then can be
labeled fluorescently. This method has been ACKNOWLEDGMENT
used successfully in the isolation of hybrid- Conflicts of interest: We disclose no conflicts.
oma cells by flow cytometric sorting (93).
A related method, which is commercially
available for use, is the gel microdroplet CITATION
encapsulation technique. This strategy uses Smith SA, Crowe JE, Jr. 2015. Use of human
an agarose droplet to encapsulate the secret- hybridoma technology to isolate human
ing cells. The agarose matrix itself is bio- monoclonal antibodies. Microbiol Spectrum
tinylated so that an avidin bridge forms with 3(1):AID-0027-2014.
biotinylated anti-immunoglobulin antibody,
which can capture the antibody secreted
from the cell (94, 95). This technique has REFERENCES
been employed successfully to use flow cyto- 1. Kohler G, Milstein C. 1975. Continuous cultures
metric sorting and sort hybridomas based on of fused cells secreting antibody of predefined
their secreted mAb (9597). The agarose specificity. Nature 256:495497.
2. Schwaber J, Cohen EP. 1973. Human x mouse
matrix is able to prevent secreted antibody
somatic cell hybrid clone secreting immu-
from cross-contaminating neighboring encap- noglobulins of both parental types. Nature
sulated cells. Unlike the direct biotinylation 244:444447.
method described previously, the droplet 3. Bloom AD, Nakamura FT. 1974. Establish-
encapsulation method requires removal of the ment of a tetraploid, immunoglobulin produc-
agarose matrix after the cell(s) is/are selected. ing cell line from the hybridization of two
human lymphocyte lines. Proc Natl Acad Sci
Unfortunately, both methods are challenging USA 71:26892692.
and require considerable optimization. 4. Olsson L, Kaplan HS. 1980. Humanhuman
Another method used for biological clon- hybridomas producing monoclonal antibodies
ing and growth of hybridomas involves of predefined antigenic specificity. Proc Natl
embedding in semisolid medium. When cells Acad Sci USA 77:54295431.
5. Stevens RH, Macy E, Morrow C, Saxon
are plated, often following fusion, in semisolid A. 1979. Characterization of a circulating
medium containing methylcellulose and the subpopulation of spontaneous anti-tetanus
HAT selection components, hybridomas grow toxoid antibody producing B cells following
in suspended isolated colonies (83, 98). Hy- in vivo booster immunization. J Immunol 122:
24982504.
bridoma colonies can be picked into growth
6. Casali P, Inghirami G, Nakamura M, Davies
medium with a high probability of being TF, Notkins AL. 1986. Human monoclonals
clonal. Building on this method, manufacturers from antigen-specific selection of B lympho-
have developed instruments (for example, cytes and transformation by EBV. Science
ClonePix [Molecular Devices LLC]) that iso- 234:476479.
late, detect, and measure relative secretion of 7. Kozbor D, Roder JC. 1981. Requirements for
the establishment of high titered human
monoclonal antibody from hybridomas. With monoclonal antibodies against tetanus toxoid
methylcellulose semisolid HAT medium and using the Epstein-Barr virus technique. J
the ability to pick fluorescent colonies in a Immunol 127:12751280.
CHAPTER 8 Use of Hybridoma Technology To Isolate Human mAbs 153

8. Crawford DH, Ando I. 1986. EB virus induc- proliferation and differentiation of human B
tion is associated with B-cell maturation. cells activated by the CD40 ligand. Int Immunol
Immunology 59:405409. 5:657663.
9. Roder JC, Cole SP, Kozbor D. 1986. The EBV- 21. Orscheschek K, Merz H, Schlegelberger B,
hybridoma technique. Methods Enzymol 121: Feller AC. 1994. An immortalized cell line with
140167. features of human follicular dendritic cells.
10. Steinitz M, Koskimies S, Klein G, Makela O. Antigen and cytokine expression analysis. Eur J
1978. Establishment of specific antibody pro- Immunol 24:26822690.
ducing human lines by antigen preselection 22. Nilsson K, Sundstrm C. 1974. Establishment
and EBV transformation. Curr Top Microbiol and characteristics of two unique cell lines from
Immunol 81:156163. patients with lymphosarcoma. Int J Cancer
11. Bernasconi NL, Traggiai E, Lanzavecchia A. 13:808823.
2002. Maintenance of serological memory by 23. Zubler RH, Erard F, Lees RK, Van Laer M,
polyclonal activation of human memory B Mingari C, Moretta L, MacDonald HR. 1985.
cells. Science 298:21992202. Mutant EL-4 thymoma cells polyclonally acti-
12. Hartmann G, Krieg AM. 2000. Mechanism vate murine and human B cells via direct cell
and function of a newly identified CpG DNA interaction. J Immunol 134:36623668.
motif in human primary B cells. J Immunol 24. Moore PA, Belvedere O, Orr A, Pieri K, LaFleur
164:944953. DW, Feng P, Soppet D, Charters M, Gentz
13. Traggiai E, Becker S, Subbarao K, Kolesnikova L, R, Parmelee D, Li Y, Galperina O, Giri J,
Uematsu Y, Gismondo MR, Murphy BR, Rappuoli Roschke V, Nardelli B, Carrell J, Sosnovtseva
R, Lanzavecchia A. 2004. An efficient method S, Greenfield W, Ruben SM, Olsen HS, Fikes J,
to make human monoclonal antibodies from Hilbert DM. 1999. BLyS: member of the tumor
memory B cells: potent neutralization of SARS necrosis factor family and B lymphocyte stim-
coronavirus. Nat Med 10:871875. ulator. Science 285:260263.
14. Kozbor D, Roder JC, Chang TH, Steplewski 25. Gorny MK. 1994. Production of human mono-
Z, Koprowski H. 1982. Human anti-tetanus clonal antibodies via fusion of Epstein-Barr
toxoid monoclonal antibody secreted by EBV- virus-transformed lymphocytes with hetero-
transformed human B cells fused with murine myeloma, p 276281. In Celis JE (ed), Cell Biology:
myeloma. Hybridoma 1:323328. A Laboratory Handbook, 2nd ed. Academic Press,
15. Foung SK, Perkins S, Raubitschek A, Larrick San Diego, CA.
J, Lizak G, Fishwild D, Engleman EG, Grumet 26. Miller G, Lipman M. 1973. Release of infec-
FC. 1984. Rescue of human monoclonal antibody tious Epstein-Barr virus by transformed mar-
production from an EBV-transformed B cell moset leukocytes. Proc Natl Acad Sci USA
line by fusion to a human-mouse hybridoma. 70:190194.
J Immunol Methods 70:8390. 27. Wallace LE, Young LS, Rowe M, Rowe D,
16. Chiorazzi N, Wasserman RL, Kunkel HG. Rickinson AB. 1987. Epstein-Barr virus-specific
1982. Use of Epstein-Barr virus-transformed B T-cell recognition of B-cell transformants ex-
cell lines for the generation of immunoglobu- pressing different EBNA 2 antigens. Int J Cancer
lin-producing human B cell hybridomas. J Exp 39:373379.
Med 156:930935. 28. Nikitin PA, Yan CM, Forte E, Bocedi A,
17. Emanuel D, Gold J, Colacino J, Lopez C, Tourigny JP, White RE, Allday MJ, Patel A,
Hammerling U. 1984. A human monoclonal Dave SS, Kim W, Hu K, Guo J, Tainter D,
antibody to cytomegalovirus (CMV). J Immunol Rusyn E, Luftig MA. 2010. An ATM/Chk2-
133:22022205. mediated DNA damage-responsive signaling
18. Lagerkvist AC, Furebring C, Borrebaeck CA. pathway suppresses Epstein-Barr virus trans-
1995. Single, antigen-specific B cells used to formation of primary human B cells. Cell Host
generate Fab fragments using CD40-mediated Microbe 8:510522.
amplification or direct PCR cloning. Biotechniques 29. Smith SA, Zhou Y, Olivarez NP, Broadwater
18:862869. AH, de Silva AM, Crowe JE Jr. 2012. Persis-
19. Ding BB, Bi E, Chen H, Yu JJ, Ye BH. 2013. tence of circulating memory B cell clones with
IL-21 and CD40L synergistically promote potential for dengue virus disease enhance-
plasma cell differentiation through upregula- ment for decades following infection. J Virol
tion of Blimp-1 in human B cells. J Immunol 86:26652675.
190:18271836. 30. Okada Y. 1962. Analysis of giant polynuclear
20. Cocks BG, de Waal Malefyt R, Galizzi JP, de cell formation caused by HVJ virus from
Vries JE, Aversa G. 1993. IL-13 induces Ehrlichs ascites tumor cells. I. Microscopic
154 SMITH AND CROWE

observation of giant polynuclear cell forma- contact between cells after their electroper-
tion. Exp Cell Res 26:98128. meabilization. Biochem Biophys Res Commun 140:
31. Morgan C, Howe C. 1968. Structure and 258266.
development of viruses as observed in the elec- 46. Bardsley DW, Liddell JE, Coakley WT,
tron microscope. IX. Entry of parainfluenza I Clarke DJ. 1990. Electroacoustic production
(Sendai) virus. J Virol 2:11221132. of murine hybridomas. J Immunol Methods
32. Okada Y. 1993. Sendai virus-induced cell 129:4147.
fusion. Methods Enzymol 221:1841. 47. Neil GA, Zimmermann U. 1993. Electrofusion.
33. Nagata S, Yamamoto K, Ueno Y, Kurata T, Methods Enzymol 220:174196.
Chiba J. 1991. Production of monoclonal 48. Nilsson K, Bennich H, Johansson SG, Pontn
antibodies by the use of pH-dependent vesic- J. 1970. Established immunoglobulin produc-
ular stomatitis virus-mediated cell fusion. ing myeloma (IgE) and lymphoblastoid (IgG)
Hybridoma 10:317322. cell lines from an IgE myeloma patient. Clin
34. Nagata S, Yamamoto K, Ueno Y, Kurata T, Exp Immunol 7:477489.
Chiba J. 1991. Preferential generation of 49. Olsson L, Kaplan HS. 1983. Humanhuman
monoclonal IgG-producing hybridomas by monoclonal antibody-producing hybridomas:
use of vesicular stomatitis virus-mediated cell technical aspects. Methods Enzymol 92:316.
fusion. Hybridoma 10:369378. 50. Brodin T, Olsson L, Sjgren HO. 1983. Cloning of
35. Kao KN, Michayluk MR. 1974. A method for human hybridoma, myeloma and lymphoma cell
high-frequency intergeneric fusion of plant lines using enriched human monocytes as feeder
protoplasts. Planta 115:355367. layer. J Immunol Methods 60:17.
36. Lentz BR. 2007. PEG as a tool to gain insight 51. Cote RJ, Morrissey DM, Houghton AN,
into membrane fusion. Eur Biophys J 36:315326. Beattie EJ Jr, Oettgen HF, Old LJ. 1983.
37. Kerkis AY, Zhdanova NS. 1992. Formation Generation of human monoclonal antibodies
and ultrastructure of somatic cell hybrids. reactive with cellular antigens. Proc Natl Acad
Electron Microsc Rev 5:124. Sci USA 80:20262030.
38. Lane RD, Crissman RS, Lachman MF. 1984. 52. Olsson L. 1983. Monoclonal antibodies in
Comparison of polyethylene glycols as fusogens clinical immunobiology. Derivation, potential,
for producing lymphocyte-myeloma hybrids. and limitations. Allergy 38:145154.
J Immunol Methods 72:7176. 53. Sikora K, Alderson T, Ellis J, Phillips J,
39. Yu X, McGraw PA, House FS, Crowe JE Jr. Watson J. 1983. Human hybridomas from
2008. An optimized electrofusion-based pro- patients with malignant disease. Br J Cancer
tocol for generating virus-specific human 47:135145.
monoclonal antibodies. J Immunol Methods 54. Hibi N, Arii S, Iizumi T, Nemoto T, Chu
336:142151. TM. 1986. Human monoclonal antibody rec-
40. Rols MP, Teissie J. 1990. Electropermea- ognizing liver-type aldolase B. Biochem J 240:
bilization of mammalian cells. Quantitative anal- 847856.
ysis of the phenomenon. Biophys J 58:10891098. 55. Matsuoka Y, Moore GE, Yagi Y, Pressman D.
41. Lo MM, Tsong TY, Conrad MK, Strittmatter 1967. Production of free light chains of immu-
SM, Hester LD, Snyder SH. 1984. Monoclonal noglobulin by a hematopoietic cell line derived
antibody production by receptor-mediated from a patient with multiple myeloma. Proc
electrically induced cell fusion. Nature 310: Soc Exp Biol Med 125:12461250.
792794. 56. Pickering JW, Gelder FB. 1982. A human
42. Wojchowski DM, Sytkowski AJ. 1986. myeloma cell line that does not express immuno-
Hybridoma production by simplified avidin- globulin but yields a high frequency of antibody-
mediated electrofusion. J Immunol Methods secreting hybridomas. J Immunol 129:406412.
90:173177. 57. Kozbor D, Tripputi P, Roder JC, Croce CM.
43. Hewish DR, Werkmeister JA. 1989. The use 1984. A human hybrid myeloma for production
of an electroporation apparatus for the pro- of human monoclonal antibodies. J Immunol
duction of murine hybridomas. J Immunol 133:30013005.
Methods 120:285289. 58. Karpas A, Dremucheva A, Czepulkowski BH.
44. Bakker Schut TC, Kraan YM, Barlag W, 2001. A human myeloma cell line suitable for
de Leij L, de Grooth BG, Greve J. 1993. the generation of human monoclonal anti-
Selective electrofusion of conjugated cells in bodies. Proc Natl Acad Sci USA 98:17991804.
flow. Biophys J 65:568572. 59. Ostberg L, Pursch E. 1983. Human X (mouse
45. Teissie J, Rols MP. 1986. Fusion of mamma- X human) hybridomas stably producing human
lian cells in culture is obtained by creating the antibodies. Hybridoma 2:361367.
CHAPTER 8 Use of Hybridoma Technology To Isolate Human mAbs 155

60. Goldstein NI, Nagle R, Villar H, Hersh E, domain II of the envelope protein. MBio 4:
Fisher PB. 1990. Isolation and characteriza- e00873913. doi:10.1128/mBio.00873-13.
tion of a human monoclonal antibody which 70. Carroll WL, Thielemans K, Dilley J, Levy
reacts with breast and colorectal carcinoma. R. 1986. Mouse x human heterohybridomas
Anticancer Res 10:14911500. as fusion partners with human B cell tumors.
61. Freedman RS, Ioannides CG, Tomasovic B, J Immunol Methods 89:6172.
Patenia R, Zhang HZ, Liang JC, Edwards CL. 71. Carroll WL, Lowder JN, Streifer R, Warnke
1991. Development of a cell surface reacting R, Levy S, Levy R. 1986. Idiotype variant cell
human monoclonal antibody recognizing ovar- populations in patients with B cell lymphoma.
ian and certain other malignancies. Hybridoma J Exp Med 164:15661580.
10:2133. 72. Brown S, Dilley J, Levy R. 1980. Immunoglobulin
62. Teng NN, Lam KS, Calvo Riera F, Kaplan HS. secretion by mouse X human hybridomas: an
1983. Construction and testing of mouse approach for the production of anti-idiotype
human heteromyelomas for human monoclonal reagents useful in monitoring patients with B
antibody production. Proc Natl Acad Sci USA cell lymphoma. J Immunol 125:10371043.
80:73087312. 73. da Silva Cardoso M, Siemoneit K, Sturm D,
63. Gorny MK, Xu JY, Karwowska S, Buchbinder Krone C, Moradpour D, Kubanek B. 1998.
A, Zolla-Pazner S. 1993. Repertoire of neutral- Isolation and characterization of human mono-
izing human monoclonal antibodies specific for clonal antibodies against hepatitis C virus enve-
the V3 domain of HIV-1 gp 120. J Immunol 150: lope glycoproteins. J Med Virol 55:2834.
635643. 74. Ogura M, Morishima Y, Ohno R, Kato Y,
64. Gorny MK, Wang XH, Williams C, Volsky B, Hirabayashi N, Nagura H, Saito H. 1985.
Revesz K, Witover B, Burda S, Urbanski M, Establishment of a novel human megakaryo-
Nyambi P, Krachmarov C, Pinter A, Zolla- blastic leukemia cell line, MEG-01, with positive
Pazner S, Nadas A. 2009. Preferential use of Philadelphia chromosome. Blood 66:13841392.
the VH5-51 gene segment by the human 75. Kubota-Koketsu R, Mizuta H, Oshita M,
immune response to code for antibodies Ideno S, Yunoki M, Kuhara M, Yamamoto
against the V3 domain of HIV-1. Mol Immunol N, Okuno Y, Ikuta K. 2009. Broad neutralizing
46:917926. human monoclonal antibodies against influenza
65. Gorny MK. 2012. Human hybridoma technol- virus from vaccinated healthy donors. Biochem
ogy. Antibody Technol J 2:15. Biophys Res Commun 387:180185.
66. Posner MR, Elboim H, Santos D. 1987. The 76. Pan Y, Sasaki T, Kubota-Koketsu R, Inoue Y,
construction and use of a humanmouse mye- Yasugi M, Yamashita A, Ramadhany R, Arai
loma analogue suitable for the routine produc- Y, Du A, Boonsathorn N, Ibrahim MS,
tion of hybridomas secreting human monoclonal Daidoji T, Nakaya T, Ono K, Okuno Y,
antibodies. Hybridoma 6:611625. Ikuta K, Watanabe Y. 2014. Human mono-
67. Yu X, Tsibane T, McGraw PA, House FS, clonal antibodies derived from a patient infected
Keefer CJ, Hicar MD, Tumpey TM, Pappas with 2009 pandemic influenza A virus broadly
C, Perrone LA, Martinez O, Stevens J, cross-neutralize group 1 influenza viruses.
Wilson IA, Aguilar PV, Altschuler EL, Basler Biochem Biophys Res Commun 450:4248.
CF, Crowe JE Jr. 2008. Neutralizing anti- 77. Akapirat S, Avihingsanon A, Ananworanich
bodies derived from the B cells of 1918 influenza J, Schuetz A, Ramasoota P, Luplertlop N,
pandemic survivors. Nature 455:532536. Ono K, Ikuta K, Utachee P, Kameoka M,
68. Smith SA, de Alwis R, Kose N, Durbin AP, Leaungwutiwong P. 2013. Variables influenc-
Whitehead SS, de Silva AM, Crowe JE Jr. ing anti-human immunodeficiency virus type 1
2013. Human monoclonal antibodies derived neutralizing human monoclonal antibody
from memory B cells following live attenuated (NhMAb) production among infected Thais.
dengue virus vaccination or natural infection Southeast Asian J Trop Med Public Health
exhibit similar characteristics. J Infect Dis 207: 44:825841.
18981908. 78. Kalantarov GF, Rudchenko SA, Lobel L,
69. Smith SA, de Alwis AR, Kose N, Harris E, Trakht I. 2002. Development of a fusion
Ibarra KD, Kahle KM, Pfaff JM, Xiang X, partner cell line for efficient production of
Doranz BJ, de Silva AM, Austin SK, Sukupolvi- human monoclonal antibodies from peripheral
Petty S, Diamond MS, Crowe JE Jr. 2013. The blood lymphocytes. Hum Antibodies 11:8596.
potent and broadly neutralizing human dengue 79. Ki r m a n I , Ka l a nt ar o v GF , L o be l L I ,
virus-specific monoclonal antibody 1C19 reveals Hibshoosh H, Estabrook A, Canfield R,
a unique cross-reactive epitope on the bc loop of Trakht I. 2002. Isolation of native human
156 SMITH AND CROWE

monoclonal autoantibodies to breast cancer. 90. Dangl JL, Parks DR, Oi VT, Herzenberg LA.
Hybrid Hybridomics 21:405414. 1982. Rapid isolation of cloned isotype switch
80. Calvert AE, Kalantarov GF, Chang GJ, Trakht I, variants using fluorescence activated cell
Blair CD, Roehrig JT. 2011. Human monoclonal sorting. Cytometry 2:395401.
antibodies to West Nile virus identify epitopes on 91. Martel F, Bazin R, Verrette S, Lemieux R.
the prM protein. Virology 410:3037. 1988. Characterization of higher avidity mono-
81. Dessain SK, Adekar SP, Stevens JB, Carpenter clonal antibodies produced by murine B-cell
KA, Skorski ML, Barnoski BL, Goldsby RA, hybridoma variants selected for increased anti-
Weinberg RA. 2004. High efficiency creation of gen binding of membrane Ig. J Immunol 141:
human monoclonal antibody-producing hybridomas. 16241629.
J Immunol Methods 291:109122. 92. Manz R, Assenmacher M, Pflger E, Miltenyi
82. Kromenaker SJ, Srienc F. 1994. Stability of S, Radbruch A. 1995. Analysis and sorting of
producer hybridoma cell lines after cell sorting: live cells according to secreted molecules, relo-
a case study. Biotechnol Prog 10:299307. cated to a cell-surface affinity matrix. Proc Natl
83. Mann CJ. 2007. Rapid isolation of antigen- Acad Sci USA 92:19211925.
specific clones from hybridoma fusions. Nat 93. Holmes P, Al-Rubeai M. 1999. Improved cell
Methods 4:12. line development by a high throughput affinity
84. Haight FA. 1967. Handbook of the Poisson capture surface display technique to select for
Distribution. John Wiley & Sons, New York, NY. high secretors. J Immunol Methods 230:141147.
85. Coller HA, Coller BS. 1986. Poisson statistical 94. Weaver J, Williams G, Klibanov A, Demain
analysis of repetitive sub-cloning by the limit- A. 1988. Gel microdroplets: rapid detection
ing dilution technique as a way of assessing and enumeration of individual microorganisms
hybridoma monoclonality. Methods Enzymol by their metabolic activity. Nat Biotechnol 6:
121:412417. 10841089.
86. Underwood PA, Bean PA. 1987. Hazards of 95. Powell KT, Weaver JC. 1990. Gel micro-
the limiting dilution methods of cloning droplets and flow cytometry: rapid determination
hybridomas. J Immunol Methods 107:119128. of antibody secretion by individual cells within a
87. Brezinsky SCG, Chiang GG, Szilvasi A, cell population. Nat Biotechnol 8:333337.
Mohan S, Shapiro RI, MacLean A, Sisk W, 96. Kenney JS, Gray F, Ancel MH, Dunne JF.
Thill G. 2003. A simple method for enriching 1995. Production of monoclonal antibodies
populations of transfected CHO cells for cells using a secretion capture report web. Nat
of higher specific productivity. J Immunol Biotechnol 13:787790.
Methods 277:141155. 97. Gray F, Kenney JS, Dunne JF. 1995. Secretion
88. Parks DR, Bryan VM, Oi VT, Herzenberg capture and report web: use of affinity derivatised
LA. 1979. Antigen-specific identification and agarose microdroplets for the selection of hybrid-
cloning of hybridomas with a fluorescence- oma cells. J Immunol Methods 182:155163.
activated cell sorter. Proc Natl Acad Sci USA 98. Davis JM, Pennington JE, Kubler AM,
76:19621966. Conscience JF. 1982. A simple, single-step
89. Jantscheff P, Winkler L, Karawajew L, technique for selecting and cloning hybridomas
Kaiser G, Bttger V, Micheel B. 1993. Hybrid for the production of monoclonal antibodies.
hybridomas producing bispecific antibodies to Immunol Methods 50:161171.
CEA and peroxidase isolated by a combination 99. Yokoyama WM, Christensen M, Dos Santos
of HAT medium selection and fluorescence G, Miller D, Ho J, Wu T, Dziegelewski M,
activated cell sorting. J Immunol Methods 163: Neethling FA. 2013. Production of monoclonal
9197. antibodies. Curr Protoc Immunol 102:129.
Humanized Mice for Studying Human
Immune Responses and Generating
Human Monoclonal Antibodies

RAMESH AKKINA1
9
INTRODUCTION

The potential uses of human pathogen-specific antibodies are enormous in


terms of both diagnostics and therapeutics. Early applications used
polyclonal sera for prophylaxis and therapies, but problems such as allergic
reactions, cost, and difficulty in their generation have led to the use of
mouse-derived monoclonal antibodies that were humanized by various
methods (1). These methods involved substituting part or all of the murine
antibody backbone with its human equivalent to derive chimeric or fully
humanized antibodies. Less labor-intensive methods used transgenic mice
harboring human immunoglobulin genes for immunization to derive human
antibodies (2). While this has hastened human antibody generation, some
limitations exist, such as differences in the maturation processes between
the mouse B cells expressing human antibodies and human B cells secreting
human antibodies. Therefore, an ideal way to produce authentic affinity-
matured human antibodies is to identify and harness the specific antibody-
producing human B-cell clones themselves. Conventional methods involved
immortalizing antigen-specific B cells from individuals who either recov-
ered from a disease or were vaccinated with a desired antigen to derive
stable antibody-producing cells. Alternatively, more recent high-throughput

1
Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0003-2012

157
158 AKKINA

methods involved rescuing the specific NK cells and other innate immune defects that
antibody genes from either specific plasma permit higher levels of human cell and tissue
cells or memory B cells (3). While these engraftments but were still less than ideal.
methods have now become routine, they A major advancement was the targeted dis-
both require collecting B cells from suitable ruption of the interleukin-2 (IL-2) receptor
human subjects. In addition to the paucity common gamma chain (IL2-Rcg) coding for
of specific pathogen-exposed human sub- the common and essential signaling com-
jects when needed and the existence of low ponent for the action of cytokines IL-2, IL-4,
numbers of antigen-specific cells, there are IL-7, IL-9, IL-15, and IL-21 (7, 8). Impairment
other practical and ethical considerations. of IL-7 and IL-15 signaling blocks native
One such consideration is the derivation of mouse NK-cell development, thus permitting
antibodies against dangerous pathogens such enhanced human cell engraftment. This muta-
as Ebola virus. These limitations pointed out tion, when introduced together with the SCID,
the need for a more practical experimental NOD, RAG1, or RAG2 gene mutations in
system that permits isolation of large quanti- different combinations by selective breeding,
ties of antigen-specific B cells against any yielded a number of new-generation severely
pathogen or antigen of interest. In this regard, immunocompromised recipient mice for far
newer-generation humanized mice harboring superior human cell engraftment (6). These
a transplanted human immune system with a include Rag2/ cg/, Rag1/ cg/ (RG),
capacity to yield antigen-specific B and plasma NOD/shi-scid/cg/ null (NOG), and NOD/
cells are expected to fill this need (4, 5, 6) and SCID/cg/ (NSG) mice. Ongoing efforts are
are discussed here. being directed toward introducing HLA class I
and II human immune system and cytokine
genes to generate new transgenic mice to per-
IMMUNODEFICIENT MOUSE STRAINS mit more robust human antibody and cellular
FOR HUMAN CELL RECONSTITUTION immune responses (see below).

A common denominator for all humanized


immune system mouse models is the trans- HUMANIZED MOUSE MODELS WITH
plantation of mature or progenitor human HUMAN IMMUNE SYSTEM
hematopoietic cells by various routes into
immunodeficient mice receptive to xenografts A variety of Hu mice are made by using dif-
without graft rejection. In this regard, there ferent strains of immunodeficient mice as well
has been a gradual evolution and improve- as by using different human cells and tissues.
ments in the generation of immunodeficient These are summarized in Table 1 and a broad
mouse strains (7). Early strains such as nude description is provided below.
mice, while lacking T cells and thus having
defects in T-cell responses, still harbored
Hu-PBL Mice
mouse B cells and NK cells and therefore
were not permissive for human cell reconsti- Hu-PBL mice are generated by the trans-
tution. The availability of severe combined plantation of human mature peripheral
immunodeficiency (SCID) mice that lacked blood mononuclear cells (PBMCs) via the
both T and B cells allowed human cell intraperitoneal (i.p.) route into SCID mice (9).
reconstitution and led to the creation of hu- More recent versions use either NSG or RG
peripheral blood lymphocyte (PBL)-SCID and mice. The transplanted human immune cells
SCID-hu mouse models (see below). Later persist for several weeks and show effector
improvements gave rise to nonobese diabetic functions. These mice can be productively
(NOD)-SCID mice that have a lower level of infected by viruses such as HIV-1 that target
CHAPTER 9 Hu Mice for Studying Immune Responses 159

TABLE 1 Summary of current Hu mouse models


Model Generation/mice used Advantages Disadvantages

Hu-PBL i.p. injection of human PBMC. Easy to prepare. Immediate use. No multilineage hematopoiesis.
SCID, NOD-SCID, NSG, NOG. Good T-cell engraftment. No primary immune response.
Graft versus host disease.
Hu-HSC Intrahepatic injection of CD34+ Easy to prepare. Multilineage No human HLA restriction.
HSC into neonates. Intravenous hematopoiesis. Primary immune
injection of CD34+ HSC into adults. response.
Rag2/ c/, NSG, NOG.
SCID-hu Coimplantation of human fetal Abundant T-cell lymphopoiesis. Surgery needed. Requires human
liver and thymic fragments under fetal tissue. No multilineage
kidney capsule. SCID or NOD-SCID. hematopoiesis. No primary
immune response. Poor
peripheral T-cell engraftment.
BLT Coimplantation of human fetal Multilineage hematopoiesis. Surgery needed. Requires human
liver and thymic fragments under T-cell education in autologous fetal tissue.
kidney capsule with additional thymus. HLA restriction.
injection of autologous CD34+ HSC.
Rag2/ c/, NOD-SCID, NSG.

cells of the human hematopoietic system. Hu-HSC Mice


Human memory B cells continue to produce
This model involves transplantation of hema-
antibodies from previous antigen exposure;
topoietic stem cells (HSCs) into a variety of
however, there is no multilineage human hema-
immunodeficient mice and has evolved sub-
topoietic cell generation and consequently no
stantially over the years (7, 12, 13). Early
primary immune response. While graft versus
versions involved the injection of hematopoi-
host disease (GVHD) by the injected T cells
etic progenitor cells (CD34+ cells) (also termed
is a drawback, this model is ideal for studying
SCID repopulating cells) into conditioned
xeno-GVHD.
adult SCID mice by intravenous (i.v.) or
intrafemoral routes. While there was de novo
SCID-Hu MICE
lymphopoiesis, T-cell development was
Coimplantation of human fetal thymus and poor. Use of new-generation IL-2 cg/ that
liver (containing hematopoietic stem cells) encompasses RG, NOG, or NSG mice led to
fragments under the SCID mouse renal cap- better engraftment. There are two versions of
sule generate mice that harbor a functional these with important differences. One is the
human thymus (called the thy/liv organoid) injection of HSCs into adult irradiated NSG/
(10). These mice primarily produce human NOG mice. While multiple hematopoietic line-
thymocytes and naive T cells. These T cells ages are generated, there is a poor yield of T
predominantly reside in the thy/liv organoid, cells. The second is intrahepatic injection of
and there is poor peripheral T-cell circula- HSC into conditioned newborn RG, NSG, or
tion. Because of the insufficient generation NOG mice that results in superior human cell
of a full spectrum of immune cells, they lack engraftment and the generation of T cells, B
the capacity to generate a human immune cells, macrophages, NK cells, and dendritic
response. Nevertheless, these models have cells (6, 13). Infection of both versions of these
been instrumental in the study of some key mice with different pathogens or immuniza-
aspects of viral pathogenesis with viruses such tion with different antigens gives rise to human
as HIV and HTLV and laid the foundation for immune responses (see below). There is also
the generation of new humanized mouse mucosal human cell engraftment permitting
models (11). HIV-1 infection by mucosal routes (14).
160 AKKINA

BLT Mice surgery is involved (13). Immunodeficient


mouse strains, namely RG, NOG, and NSG,
This model is a slight modification and con-
are commonly used (6, 20, 21, 22, 23). Injection
sequent improvement of the earlier SCID-hu
of HSC into neonatal mice versus adult mice
mouse model. The name derives from trans-
gives far superior engraftment with the gen-
plantation of bone marrow, liver, and thymus
eration of human immune-competent mice
(BLT); the main difference from SCID-hu
that harbor all four needed immune cell
mouse model is the additional transplantation
subsets, namely T cells, B cells, macrophages,
of autologous HSCs purified from fetal liver
and dendritic cells. Newborn mice, preferably
(13, 15, 16). The original BLT version used
within 3 days of birth, are injected with CD34+
NOD-SCID mice, whereas the new improved
HSCs from different sources that include cord
versions use NSG, NOG, or RG mice (17, 18).
blood, fetal liver, or human bone marrow. Of
Superior human cell engraftment with the
these, the efficiency and duration of engraft-
generation of T cells, B cells, macrophages,
ment appear to be better with the fetal liver-
NK cells, and dendritic cells is seen. The
derived CD34+ cells because of their more
presence of autologous human thymus per-
primitive lineage status in development. The
mits appropriate T-cell education and human
following protocol is routinely used in our
T-cell restriction.
laboratory and yields well-engrafted mice.
Single-cell suspension is prepared from frag-
Hu-Liver-HSC Mice ments of human fetal liver (16 to 20 weeks
gestation) by enzymatic digestion with colla-
The models described above are restricted to
genase, DNase, and hyaluronidase. The cells
human immune system transplantation with
are incubated with CD34 antibody and later
the generation of human immune cell subsets.
subjected to immunomagnetic bead-based
As noted, human pathogens that infect the
positive section. The purity of CD34+ cells
human hematopoietic system can be studied
generally ranges between 90% and 99% after
in addition to generating human immune
two successive cycles of selection. The purified
responses against a variety of antigens. To
cells are cultured overnight in a human
further broaden their application in infectious
cytokine media mix containing IL-3, IL-6,
disease research, a recent development is the
and stem cell factor (25 ng of each per ml).
derivation of Hu-HSC mice that permits
Preferably, fresh cells are injected into
infection with other human-specific patho-
neonates, although previously frozen cells
gens with a predilection to infect other human
can also be used. The neonatal mice are
organ systems such as the liver. Transgenic
irradiated at 350 rads 2 to 4 hours before cell
mice that simultaneously permit human
injections. We routinely use 5 105 human
hepatocyte and HSC engraftment were re-
fetal liver-derived CD34+ cells per mouse pup
cently developed (19). This permitted creation
to ensure consistent engraftment, although
of Hu-liver-HSC mice susceptible to the hepa-
lower numbers of cells can be used. Cells are
titis C virus and with a capacity for human
injected via insulin syringe in a 25-ml volume
immune response.
intrahepatically by visualizing the dark area
occupied by the liver under the relatively
OVERVIEW OF Hu MOUSE transparent skin (Fig. 2). Postinjection, pups
PREPARATION, INFECTION, are returned to their mothers and weaned
AND IMMUNIZATION 3 weeks later. Mice are housed in BSL-2
conditions. The engrafted mice are screened
A general outline of Hu mouse preparations to determine the levels of human CD45+ cells
is depicted in Fig. 1. Preparation of Hu-HSC in peripheral blood at approximately 12 weeks
mice is not technically intensive since no of age. In general, we obtain mice with 40%
CHAPTER 9 Hu Mice for Studying Immune Responses 161

FIGURE 1 Schematic for generation of Hu-HSC RG (Rag-hu) and BLT mice. doi:10.1128/microbiolspec.AID-
0003-2012.f1.

to 90% human cell engraftment. Human cell use of a trocar. Later, each of the mice is
engraftment is seen in primary and secondary injected (i.v., tail route) with autologous 2.5
lymphoid organs. In addition, mucosal engraft- 105 CD34+ HSC purified from the remaining
ment with human cells is also seen in the fetal liver. The reconstituted mice are evalu-
female reproductive tract as well as in the gut, ated for human cell engraftment at 12 to16
permitting HIV-1 mucosal transmission. In weeks posttransplantation before the use for
general, 20 to 30 Hu-HSC mice can be various experiments. In general, 15 to 20 mice
made with a typical batch of fetal liver derived can be made with a typical set of fetal tissues.
CD34+ cells. While different human pathogens and
Original preparations of BLT mice used different immunogens are studied in Hu
NOD-SCID mice, and these have been effec- mice based on the need of the investigator and
tively used in many experimental settings the types of human tissues transplanted, the
including HIV-1 mucosal viral transmission following general scheme provides a broad
(15, 16, 24). Owing to far superior engraft- overview of these protocols. Viral pathogens
ment, more recent protocols use NSG mice such as HIV-1, dengue, and Epstein-Barr virus
(17, 25, 26). Male mice are preferred for (EBV) have been studied more frequently to
surgical tissue implantation because of their date in these new models. With HIV-1, the
larger kidney size. Adult mice are conditioned most common route of experimental infection
by sublethal whole body irradiation at 325 rads is via the i.p. route for pathogenesis and ther-
before transplantation. Human fetal thymic apeutic studies. A typical infection involves
and liver tissues are dissected into 1-mm either the CCR5-tropic viral strain BaL or the
fragments and introduced together under the CXCR4-tropic strain NL4-3. Injection with
left kidney capsule of anesthetized mice by the 1 105 IU gives rise to viremia within a week
162 AKKINA

protocol using BLT mice, recombinant HIV-1


envelope or WNV-E protein were mixed with
a synthetic adjuvant IC31 and injected i.m. in
both quadriceps muscles (17). Three doses were
given on days 0, 21, and 45, and mice were
monitored for immune responses for 90 days.

HUMAN CELL RECONSTITUTION AND


ANTIBODY RESPONSES IN Hu MICE

A number of reports documented the gen-


eration of antigen-specific human antibody
responses in Hu-HSC mice (29, 30, 31, 32,
33, 34, 35, 36). Both IgM and IgG responses
were reported by different investigators,
although, in general, IgG responses were
FIGURE 2 Intrahepatic injection of CD34+ hema- found to be weak. Human antibody repertoire
topoietic stem cells (HSCs) into newborn immu- in Hu-HSC RG mice by analysis of the length
nodecient RG mouse. doi:10.1128/microbiolspec. of the CDR3 hypervariable regions revealed
AID-0003-2012.f2.
that the IgM B-cell repertoire was similar
to that seen in normal healthy individuals,
and persistent life-long infection, based on thus indicating no obvious limitations to
the maintenance of human cell engraftment generate a wide spectrum of human anti-
that in turn depends on the quality of the bodies of various specificities (21). However,
HSC injected (22, 27). For dengue viral infec- immunizations of Hu-HSC RG mice with TT
tion, the viral inoculum is delivered by i.p., and HBV vaccines gave a predominantly IgM
subcutaneous, or intradermal routes (28, 29). response with limited antigen-specific IgG
Infected mice develop acute viremia generally production, indicating a general failure to
lasting for 3 weeks. With EBV 1 105 to 1 106 class switch. The paucity of antigen-specific
RIU are injected i.p., and immune responses IgG is puzzling given that these mice do
are assayed at 4 to 10 weeks. With HIV-1, accumulate total serum IgG efficiently. One
antibody responses are monitored at approx- study used PBMC transfected with a human
imately 4 to 6 weeks. IgM responses can T-cell receptor specific for influenza hemag-
be seen with dengue viral infection within 2 glutinin (HA) peptide and bearing an HLA
weeks at the earliest, whereas IgG responses matched with that of the transplanted HSC
take much longer, usually about 8 weeks. used to prepare the Hu mice. When these T
For experimental immunizations, a variety cells were transferred into the respective
of antigens have been used. These include mice, anti-HA human IgG response was
tetanus toxoid (TT), hepatitis B virus (HBV), observed, suggesting that isotype switch
HIV-1, and West Nile virus envelope (WNV-E) deficiency is not due to an intrinsic defect
antigens among others (7, 17, 21). With regard in the B cells but rather to impairment in T-
to human vaccine preparations, three doses cell cooperation (37). This impairment may
each of HBV or TT vaccine (each corre- be due to human T-cell restriction by murine
sponding to 1/10 the human dose) are given major histocompatibility complex (MHC) in
intramuscularly (i.m.) 2 weeks apart to Hu- addition to a potential T-cell dysfunction.
HSC RG mice (21), and antibody responses are When IgD+CD19+ naive B cells from Hu-HSC
evaluated starting 2 weeks later. In a recent mice were treated in vitro with anti-CD40
CHAPTER 9 Hu Mice for Studying Immune Responses 163

antibodies, IL-2, and IL-21 in the presence of A recent study evaluated BLT mice more
antigen, they became activated and secreted thoroughly to determine the antigen-specific
IgG, again confirming the functionality of the antibody responses by immunization with
B cells in these mice. In evaluating T-cell adjuvanted HIV-1 and WNV envelope anti-
help, another study using HLA-DR4 (MHC gens (17). Profound differences were noted
class II) transgenic mice and reconstitution both in terms of B-cell composition and
with matching HSC reported improved im- antibody responses in comparison with
mune responses that included IgG class healthy human immune responses. Even re-
switching and higher levels of IgG produc- peated booster immunizations did not result
tion (38). A more recent phenotypic analysis in secondary responses characterized by gen-
of B cells from Hu-HSC mice prepared eration of IgG. Unlike in the human, there
by using adult NSG mice indicated a normal was an abundance of a B-1 like B-cell
B-cell developmental pathway (39). How- population (CD19+ CD5+). The predominant
ever, molecular analysis of single B cells antibody response characterized by the IgM
indicated that, while the overall distribution phenotype and lack of IgG is attributed to
of Vh genes reflected a normal human anti- the CD5+ B-cell subset that is believed to
body repertoire, mature B-cell subsets showed produce natural antibody by using a T-cell-
autoimmune characteristics (39). Overall, independent pathway.
the wide variations seen in Hu-HSC mice
with regard to antibody production and class
switch could be attributed to a number of T-CELL RESPONSES AND HLA
factors. These include lack of proper human RESTRICTION IN HU MICE
T-cell restriction and help as well as differ-
ences in protocols, including utilizing neonatal Initial reports showed induction of antigen-
mice versus adult mice for HSC transplanta- specific T-cell responses in Hu-HSC mice
tion and HSC sources, namely cord blood and against various human pathogens (14, 42,
fetal liver. In any case, additional improve- 43). Immune control of HIV-1 and EBV
ments are necessary as discussed below. infection was abolished by the depletion of
SCID-hu mice have been further improved CD8 T cells in Hu-HSC mice, thus provid-
to generate BLT mice that provide a more ing additional indirect evidence for their
appropriate thymic microenvironment for role in protection (44, 45). However, other
human T-cell development and improved studies reported deficiencies in T-cell responses
T-B cell cooperation (15, 16). This resulted (37, 46, 47). While polyclonal stimulation of
in a more robust T-cell development and Hu-HSC mouse splenocytes by phytohemag-
multilineage hematopoiesis including the glutinin, anti-CD3/anti-CD28 antibodies, and
generation of B cells, macrophages, and den- phorbol myristate acetate/ionomycin lead to
dritic cells. The lineage-specific differentia- cell proliferation and cytokine secretion, they
tion, positive and negative selection of T cells were 10-fold less than what is seen with
is expected to occur in the autologous human human PBMC, suggesting a functional defect.
thymus. T cells in these mice were shown Moreover, human T cells responded poorly to
to generate MHC class I and II restricted in vivo immunizations as shown by the lack
human immune responses and offer T-cell of interferon-g or IL-4 secretion after specific
help to the antigen-stimulated B cells (16, 40). antigen restimulation ex vivo. Specific CD4
A number of early studies have shown both and CD8 responses were measured by cyto-
IgM and IgG antigen-specific responses, kine secretion assays, cell proliferation assays,
albeit with varied robustness. However, later or cytotoxic assays in vitro by restimulation
studies failed to show antibody class switching of cells from mice infected with either HIV-1
despite repeated booster immunizations (41). or EBV. Even in reports showing immune
164 AKKINA

activity, the responses are low and are attrib- GENERATION OF HUMAN MONOCLONAL
uted to several factors. The overall low level of ANTIBODIES IN Hu MICE
T cells is believed to result from a potential
lymphopenia-induced T-cell activation among Since humanized mice are shown to harbor
other causes. Another reason for low levels a normal human antibody repertoire, they
of T cells in Hu-HSC mice is thought to be a can be exploited to generate a broad spectrum
lack of HLA restriction, since T-cell selection of antibodies, both neutralizing and nonneu-
is happening in xenogenic mouse thymus in tralizing. The scope of their use is further
a H-2-restricted fashion, which might not broadened by the creation of new models,
be efficient for human cells. Furthermore, such as those harboring human liver, so that
the low numbers of T cells generated also agents like hepatitis viruses can also be used
display poor survival in the periphery owing to to generate specific antibodies. A general
their suboptimal interactions with mouse schematic summarizing different methods of
APCs and weak signaling (30). Supporting deriving human monoclonal antibodies in Hu
these possibilities, it was recently reported mice is presented in Fig. 3. The broad scheme
that when RG mice transgenic for HLA-DR4 would involve either infection or vaccina-
were reconstituted with matching HLA-DR4 tion with a desired antigen with or without
CD34 cord blood cells, the number of thymic an adjuvant. At the peak time of immune
and peripheral T cells was drastically increased. response, the splenocytes containing the B
Shultz et al generated class I HLA-A2 trans- cells are harvested for later selection, expan-
genic mice and reconstituted them with sion, and immortalization. Alternatively, anti-
matched HSC (6). These mice showed HLA gen-specific individual B cells can be sorted
restricted cellular immune responses to EBV. postimmunization, and their antibody genes
Based on these data, it has now become evi- can be cloned and later incorporated into an
dent that enforced expression of HLA-A2 and expression system to yield large quantities of
HLA-DR4 enables HLA-restricted T-cell antibodies. Furthermore, the specific antibody
functions which also correlate with improved gene sequences can also be class switched
cytokine secretion and IgG production. An- based on what type of antibody is desired for
other desirable alteration in these mouse therapeutic purposes by the use of molecular
models is to knock out the murine MHC techniques. Following some of these lines, one
expression such that unwanted H-2-restricted recent report described the entire process of
responses can be avoided by the transplanted generating human monoclonal antibodies by
human immune cells. T-cell responses in BLT using Hu-HSC RG mice (21). This is summa-
mice appear to be more normal because of rized in brief here as a basis for broader
reconstitution with autologous human thymus applications. Mice were immunized with
and HSC, thus permitting HLA restriction commercially available TT and HBc vaccines
of T-cell responses and more efficient T- and as described above. Postvaccination, memory
B-cell interactions and cooperation. HIV-1 B cells expressing surface immunoglobulins
infection in BLT mice demonstrated epitope- were fluorescence-activated cell sorter sorted
specific T-cell responses to HIV antigens in a from splenic and mesenteric lymph node
class I restricted manner in a recent study single-cell suspensions. The sorted B cells
(48). Anti-HIV-1 CD8 responses were found were immortalized by transduction with a
to mimic those in humans in terms of their retroviral vector encoding BCL6 and BCL-XL
specificity, kinetics, and immunodominance. genes and cultured in the presence of CD40L
Furthermore, it was found that mice express- and IL-2. Another novel method that can be
ing the particular HLA class allele HLA-B 57 used is STAT5 overexpression to immortalize
showed enhanced control of HIV infection as B cells (49). Supernatants from the immor-
seen in humans that bear the same allele. talized B-cell pools cultured in microtiter
CHAPTER 9 Hu Mice for Studying Immune Responses 165

FIGURE 3 Schematic for deriving human monoclonal antibodies using Hu mice. doi:10.1128/
microbiolspec.AID-0003-2012.f3.
166 AKKINA

plates were tested by enzyme-linked immuno- eration of an anti-Kaposis sarcoma-associated


sorbent assay to identify specific antibody- herpesvirus-antibody immune response (16, 43,
producing wells. Cells from the desired anti- 55, 56, 57). More recent studies have expanded
body-positive wells were subjected to limiting the use of Hu mice to other nonviral pathogens.
dilution culturing to obtain monoclonal B-cell These encompass work with drug-resistant
lines. In this study, only IgM antibody-secret- Salmonella enterica serovar Typhimurium
ing B-cell clones could be obtained, although (58, 59), persistent infection with the malaria
IgG responses were seen in the immunized parasite Plasmodium falciparum (60), and
mice, albeit at lower levels. Thus, further detection of Hu mouse adaptive and innate
improvements in the protocol are necessary immune responses against Leishmania (61).
to capture and immortalize these rare IgG- In addition to live pathogens, a number of
producing B cells. Other options available for antigens and human vaccine preparations have
efficient B-cell immortalization involve trans- also been tested to evaluate Hu mouse human
formation of antibody-producing B cells acti- immune responses. Immunization with DNP
vated by TLR9 agonists like CpG followed by (23)-KLH antigen generated human T-cell
high-efficiency electrofusion with myeloma proliferation and human IgG responses (40).
cells (50). Alternatively, using a molecular Toxic shock syndrome toxin 1 caused an
cloning approach, antigen-specific antibody expansion of human T cells and activation
genes can be rescued from the respective B of human dendritic cells (16). Administration
cells for high-efficiency expression in a surro- of a variety of vaccines demonstrated adap-
gate system, as has been accomplished with a tive immune responses including influenza-
number of antigens (51). specific human CD8+ T cells, human IgM
antibody responses to tetanus toxoid and
HBV (21, 62). This list is only a partial repre-
HUMAN PATHOGENS AND OTHER sentation of that reported in the current
ANTIGENS STUDIED IN Hu MICE literature which is expected to grow because
the use of these mouse models is expected
A variety of human pathogens, particularly to become more widespread.
viruses, have been studied in new-generation
Hu mice. HIV-1 is by far the most widely
studied pathogen (22, 36, 47). Hu mice have ADVANTAGES AND DISADVANTAGES
also been used to study HTLV-1 proviral OF DIFFERENT Hu MOUSE MODELS
integration and the induction of T-cell lym-
phomas (52, 53). Studies of other virus families From the standpoint of generating human
have also been gaining momentum. A number monoclonal antibodies, there are advantages
of studies focusing on dengue viral infection and disadvantages with each of the two new
showed viremia with concomitant humoral Hu-HSC and BLT mouse models. With regard
and cellular responses (28, 29). Hu mice with to the ease of preparation, the Hu-HSC model
human hepatocyte reconstitution allowed is relatively easy to create since only a quick
infection with hepatotropic viruses such as intrahepatic injection of HSC is needed and
hepatitis C virus and HBV inducing patholo- does not involve surgery. Additionally, human
gies and immune responses (19, 54). A variety HSC from easily procurable sources such as
of human herpes viruses have also been studied. cord blood can be used, and larger cohorts of
These reports documented HLA-restricted mice from a single donor can be made.
adaptive T-cell immune responses to EBV, However, a disadvantage with this model is
cytomegalovirus reactivation from latency, the lack of proper human T-cell restriction
protective innate and adaptive immune re- and ideal T- and B-cell cooperation because of
sponses against intravaginal HSV-2, and gen- the absence of an autologous human thymus.
CHAPTER 9 Hu Mice for Studying Immune Responses 167

Coexpression of HLA class I and II genes by tor remove xenografted human cells that do
transgenesis will overcome this deficiency. A not express the cognate ligand CD47 (dont
particular advantage with the BLT mouse eat me marker of self) (63, 64). In this context,
model is the presence of transplanted human it has been recently shown that human SIRPa
thymus, thus permitting proper T-cell educa- transgenic mice exhibit improved human
tion and T-cell restriction thus offering a cell reconstitution (65). Conversely, human
better T- and B-cell cooperation. Both IgM HSC stably transduced with murine CD47
and IgG responses are seen, although the IgG ligand encoding lentiviral vectors also showed
responses are found to be weaker, akin to that increased engraftment (66). Thus, improve-
seen with the hu-HSC model. Major disad- ment of constitutive CD47-SIRPa interactions
vantages are the complicated surgery required in Hu mice will enhance the survival of human
to implant human fetal tissues under the cells in the mouse environment and will
kidney capsule, and the number of mice that provide more robust and lasting human cell
can be generated from a single fetal donor engraftment. As mentioned above, the expres-
tissue is limited. Large-scale exploitation of sion of human HLA-DR4 in transgenic mice
this model also poses challenges owing to the also leads to increased T-cell numbers in
requirement for fetal tissues that are limited reconstituted mice. Coexpression of HLA class
in supply and often difficult to procure. I and class II in doubly transgenic mice is
likely to further improve human T-cell recon-
stitution and T-B cell cooperation, as well as
LIMITATIONS TO BE OVERCOME, help and mediate HLA-restricted immune
CURRENT ADVANCES, AND FUTURE responses (30). The knock-in of the human
PROSPECTS HLA class I and class II genes in mouse
MHC loci will preclude the unwanted mouse
While the current human immunocompe- H-2-restricted human cell immune responses.
tent mouse models have come a long way Since many murine cytokines and growth
since the description of the original human- factors are poorly cross-reactive with their
mouse chimeras, there are several limitations corresponding human cell receptors, thus
that need to be overcome (9). These encom- contributing to suboptimal human cell devel-
pass (i) residual innate immunity in the immu- opment and maintenance, supplying these
nodeficient mouse strains, thus requiring in trans either by injection or by transgenesis
irradiation and prior conditioning; (ii) less overcomes these deficiencies. These cytokines
than ideal T-cell numbers and lack of full include granulocyte-macrophage colony-stim-
maturation of B cells; (iii) lack of human HLA ulating factor (GM-CSF), IL-4, macrophage
class I and class II restriction in hu-HSC mouse colony-stimulating factor (M-CSF) for mono-
models and lack of appropriate levels of HLA cyte/macrophage, IL-7 for T cells, IL-15 for
APCs in the BLT mouse model; (iv) deficiencies NK cells, and erythropoietin for erythrocytes
in T- and B-cell cooperation resulting in low (67). Knock-in replacement of mouse cytokine
levels of antibody responses and inefficient genes with their human equivalents in respec-
immunoglobulin class switch; and, finally, (v) tive loci has an additional advantage of their
poorly cross-reactive native murine cytokines constitutive expression governed by the
and required growth factors. mouse regulatory elements. Indeed, with the
Some of these deficiencies are currently use of this approach, respective transgenic
being tackled and rectified as detailed below. mouse strains have been developed. Expres-
In addition to T cells, B cells, and NK cells, sion of human thrombopoietin resulted in
macrophages are also found to contribute higher human cell engraftment and better
to xenograft rejection in Hu mice. Mouse HSC maintenance (68). Transgenic mice
macrophages expressing native SIRPa recep- with human IL-3, GM-CSF (67), and M-CSF
168 AKKINA

(69) knock-in genes exhibited improved 4. Berges BK, Rowan MR. 2011. The utility of the
myeloid differentiation and function, thus new generation of humanized mice to study HIV-
1 infection: transmission, prevention, pathogen-
demonstrating the benefits of these enabling esis, and treatment. Retrovirology 8:65. doi:10.1186/
strategies. Thus, as can be seen from above, the 1742-4690-8-65.
current ongoing intensive work in different 5. Nischang M, Gers-Huber G, Audige A, Akkina
areas of generating Hu mice has identified R, Speck RF. 2012. Modeling HIV infection and
therapies in humanized mice. Swiss Med Wkly
several areas for improving the existing
142:w13618. doi:10.4414/smw.2012.13618.
human immunocompetent models. However, 6. Shultz LD, Brehm MA, Bavari S, Greiner DL.
these different strategies need to converge to 2011. Humanized mice as a preclinical tool for
yield a better mouse model. This will involve infectious disease and biomedical research. Ann
breeding a composite recipient mouse strain N Y Acad Sci 1245:5054.
incorporating all desirable attributes. Such a 7. Akkina R. 2013. Human immune responses and
potential for vaccine assessment in humanized
mouse should permit ideal engraftment of mice. Curr Opin Immunol 25:403409.
human HSC and give rise to a Hu mouse 8. Ito M, Kobayashi K, Nakahata T. 2008. NOD/
capable of a robust human antibody response Shi-scid IL2rgamma(null) (NOG) mice more
encompassing immunoglobulin class switch- appropriate for humanized mouse models.
ing and high-affinity maturation. Given the Curr Top Microbiol Immunol 324:5376.
9. Mosier DE. 2000. Human xenograft models
recent rapid progress, such an ideal system for virus infection. Virology 271:215219.
will soon be available. 10. McCune JM. 1996. Development and applications
of the SCID-hu mouse model. Semin Immunol
8:187196.
ACKNOWLEDGMENTS 11. Jamieson BD, Zack JA. 1999. Murine models
for HIV disease. AIDS 13(Suppl A):S5S11.
I thank members of my laboratory for their 12. Legrand N, Ploss A, Balling R, Becker PD,
contributions to Hu mouse research and Borsotti C, Brezillon N, Debarry J, de Jong
Jonathan LeCureux for assistance with the Y, Deng H, Di Santo JP, Eisenbarth S, Eynon
manuscript. Work in my laboratory is sup- E, Flavell RA, Guzman CA, Huntington ND,
ported by NIH grants AI073255, AI095101, Kremsdorf D, Manns MP, Manz MG,
Mention JJ, Ott M, Rathinam C, Rice CM,
AI100845, HL074704, HL94257, U54AI065357, Rongvaux A, Stevens S, Spits H, Strick-
and PO1AI099783. Marchand H, Takizawa H, van Lent AU,
Conflicts of interest: I declare no conflicts. Wang C, Weijer K, Willinger T, Ziegler P.
2009. Humanized mice for modeling human
infectious disease: challenges, progress, and
CITATION outlook. Cell Host Microbe 6:59.
13. Akkina R. 2013. New generation humanized
Akkina R. 2014. Humanized mice (Hu mice)
mice for virus research: comparative aspects and
for studying human immune responses and future prospects. Virology 435:1428.
generating human monoclonal antibodies. 14. Berges BK, Akkina SR, Folkvord JM, Connick
Microbiol Spectrum 2(2):AID-0003-2012. E, Akkina R. 2008. Mucosal transmission of R5
and X4 tropic HIV-1 via vaginal and rectal routes
in humanized Rag2/ gammac / (RAG-hu)
REFERENCES mice. Virology 373:342351.
1. Presta LG. 2008. Molecular engineering and 15. Lan P, Tonomura N, Shimizu A, Wang S,
design of therapeutic antibodies. Curr Opin Yang YG. 2006. Reconstitution of a functional
Immunol 20:460470. human immune system in immunodeficient mice
2. Lonberg N. 2005. Human antibodies from trans- through combined human fetal thymus/liver and
genic animals. Nat Biotechnol 23:11171125. CD34+ cell transplantation. Blood 108: 487492.
3. Jakobovits A, Amado RG, Yang X, Roskos L, 16. Melkus MW, Estes JD, Padgett-Thomas A,
Schwab G. 2007. From XenoMouse technology to Gatlin J, Denton PW, Othieno FA, Wege AK,
panitumumab, the first fully human antibody Haase AT, Garcia JV. 2006. Humanized mice
product from transgenic mice. Nat Biotechnol 25: mount specific adaptive and innate immune re-
11341143. sponses to EBV and TSST-1. Nat Med 12:13161322.
CHAPTER 9 Hu Mice for Studying Immune Responses 169

17. Biswas S, Chang H, Sarkis PT, Fikrig E, Zhu Q, 28. Jaiswal S, Pazoles P, Woda M, Shultz LD,
Marasco WA. 2011. Humoral immune responses Greiner DL, Brehm MA, Mathew A. 2012.
in humanized BLT mice immunized with West Enhanced humoral and HLA-A2-restricted den-
Nile virus and HIV-1 envelope proteins are largely gue virus-specific T-cell responses in humanized
mediated via human CD5+ B cells. Immunology BLT NSG mice. Immunology 136:334343.
134:419433. 29. Kuruvilla JG, Troyer RM, Devi S, Akkina R.
18. Stoddart CA, Maidji E, Galkina SA, Kosikova 2007. Dengue virus infection and immune re-
G, Rivera JM, Moreno ME, Sloan B, Joshi P, sponse in humanized RAG2/ gc/ (RAG-hu)
Long BR. 2011. Superior human leukocyte recon- mice. Virology 369:143152.
stitution and susceptibility to vaginal HIV trans- 30. Garcia S, Freitas AA. 2012. Humanized mice:
mission in humanized NOD-scid IL-2R g/ current states and perspectives. Immunol Lett
(NSG) BLT mice. Virology 417:154160. 146:17.
19. Washburn ML, Bility MT, Zhang L, Kovalev 31. Gorantla S, Sneller H, Walters L, Sharp JG,
GI, Buntzman A, Frelinger JA, Barry W, Pirruccello SJ, West JT, Wood C, Dewhurst
Ploss A, Rice CM, Su L. 2011. A humanized S, Gendelman HE, Poluektova L. 2007. Human
mouse model to study hepatitis C virus infection, immunodeficiency virus type 1 pathobiology
immune response, and liver disease. Gastroenter- studied in humanized BALB/c-Rag2 / g c /
ology 140:13341344. mice. J Virol 81:27002712.
20. Akkina R, Berges BK, Palmer BE, Remling L, 32. Ishikawa F, Yasukawa M, Lyons B, Yoshida
Neff CP, Kuruvilla J, Connick E, Folkvord J, S, Miyamoto T, Yoshimoto G, Watanabe T,
Gagliardi K, Kassu A, Akkina SR. 2011. Human- Akashi K, Shultz LD, Harada M. 2005.
ized Rag1/ gc/ mice support multilineage Development of functional human blood and
hematopoiesis and are susceptible to HIV-1 immune systems in NOD/SCID/IL2 receptor
infection via systemic and vaginal routes. PloS {gamma} chain(null) mice. Blood 106:15651573.
One 6:e20169. doi:10.1371/journal.pone. 0020169. 33. Matsumura T, Kametani Y, Ando K, Hirano
21. Becker PD, Legrand N, van Geelen CM, Y, Katano I, Ito R, Shiina M, Tsukamoto H,
Noerder M, Huntington ND, Lim A, Yasuda E, Saito Y, Tokuda Y, Kato S, Ito M, Motoyoshi
Diehl SA, Scheeren FA, Ott M, Weijer K, K, Habu S. 2003. Functional CD5+ B cells
Wedemeyer H, Di Santo JP, Beaumont T, develop predominantly in the spleen of NOD/
Guzman CA, Spits H. 2010. Generation of human SCID/gcnull (NOG) mice transplanted either
antigen-specific monoclonal IgM antibodies using with human umbilical cord blood, bone mar-
vaccinated human immune system mice. PloS row, or mobilized peripheral blood CD34+
One 5:e13137. doi:10.1371/journal.pone.0013137. cells. Exp Hematol 31:789797.
22. Berges BK, Wheat WH, Palmer BE, Connick 34. Shultz LD, Saito Y, Najima Y, Tanaka S, Ochi
E, Akkina R. 2006. HIV-1 infection and CD4 T T, Tomizawa M, Doi T, Sone A, Suzuki N,
cell depletion in the humanized Rag2/ gc/ Fujiwara H, Yasukawa M, Ishikawa F. 2010.
(RAG-hu) mouse model. Retrovirology 3:76. Generation of functional human T-cell subsets
doi:10.1186/1742-4690-3-76. with HLA-restricted immune responses in HLA
23. McDermott SP, Eppert K, Lechman ER, class I expressing NOD/SCID/IL2r gamma(null)
Doedens M, Dick JE. 2010. Comparison of humanized mice. Proc Natl Acad Sci USA
human cord blood engraftment between immu- 107:1302213027.
nocompromised mouse strains. Blood 116:193200. 35. Traggiai E, Chicha L, Mazzucchelli L, Bronz
24. Denton PW, Garcia JV. 2011. Humanized mouse L, Piffaretti JC, Lanzavecchia A, Manz MG.
models of HIV infection. AIDS Rev 13:135148. 2004. Development of a human adaptive immune
25. Denton PW, Olesen R, Choudhary SK, Archin system in cord blood cell-transplanted mice. Science
NM, Wahl A, Swanson MD, Chateau M, Nochi 304:104107.
T, Krisko JF, Spagnuolo RA, Margolis DM, 36. Watanabe S, Terashima K, Ohta S, Horibata
Garcia JV. 2012. Generation of HIV latency in S, Yajima M, Shiozawa Y, Dewan MZ, Yu Z,
humanized BLT mice. J Virol 86:630634. Ito M, Morio T, Shimizu N, Honda M,
26. Marsden MD, Kovochich M, Suree N, Shimizu Yamamoto N. 2007. Hematopoietic stem cell-
S, Mehta R, Cortado R, Bristol G, An DS, Zack engrafted NOD/SCID/IL2Rgnull mice develop
JA. 2012. HIV latency in the humanized BLT human lymphoid systems and induce long-lasting
mouse. J Virol 86:339347. HIV-1 infection with specific humoral immune
27. Berges BK, Akkina SR, Remling L, Akkina R. responses. Blood 109:212218.
2010. Humanized Rag2/ gc/ (RAG-hu) mice 37. Watanabe Y, Takahashi T, Okajima A,
can sustain long-term chronic HIV-1 infection Shiokawa M, Ishii N, Katano I, Ito R, Ito
lasting more than a year. Virology 397: 100103. M, Minegishi M, Minegishi N, Tsuchiya S,
170 AKKINA

Sugamura K. 2009. The analysis of the func- MO, Behnke S, Frey J, Oxenius A, Joller H,
tions of human B and T cells in humanized Aguzzi A, Manz MG, Speck RF. 2006. Dissem-
NOD/shi-scid/gc null (NOG) mice (hu-HSC inated and sustained HIV infection in CD34+ cord
NOG mice). Int Immunol 21:843858. blood cell-transplanted Rag2/g c/ mice. Proc
38. Danner R, Chaudhari SN, Rosenberger J, Natl Acad Sci USA 103:1595115956.
Surls J, Richie TL, Brumeanu TD, Casares S. 48. Dudek TE, No DC, Seung E, Vrbanac VD,
2011. Expression of HLA class II molecules Fadda L, Bhoumik P, Boutwell CL, Power
in humanized NOD.Rag1KO.IL2RgcKO mice is KA, Gladden AD, Battis L, Mellors EF, Tivey
critical for development and function of human TR, Gao X, Altfeld M, Luster AD, Tager AM,
T and B cells. PloS One 6:e19826. doi:10.1371/ Allen TM. 2012. Rapid evolution of HIV-1 to
journal.pone.0019826. functional CD8(+) T cell responses in humanized
39. Chang H, Biswas S, Tallarico AS, Sarkis PT, BLT mice. Sci Transl Med 4:143ra98. doi:10.1126/
Geng S, Panditrao MM, Zhu Q, Marasco WA. scitranslmed.3003984.
2012. Human B-cell ontogeny in humanized 49. Scheeren FA, van Geelen CM, Yasuda E,
NOD/SCID gcnull mice generates a diverse yet Spits H, Beaumont T. 2011. Antigen-specific
auto/poly- and HIV-1-reactive antibody reper- monoclonal antibodies isolated from B cells
toire. Genes Immun 13:399410. expressing constitutively active STAT5. PloS
40. Tonomura N, Habiro K, Shimizu A, Sykes M, One 6:e17189. doi:10.1371/journal.pone.0017189.
Yang YG. 2008. Antigen-specific human T-cell 50. Crowe JE Jr. 2009. Recent advances in the study
responses and T cell-dependent production of human antibody responses to influenza virus
of human antibodies in a humanized mouse using optimized human hybridoma approaches.
model. Blood 111:42934296. Vaccine 27(Suppl 6):G47G51.
41. Rajesh D, Zhou Y, Jankowska-Gan E, Roenneburg 51. Smith K, Garman L, Wrammert J, Zheng NY,
DA, Dart ML, Torrealba J, Burlingham WJ. Capra JD, Ahmed R, Wilson PC. 2009. Rapid
2010. Th1 and Th17 immunocompetence in generation of fully human monoclonal antibodies
humanized NOD/SCID/IL2rgammanull mice. specific to a vaccinating antigen. Nat Protoc
Hum Immunol 71:551559. 4:372384.
42. Hiramatsu H, Nishikomori R, Heike T, Ito 52. Banerjee P, Tripp A, Lairmore MD, Crawford
M, Kobayashi K, Katamura K, Nakahata T. L, Sieburg M, Ramos JC, Harrington W
2003. Complete reconstitution of human lym- Jr, Beilke MA, Feuer G. 2010. Adult T-cell
phocytes from cord blood CD34+ cells using the leukemia/lymphoma development in HTLV-
NOD/SCID/gammacnull mice model. Blood 1-infected humanized SCID mice. Blood 115:
102:873880. 26402648.
43. Strowig T, Gurer C, Ploss A, Liu YF, Arrey F, 53. Yamamoto I, Takajo I, Umeki K, Morishita
Sashihara J, Koo G, Rice CM, Young JW, K, Hatakeyama K, Kataoka H, Nomura H,
Chadburn A, Cohen JI, Munz C. 2009. Priming Okayama A. 2010. Multiple integrations of
of protective T cell responses against virus- human T-lymphotropic virus type 1 proviruses in
induced tumors in mice with human immune the engrafted cells from the asymptomatic carriers
system components. J Exp Med 206:14231434. in NOD/SCID/gammacnull mice. Intervirology
44. Gorantla S, Makarov E, Finke-Dwyer J, 53:229239.
Gebhart CL, Domm W, Dewhurst S, Gendelman 54. He Z, Zhang H, Zhang X, Xie D, Chen Y,
HE, Poluektova LY. 2010. CD8+ cell depletion Wangensteen KJ, Ekker SC, Firpo M, Liu C,
accelerates HIV-1 immunopathology in human- Xiang D, Zi X, Hui L, Yang G, Ding X, Hu Y,
ized mice. J Immunol 184:70827091. Wang X. 2010. Liver xeno-repopulation with
45. Yajima M, Imadome K, Nakagawa A, Watanabe human hepatocytes in Fah/Rag2/ mice after
S, Terashima K, Nakamura H, Ito M, Shimizu N, pharmacological immunosuppression. Am J
Yamamoto N, Fujiwara S. 2009. T cell-mediated Pathol 177:13111319.
control of Epstein-Barr virus infection in human- 55. Kwant-Mitchell A, Ashkar AA, Rosenthal
ized mice. J Infect Dis 200:16111615. KL. 2009. Mucosal innate and adaptive im-
46. An DS, Poon B, Ho Tsong Fang R, Weijer K, mune responses against herpes simplex virus
Blom B, Spits H, Chen IS, Uittenbogaart CH. type 2 in a humanized mouse model. J Virol
2007. Use of a novel chimeric mouse model with a 83:1066410676.
functionally active human immune system to 56. Parsons CH, Adang LA, Overdevest J, OConnor
study human immunodeficiency virus type 1 CM, Taylor JR Jr, Camerini D, Kedes DH. 2006.
infection. Clin Vaccine Immunol 14:391396. KSHV targets multiple leukocyte lineages during
47. Baenziger S, Tussiwand R, Schlaepfer E, long-term productive infection in NOD/SCID
Mazzucchelli L, Heikenwalder M, Kurrer mice. J Clin Invest 116:19631973.
CHAPTER 9 Hu Mice for Studying Immune Responses 171

57. Smith MS, Goldman DC, Bailey AS, Pfaffle xenograft rejection by macrophages. Proc Natl
DL, Kreklywich CN, Spencer DB, Othieno Acad Sci USA 104:50625066.
FA, Streblow DN, Garcia JV, Fleming WH, 64. van den Berg TK, van der Schoot CE. 2008.
Nelson JA. 2010. Granulocyte-colony stimu- Innate immune self recognition: a role for CD47-
lating factor reactivates human cytomegalovi- SIRPalpha interactions in hematopoietic stem
rus in a latently infected humanized mouse cell transplantation. Trends Immunol 29:203206.
model. Cell Host Microbe 8:284291. 65. Strowig T, Rongvaux A, Rathinam C, Takizawa
58. Firoz Mian M, Pek EA, Chenoweth MJ, H, Borsotti C, Philbrick W, Eynon EE, Manz
Ashkar AA. 2011. Humanized mice are sus- MG, Flavell RA. 2011. Transgenic expression of
ceptible to Salmonella typhi infection. Cell Mol human signal regulatory protein alpha in Rag2/
Immunol 8:8387. gamma(c)/ mice improves engraftment of
59. Libby SJ, Brehm MA, Greiner DL, Shultz LD, human hematopoietic cells in humanized mice.
McClelland M, Smith KD, Cookson BT, Proc Natl Acad Sci USA 108:1321813223.
Karlinsey JE, Kinkel TL, Porwollik S, Canals 66. Legrand N, Huntington ND, Nagasawa M,
R, Cummings LA, Fang FC. 2010. Humanized Bakker AQ, Schotte R, Strick-Marchand H,
nonobese diabetic-scidIL2rgnull mice are sus- de Geus SJ, Pouw SM, Bohne M, Voordouw
ceptible to lethal Salmonella Typhi infection. A, Weijer K, Di Santo JP, Spits H. 2011.
Proc Natl Acad Sci USA 107:1558915594. Functional CD47/signal regulatory protein alpha
60. Arnold L, Tyagi RK, Meija P, Swetman C, (SIRPa) interaction is required for optimal human
Gleeson J, Perignon JL, Druilhe P. 2011. T- and natural killer- (NK) cell homeostasis in
Further improvements of the P. falciparum vivo. Proc Natl Acad Sci USA 108:1322413229.
humanized mouse model. PloS One 6:e18045. 67. Willinger T, Rongvaux A, Strowig T, Manz MG,
doi:10.1371/journal.pone.0018045. Flavell RA. 2011. Improving human hemato-
61. Wege AK, Florian C, Ernst W, Zimara N, lymphoid-system mice by cytokine knock-in
Schleicher U, Hanses F, Schmid M, Ritter U. gene replacement. Trends Immunol 32:321327.
2012. Leishmania major infection in humanized 68. Rongvaux A, Willinger T, Takizawa H, Rathinam
mice induces systemic infection and provokes C, Auerbach W, Murphy AJ, Valenzuela DM,
a nonprotective human immune response. PLoS Yancopoulos GD, Eynon EE, Stevens S, Manz
Negl Trop Dis 6:e1741. doi:10.1371/journal.pntd. MG, Flavell RA. 2011. Human thrombopoietin
0001741. knockin mice efficiently support human hem-
62. Yu CI, Gallegos M, Marches F, Zurawski atopoiesis in vivo. Proc Natl Acad Sci USA 108:
G, Ramilo O, Garcia-Sastre A, Banchereau J, 23782383.
Palucka AK. 2008. Broad influenza-specific CD8+ 69. Rathinam C, Poueymirou WT, Rojas J,
T-cell responses in humanized mice vaccinated Murphy AJ, Valenzuela DM, Yancopoulos
with influenza virus vaccines. Blood 112:36713678. GD, Rongvaux A, Eynon EE, Manz MG, Flavell
63. Ide K, Wang H, Tahara H, Liu J, Wang X, RA. 2011. Efficient differentiation and function of
Asahara T, Sykes M, Yang YG, Ohdan H. human macrophages in humanized CSF-1 mice.
2007. Role for CD47-SIRPalpha signaling in Blood 118:31193128.
Antibodies: Computer-Aided
Prediction of Structure and
Design of Function

ALEXANDER M. SEVY1 and JENS MEILER1


10
INTRODUCTION

The central role antibodies play in our immune system makes them
important targets for computation-based structural modeling. Antibodies
consist of a constant and a variable region (Fig. 1). The constant region
is virtually identical in all antibodies of the same isotype, while the variable
region differs from one B-cell-derived antibody to the next. The variable
region of an antibody is the business end, the region that recognizes
its antigen via so-called complementarity-determining regions (CDRs).
Their large size (150 kDa) and inherent variability, in particular in the
CDRs, make antibodies a formidable challenge for molecular modeling.
Before we begin to model antibodies, it is useful to briefly review their
overall structure.

Relation of Antibody Sequence, Structure, and Function


The fundamental unit within an antibody is an immunoglobulin (IG)
domain of around 70 to 110 amino acids that adopts the characteristic IG
b-sandwich fold. Antibodies are homodimers of heterodimers, where each

1
Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37212.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0024-2014

173
174 SEVY AND MEILER

heterodimer consists of one heavy and one IG domains, the most N-terminal one being
light chain (Fig. 1) (1), each chain having variable, and all others constant. The two N-
multiple IG domains. A mammalian antibody terminal IG domains of the heavy chain-
light chain consists of two IG domains, the interact with the two IG domains of a light
C-terminal one called constant and the N- c h a i n t o f o r m h e t e r o d i me r s . T h e s e
terminal one, variable. The mammalian heterodimers homo-dimerize via the C-termi-
antibody heavy chain consists of four or five nal IG domains of the heavy chain to form

FIGURE 1 Challenges in antibody modeling. Though all antibodies share a common core structure (center
panel, PDB ID 1IGT [1]; heavy chains in magenta, light chains in yellow), slight differences in variable
regions and especially CDR loops can have a great effect on function. The vast sequence space generated
by genetic recombination in V, D, and J genes (A) results in many different CDR loop conformations.
Modeling of CDR loops from sequence information alone is a necessary computational task for accurate
structure prediction (B). The ability to simulate the afnity maturation process in silico is another
important task that can be used to generate an antibody with either increased higher afnity for its
native target, or for a completely novel target (C) (matured residues shown in cyan). Accurate antibody
modeling requires not only the ability to model an antibody alone, but also the ability to model its
interaction with a given antigen. Computational docking techniques achieve this by sampling different
positions of an antibody on its target to nd the most favorable position (D). doi:10.1128/microbiolspec.
AID-0024-2014.f1
CHAPTER 10 Computer-Aided Prediction of Structure 175

the final antibody. This domain arrangement be small compared to the total immune
ensures that the variable domains of heavy repertoire, leaving room for structure predic-
and light chains colocalize in space to form tion of important antibodies with unknown
the so-called paratope. structure. As antibody structures in the PDB
Each of these variable domains contains have increased exponentially in recent years
three CDRs that are the second (CDR1), (Fig. 2), computational biologists have gained
fourth (CDR2), and sixth (CDR3) loop a greater understanding of the molecular
regions of the b-sandwich, locating the CDRs determinants of proper loop folding and
on the same end of the IG fold. The amino antigen binding, ultimately allowing high-
acid sequence within the CDRs is determined throughput, accurate structural modeling on
by a process called somatic recombination, a scale infeasible for experimental methods
where an IG domain is assembled by combin- alone. Understanding the structural determin-
ing randomly chosen gene segmentsV and ants of antibody-antigen interaction (i.e., how
J for the light chain and V, D, and J for the the paratope engages the epitope) is critical for
heavy chain (Fig. 1A). This process generates a understanding antibody function and process-
large number of antibody sequences, as there es such as affinity maturation. As the number
are multiple copies of each gene type-somatic of cocrystal structures of antibody and antigen
recombination combined with junctional will always be small, computational docking
diversity at the joints of the gene segments algorithms can provide models for antibody-
creates a theoretical limit of around 1011 antigen complexes that are not experimentally
unique V(D)J sequences. In particular, CDR3 determined (Fig. 1D). Note that computational
of the heavy chain (HCDR3), encoded by structure prediction usually does not replace
the D-gene, is highly variable in length, the experiment but complements experimen-
structure, and dynamics (Fig. 1C). These tal data. For examples, starting from an exper-
germline antibodies are further modified in imental structure of antibody and antigen, their
a process called affinity maturation (Fig. 1D). interaction could be modeled; starting from
During B cell proliferation the genes encod- one antibody-antigen complex, models for
ing the variable domains experience an affinity-matured antibody-antigen complexes
increased rate of point mutation. This somat- can be constructed; or modeling can add atomic
ic hypermutation causes amino acid changes detail not present in lower-resolution electron
in the paratopes of daughter B cells, a pro- density maps obtained, for example, through
cess that allows tighter interaction with electron microscopy (EM). Computational
the epitope region of the antigen, i.e., aff- design of antibody-antigen complexes can be
inity maturation. used, for example, to study the process of
affinity maturation in silico or identify anti-
bodies with novel sequences not present or
Motivations for Antibody Modeling
not yet observed in nature (Fig. 1B). Compu-
The large number of theoretically possible tational antibody engineering is also applied to
antibodies and the large number of anti- humanize or stabilize therapeutic antibodies.
bodies actually present in humans prohibit
a comprehensive experimental character-
Challenges for Antibody Modeling
ization of antibody structure and dynamics.
While great progress has been made in Challenges when predicting the structures
antibody structure determination via crystal- of antibodies via comparative modeling in-
lization (currently around 2,000 depositions clude how changes in IG sequence change
in the Protein Data Bank [PDB] contain the the relative orientation of the two variable
phrase antibody), the number of experimen- IG domains in the complex and therefore
tal structures available in the PDB will always influence the paratope structure. Obviously,
176 SEVY AND MEILER

FIGURE 2 Exponential growth of structurally determined antibodies. Total antibody structures in the
Protein Data Bank (PDB) are shown by year. The increase in structures enables more accurate
computational approaches to antibody modeling and engineering. doi:10.1128/microbiolspec.AID-
0024-2014.f2

modeling the conformation of CDR loops is COMPARATIVE MODELING


a substantial challenge, in particular for CDR3, OF ANTIBODIES
which has an average length of around 15
amino acids in humans and can be as long as Given the large number of theoretically pos-
30 residues or morewell beyond the loop sible and actually existing antibodies, experi-
lengths tested in a typical loop construction mental structure determination will remain
benchmark (4, 8, and 12 amino acids). The reserved to a small fraction of particularly
plasticity of the paratope with the often important antibodies. Therefore, computa-
flexible CDRs presents a formidable challenge tional construction of a structural model is
to antibody/antigen docking simulations, as of central importance. A particular focus of
they require flexibility of both paratope and comparative modeling techniques is accurate
epitope, creating a huge conformational search modeling of the CDRs.
space. When engineering antibodies, these
challenges are multiplied by the enormous
Canonical Structure of CDRs
number of possible antibody sequences and
the resulting gigantic size of the sequence The concept of canonical structures of light
space that needs to be sampled, in addition to CDR (LCDR) and heavy CDR (HCDR) loops
the conformational space. These challenges can be traced back to seminal work by
result in another formidable motivation for Chothia, Thornton, Lesk, and others in the
modeling antibodies: the benchmarking of new 1980s and 1990s describing the conforma-
computational techniques. The challenges tional space sampled by all CDR loops
related to modeling antibodies combined with with known structure and linking these to
the availability of many experimental struc- conserved residues among these sequences
tures make antibody structure prediction and (24). Formally, the canonical structure hypo-
functional design an important playground to thesis states that the CDR loops of anti-
test new algorithms. bodies typically adopt one of a discrete set
CHAPTER 10 Computer-Aided Prediction of Structure 177

of conformations and that these conforma- accuracy in predicting the structure of non-
tions can be inferred from the amino acid HCDR3 loops based on their gene source and
sequence. This concept has been pursued to primary sequence, lending credibility to the
discretize the conformational space of CDRs use of cluster analysis in non-HCDR3 loop
and predict a loop conformation based on its modeling.
primary sequence. The set of canonical con-
formations has been relatively well defined for
Predicting the Conformation of HCDR3
all LCDR loops and for the heavy HCDR1 and
2 loops. However, the HCDR3 loop is by far Prediction of HCDR3 loop conformations
the most variable in sequence and length, presents a considerably larger challenge, as
drastically increasing the conformational they are much more structurally diverse
space it can sample. As the number of experi- and tend to be longer, ranging usually from
mentally determined antibody structures has 5 to 26 residues, with an average of 16 residues
exploded since the initial reports of canonical (6). In rare cases they can be substantially
structures, attempts to group CDR loops and longer. HCDR3 loops are generally divided
create a definitive set of loop conformations into torso and head regions for clustering
have continued to add new clusters to the purposes (7), and loops are characterized by
known set. While it is likely that this trend will either a bulged or extended beta-sheet con-
continue for some time, the fact that the formation in the torso region. Since HCDR3
number of clusters is still orders of magnitude loops cannot typically be placed into a confor-
lower than the known antibody repertoire mational cluster based on sequence alone,
validates the original canonical structure recent work has focused on developing a set
hypothesis by Chothia et al. for CDRs other of rules to predict certain aspects of the
than HCDR3 (2). conformation based on key residue positions
Studies of canonical non-HCDR3 loop (8). The more stable bulged conformation
conformations have focused on clustering tends to be preferred by HCDR3 loops,
known structures and deriving common stabilized by a hydrogen bond between a
characteristics in primary sequence to en- conserved tryptophan and a backbone car-
able a priori prediction of a loop confor- bonyl. One rule dictates that an aspartate
mation based on sequence alone. The number residue two positions upstream of the con-
of canonical loop conformations has increased served tryptophan is sufficient to displace this
along with experimental structures available hydrogen bond and results in a shift to the
for analysis; initial studies using only 17 struc- extended conformation. Another states that
tures identified 18 non-HCDR3 clusters (2), the position of a basic residue opposite this
whereas more recent studies using 1,200 aspartate dictates the formation of a bulged
structures have increased the number of non- or extended conformation. Taken together,
HCDR3 clusters to 72 (5). Clusters are iden- these rules are able to correctly predict torso
tified by their loop type and length, with conformation with 85% accuracy, compara-
the majority of non-HCDR3 loops falling ble to that of non-HCDR3 loops.
between 8 and 13 residues in length. Though Though the torso conformation of HCDR3
the number of non-HCDR3 loop clusters has loops can be predicted by sequence analysis,
increased with each subsequent analysis, the the antigen-binding head region of the loop
overall clustering pattern still maintains a remains intractable to clustering, leading to
high degree of uniformity and predictability. considerable efforts to use de novo modeling to
Figure 3 shows median loop structures of predict HCDR3 conformation. Currently, soft-
loops deriving from the largest cluster for each ware packages such as Rosetta (9) and Prime
of the CDR L1, L2, L3, H1, and H2 loops (5). (10) have been adapted to predict low-energy
Studies have consistently reported 85 to 90% loop conformations, either based on peptide
178 SEVY AND MEILER

FIGURE 3 Canonical CDR loop conformations. Pictured above are median loop structures representing
the largest cluster of (A) CDR L1, (B) L2, (C) L3, (D) H1, and (E) H2. Light and heavy chain loop variability
varies widely between the CDR loops, with heavy chain loops tending to be more variable. doi:10.1128/
microbiolspec.AID-0024-2014.f3

fragments gleaned from the PDB or generated current algorithms for de novo modeling
de novo. These protocols have achieved vary- remain insufficient to address a large propor-
ing degrees of success: HCDR3 loops shorter tion of important antibodies. However, the
than 12 to 14 residues can be consistently recent advances in predicting the conforma-
predicted within a reasonable margin of error tion of the HCDR3 base via clustering and
(2 ), while longer loops are less predictable improvements in de novo loop construction
and tend to have higher deviation. As the promise to enable more reliable HCDR3
average HCDR3 loop is 16 residues in length, prediction in the near future.
CHAPTER 10 Computer-Aided Prediction of Structure 179

Programs, Platforms, and Servers the highest sequence homology. The relative
Dedicated to Antibody Modeling orientation of the heavy and light chain V
domains has a significant impact on the
Because of the large number of antibody-
antigen-binding properties of an antibody
based therapeutics, there is substantial inter-
(13). Although the framework regions are
est in determining the structure of antibodies
well conserved, combining heavy and light
in a high-throughput, accurate manner. Exper-
chain frameworks from different templates
imental methods such as X-ray crystallogra-
results in just one and possibly incorrect
phy and nuclear magnetic resonance (NMR)
relative orientation of the heavy and light
spectroscopy, while highly informative, are
chain V domains, introducing error into the
labor-, time-, and resource-intensive. In addi-
predicted antibody structure. In addition,
tion, complete antibodies tend to be too large although LCDR loops and HCDR1 and 2
for NMR spectroscopy. X-ray crystallography, have canonical conformations, grafting these
in the absence of the antigen, often struggles to loops onto a disparate framework can result
determine the conformation of a long HCDR3 in errors in the relative placement of these
loop in the biologically relevant conforma- loops and their interactions. To address these
tion needed to engage the antigen. Sometimes problems, modeling servers have introduced
the HCDR3 loop can be locked into a differ- several different solutions. Some servers such
ent conformation by crystal packing or its as PIGS allow the user a great deal of input
coordinates cannot be determined because regarding the manner in which chimeras are
of flexibility. Therefore, many groups have built, allowing the user to tune these para-
worked on automated protocols to computa- meters for each model (14). The MOE and
tionally model antibody variable region struc- WAM modeling servers build an ensemble of
tures to predict the conformational space chimeras built from different templates and
including the conformation needed for bind- use force field energy minimization algorithms
ing. This has resulted in a number of publicly to relieve clashes and determine the most likely
accessible antibody-modeling servers, which conformation (15, 16). RosettaAntibody takes a
use a combination of comparative modeling, similar approach but uses a knowledge-based
de novo structure prediction, and energy min- potential rather than molecular mechanics
imization to generate an ensemble of potential force field to relieve clashes in framework
conformations. Recently, organized efforts and loop placements (17).
such as the Antibody Modeling Assessment Another major challenge is modeling the
(AMA) have focused on comparing these noncanonical HCDR3 loop. The sequence
modeling servers and determining the accu- and structural variability of this loop make
racy of antibody-modeling techniques relative it difficult to model it simply by homology
to one another (11, 12). in most cases. The PIGS server attempts
Antibody-modeling servers typically rely to model HCDR3 in the same manner as it
on comparative modeling to model frame- models the other loops, by sequence homol-
work regions and CDR loops, with the notable ogy to known HCDR3 structures (14). This
exception of HCDR3. Framework regions are approach can be effective in cases where
well conserved between antibodies, and a a similar HCDR3 exists in the PDB, and as
suitable template can usually be found more structures are added, this likelihood in-
among antibodies with experimentally deter- creases. The MOE server also grafts HCDR3
mined structures in the PDB. One concern loops from a template, followed by a more
with comparative modeling is the issue of complicated protocol of HCDR3 clustering,
generating chimeras, combining heavy and force field energy minimization to build an
light chain frameworks and CDR loops from ensemble of structures (16). However, the
different templates to use the template with variability of the HCDR3 loop is such that a
180 SEVY AND MEILER

more sophisticated modeling technique is nec- ANTIBODY DOCKING AND


essary in many cases. Other notable approaches EPITOPE MAPPING
for HCDR3 loop modeling involve either
fragment-based or de novo modeling. Rosetta Another significant challenge in modeling
Antibody uses a fragment-based approach, antibody-antigen complexes is docking the
pulling short peptide fragments from the antibody onto its epitope on the surface of
PDB and using robotics-based algorithms in the antigen. Though general protein-protein
conjunction with knowledge-based potentials docking has been successful in many cases,
to close the HCDR3 loop in an energetically antibody-antigen docking represents a special
favorable conformation (17). WAM uses a challenge among protein-protein docking cases.
similar approach, modeling based on PDB- Although useful in other protein-protein dock-
derived peptides based on different parameters ing problems, shape complementarity is not a
of the HCDR3, such as length and predicted good determinant of correct antibody place-
kinked conformation. However, the server ment, as epitopes and paratopes are typically
follows fragment assembly with a force field- flat. Binding affinity is instead determined
based minimization to achieve a local energy by hydrophobic hot spot and electrostatic
minimum (15). interactions (19, 20). Short, aggregation-prone
Since the introduction of these antibody- regions are common in many antibodies, con-
modeling servers, a standing question has sisting of mainly aromatic residues and con-
been which method is most effectivein centrated in HCDR2 loops, and can contribute
user friendliness, structural accuracy, and significantly to buried surface area in anti-
transparency of results. To this end, the body-antigen interactions (21). In addition,
Antibody Modeling Assessment has held a the model for the antibody used as a starting
biannual blinded study to give groups the point for docking might have to be altered
chance to model the structures of unpub- when the epitope is engaged: if a single con-
lished antigen-binding fragment (Fab) X- formation is used, conformation of CDR loops
ray structures to assess the state of the field. or the relative orientation of the V domains
The assessments in 2011 and 2014 involved might have to be changed when engaging the
blind prediction of 9 and 11 Fab fragments, epitope. If an ensemble of states is used, the
respectively, and analyzed the results by a best conformation needs to be selected and
number of parameters, including total root possibly refined further for optimal binding
mean square deviation (RMSD), loop and (22, 23). Additionally, if the location of the
framework-specific RMSD, and overall epitope is unknown, a global search needs to
structural integrity. Overall RMSD values be performed. This adds another degree of
for all servers are generally within 1.0 to complexity by requiring the docking algorithm
1.5 , with results varying between frame- to sample epitopes across the entire surface
work and CDR loops. Framework regions of the antigen. Several approaches have been
were generally predicted most accurately, implemented to address these challenges, such
with RMSD values consistently within 1.0 . as algorithms to remodel antibody loop con-
The highest deviation was seen for HCDR3 formations in the presence of the antigen and
loops, with RMSD values ranging from 0.5 application of experimental data to inform
to 8.0 and predictive ability depending the energy evaluation of docked models.
heavily on the details of individual Fab frag-
ments. Overall, the servers are comparable
Antibody Docking Algorithms
in their modeling accuracy, with variation
dependent on particulars of the Fab frag- One docking algorithm that has had success
ment and metric used to analyze accuracy in the realm of antibody-antigen docking
(11, 12, 18). is the RosettaDock algorithm, originally
CHAPTER 10 Computer-Aided Prediction of Structure 181

described by Gray et al. (24). The algorithm alternating low- and high-resolution pertur-
uses alternating rounds of low-resolution bation and minimization steps. However, it
rigid body perturbations and high-resolu- adds additional perturbation moves that are
tion side-chain and backbone minimization specific to antibody-binding motifs, such as
to generate a model of the docked complex. CDR loop remodeling and reorientation of the
The RosettaDock protocol relies on random angle between the V domains. This addresses
perturbation of the complex and creates the issue of antibody CDR loops adopting
large numbers (105) of models to capture alternate conformations in the bound and free
a global energy minimum. The original states, and the algorithm is designed to accom-
protocol was able to recover native confor- modate slight errors in comparative modeling
mations with an RMSD on the order of 5 to that may disrupt adoption of the native con-
10 for local searches, with a higher error for formation during docking. Although Snug-
global searches. Encouragingly, the antibody- Dock benchmarking shows similar results to
antigen docked complexes showed a strong RosettaDock when using the lowest energy
energy funnel, with low energy structures models for docking, ensemble docking using
corresponding to a low RMSD to the native SnugDock shows a marked improvement of
structure. This funnel validates the creation of the energy funnel and increase of high-
a large number of complexes, with confidence and medium-quality structures. This result is
that those with the lowest scores are most consistent with a conformational selection
likely to recover the native conformation. paradigm for the initial antibody-antigen
Since its original publication, the Rosetta- interaction with a subsequent induced fit.
Dock protocol has been benchmarked thor-
oughly with large numbers of both native and
Incorporation of Experimental Data
homology-modeled structures. Advances in
To Guide Antibody Docking and
comparative antibody-modeling capabilities
Epitope Mapping
beg the question of how well these compara-
tive models can be docking into their native The ability to identify a precise binding epi-
antigens to produce a native-like structure. In tope of an antibody with its corresponding
general, antibody-modeling servers provide antigen is of obvious use for any antibody
similar results in terms of success in docking being studied. The knowledge of a binding
comparative models. However, since scores epitope at the individual residue level allows
fail to correlate perfectly with RMSD values, more detailed analysis of the nature of an
with the lowest RMSD model frequently not antibodys interaction with its antigen, includ-
being the best scoring and vice versa, the best ing prediction of the mechanism of neutrali-
results have been obtained with ensemble zation of an antibody, escape mutations that
docking models, using multiple models from may evade binding, specificity of an antibody
the lower end of the energy landscape as in binding-related antigens, etc. However,
inputs for docking. Using such an ensemble epitope mapping is an area that has relied on
approach has been shown to flatten the energy low- and medium-throughput experimental
funnel and increase the proportion of models techniques, which can make it difficult to map
deemed high and medium quality (9). the epitopes of large numbers of antibodies.
Additional docking protocols have been pub- Experimental methods such as EM, NMR
lished based on the core algorithm in Rosetta- spectroscopy chemical shift mapping, competi-
Dock but tailored specifically to antibody-antigen tive enzyme-linked immunosorbent assays,
complexes. In particular, the SnugDock algo- site-directed mutagenesis, force spectroscopy,
rithm has been shown to increase docking and hydrogen-deuterium exchange have made
accuracy for antibody-antigen complexes (25). it possible to obtain structural information
This algorithm uses the same approach of on the antibody-antigen complex, including
182 SEVY AND MEILER

epitope and paratope, without the need for a the antibody is placed in the vicinity of
crystal structure (2631). Each technique faces the epitope to begin with. Such approaches
its own challenges that make high-throughput reduce the conformational space that needs to
epitope mapping difficult (32), but these data be sampled.
have been used successfully to create reliable In addition to reducing sampling space,
computational models. A comprehensive limited experimental data can also be used
example of integrating experimental data to improve scoring. One major issue with
into computational modeling is shown in docking using current methods is that a near-
Fig. 4: data from EM, hydrogen-deuterium native conformation is frequently present in
exchange, and site-directed mutagenesis were the large set of models, but the distinguish-
used to create a high-resolution model of ing power of the energy function is not
an antibody-antigen complex for an influenza sufficient to identify the near-native confor-
hemagglutinin directed antibody (33). mation without any a priori knowledge of the
Computational methods have also made structure. Low-resolution epitope mapping
great strides in recent years to enable the using experimental methods can provide an
large-scale, rapid mapping of binding epi- extra distinguishing feature to eliminate in-
tope. As with experimental methods, com- correct models and identify those that are
putational approaches come with their own most likely to adopt the native conformation.
disadvantages and cannot be used reliably Simonelli et al. have validated this hypothesis
in isolation. However, various hybrid meth- by using NMR chemical shift mapping to
ods that use different types of experimental identify an epitope before the start of compu-
data to reduce the complexity of computa- tational docking (26). Models that do not agree
tional searches have shown great promise in with the experimental results can be discarded,
providing a feasible approach to the epitope and lower scoring models are more likely to
mapping problem. A promising approach represent the native conformation.
to increase the accuracy of docking predic- Docking algorithms have been adapted
tions is the incorporation of experimental data to incorporate experimental data, which in-
to supplement in silico epitope prediction. creases the accuracy of the final docked
RosettaDock, in particular, has been bench- complex and the residues involved in the
marked on both local and global searches antibody-antigen interface. There are many
and predictably shows better results when different steps during the docking protocol in
perturbations are kept to a minimum and which experimental data can be incorporated.

FIGURE 4 Integrating experimental data to aid computational modeling. (A) Low-resolution cryo-EM
maps, (A and B) combined with hydrogen-deuterium exchange (DXMS) data and site-directed
mutagenesis, were used to generate a docked model of a potent anti-inuenza antibody. Reprinted
from Journal of Clinical Investigation (33) with permission from the publisher. doi:10.1128/microbiolspec.
AID-0024-2014.f4
CHAPTER 10 Computer-Aided Prediction of Structure 183

Mutagenesis data and cryo-EM density maps identifies pairs of residues within peptide
have been used to constrain the initial place- fragments that covary with the binding by
ment of the antibody on the surface of the the target antibody, allowing for identification
antigen prior to docking, allowing for a more of key residues both proximal and distal to
limited search and resulting in more native- the epitope. This method has been validated
like contacts (3335). In addition, complex using antibodies targeting viral protein gp120
scores can be weighted such that residues from HIV and is beneficial as a complement
experimentally shown to interact are encour- to docking. Another similar approach uses
aged to do so during the docking protocol neutralization data of an antibody with dif-
(34, 35). This approach drives the formation of ferent viral strains varying by sequence. This
models that contain the correct residue algorithm identifies mutations that tend to
pairwise interactions across the interface, correlate with a decrease in neutralization
while allowing for the creation of new inter- and proposes these as the binding epitope.
acting pairs within this framework. Final In addition, the algorithm can incorporate
models can then be clustered and analyzed structural data to eliminate purported epitope
visually for adherence to experimental data, residues on the basis of solvent-accessible
eliminating those that do not contain key surface area (40, 41). This method can be
residues at the interface or have topologies beneficial in cases where a large body of
inconsistent with EM data. neutralization data already exists in the liter-
Another source of experimental data that ature for an antibody, as extra experimental
has been used to improve computational data do not have to be collected solely for use
epitope prediction is hydrogen-deuterium by the algorithm.
exchange (36). This technique involves the
dissociation of amide hydrogens on a pro-
tein backbone and replacement with deute- ANTIBODY DESIGN
rium from a solvent. The extent of exchange
can then be quantified using NMR or mass The computational design of antibodies is not
spectrometry. Antibody binding to its antigen only the most stringent test of our under-
causes a decrease in solvent accessibility of standing of the rules that govern antibody
peptides at the epitope and a concomitant structure and interaction, but it also has
decrease in deuterium incorporation. Epitopes exciting applications in designing an anti-
of many different antibodies have been body optimized for a given epitope (affinity
mapped using this technique, with higher maturation) or an antibody that recognizes
throughput than other methods such as X-ray multiple similar target epitopes (broad neu-
crystallography or mutagenesis (27, 28, 37). tralization). Through this approach the rela-
Several studies have taken advantage of this tion between the sequence, structure, and
phenomenon to improve docking predictions activity of antibodies can be better understood,
by constraining the initial placement of the as the sequence and structural space can be
antibody and rewarding residue interactions explored in a more comprehensive manner
that agree with hydrogen-deuterium exchange than possible by analysis of naturally occur-
data (33, 38). ring antibodies only. Recently, an important
Other approaches to epitope mapping proof-of-principle experiment for computer-
have moved away from docking and instead aided epitope-focused vaccine design was
use bioinformatics strategies to identify reported (42). For this paradigm to reach its
interacting regions. One such approach uses full potential, knowledge of the optimal anti-
binding of randomly created peptide libraries bodies to engage an epitope and the relation
to determine motifs that are important for between sequence, structure, and activity
antibody recognition (39). The algorithm then inferred from computational design must be
184 SEVY AND MEILER

integrated. Besides the obvious application arbitrary number of binding partners (50, 51).
for the development of better therapeutic This technique has been applied to explore
antibodies, computational antibody engineer- the changes in antibody sequence and con-
ing also has the potential for formulation formation responsible for the shift from a
and humanization of therapeutic antibodies polyspecific, germline antibody to one with
(4346). higher affinity for a single target. In comple-
mentary work, Babor and Kortemme and
Willis et al. used multistate design to show
Broad Neutralization versus
that antibody germline sequences are optimal
Afnity Maturation
for conformational flexibility of both CDR
Affinity maturation is a process by which loops and framework residues, allowing the
the variable region of an antibody under- binding of multiple targets, whereas affinity-
goes somatic hypermutation to introduce matured antibodies have decreased flexibility
point mutations in the framework and CDR (52, 53). These authors have also identified
loops to select for a variant with increased the key residues responsible for either mono-
binding affinity for its target. Along with or polyspecificity for several commonly seen
V(D)J recombination and junctional diversity, germline genes. These studies validate the
it is a fundamental reason why a finite set biological relevance of design algorithms,
of antibody sequences is able to recognize a since sequences can be both computationally
virtually infinite array of antigens (47). Exper- matured and reverted to germline by using
imental methods can be used to re-create different sets of antigens as inputs.
maturation in an ex vivo context to create an
antibody with higher affinity for a given target.
In silico Afnity Maturation
For example, phage display combined with
random mutagenesis can screen for mutations The understanding of the nature of affinity
in a high-throughput manner to create new, maturation, as well as the ability to selec-
tighter-binding antibodies (48). This form of tively modify the specificity of antibody
directed evolution has been successfull in sequences, has led to advances in antibody
the maturation of many types of antibodies engineering that enable maturing antibodies
(48). Additionally, computational algorithms in silico to create new sequences with higher
have been developed that mimic the process of affinity. In one case, Clark et al. were able to
directed evolution to produce similar results. use computational design to mature an anti-
Computational methods have been successful body and generate candidate sequences with
in both studying the nature of affinity matu- higher predicted affinity (54). Using a combi-
ration in germline and matured antibodies nation of side chain repacking and electrostat-
and in further refining matured antibodies to ic optimization, a triple mutant was created
increase affinity even more. with 10 times higher affinity. A comparable
Affinity maturation can be simulated in increase in affinity was achieved by Lippow
silico to analyze compromises in an antibody et al. by redesigning an antilysozyme antibody
sequence that lead to a decrease in poly- along with the therapeutic antibody cetux-
specificity with a concomitant increase in imab (55). The design protocol was also able to
specificity for a single partner. For example, predict mutations in bevacizumab that had
a general computational method for the de- been previously shown to increase affinity.
sign of antibody CDR loops for targeted epi- The designed mutations primarily affected the
tope binding was introduced by Pantazes and electrostatic nature of the binding interface,
Maranas (49). Computational methods such as either by removing a poorly satisfied polar
multistate design are capable of determining residue at the interface or by adding a polar
the protein sequence optimal for binding an residue at the solvent-facing periphery of the
CHAPTER 10 Computer-Aided Prediction of Structure 185

interface (55). A similar approach has been Eliciting Neutralizing Antibodies


taken to increase the species cross-reactivity through Antigen Design
of an antibody, rather than increasing affinity
Although much focus has been directed
for a previously targeted antigen. By analyzing
at engineering antibodies with desired
sequence differences between two serine pro-
properties, recent work has targeted the
tease orthologs, Farady et al. created novel
opposite side of the problem: engineering
antibody designs by restricting the search
an antigen that can elicit a desired antibody
space to positions that contact points of dif-
in an effective and reproducible manner.
ference between orthologs (56). In this man-
This comes with the ultimate goal of the
ner they were able to target positions that
rational design of antigens to be used in
would be most likely to establish new contacts
vaccination that can elicit antibodies targeting
across the binding interface to enable interac-
a precise, conserved, and neutralizing epitope.
tion at a reasonable affinity. This method was
Giles et al. attempted to create a broadly
able to create antibody mutants with increases
reactive antigen by identifying divergent
in affinity of over two orders of magnitude.
sequences between different clades of influ-
One significant limitation of most com-
enza hemagglutinin and clustering the con-
putational design protocols is that they
sensus sequences. In this way they created a
require a high-resolution crystal structure
compromise antigen, incorporating elements
of the antibody-antigen complex, or alter-
of various clades designed to elicit antibodies
natively high-resolution structures of each
binding all of them (59). This strategy was
component separately. However, several anti-
validated with the finding that the broadly
body designs have been made for complexes
reactive antigen was capable of eliciting
that do not have a solved structure available,
greater antibody breadth than a polyvalent
using a combination of comparative modeling,
virus-like particle displaying the native anti-
protein-protein docking, and design. Barderas
gens (60). Wu et al. pursued a similar goal,
et al. used experimental epitope mapping data
instead using HIV surface protein gp120 as
to dock a comparative model of an antigastrin
the antigen. This group used computational
antibody onto the surface of its target (57).
design to engineer a modified gp120 that
They then used the docked models to estimate
maintained the structure of the neutralizing
regions of antibody-antigen interaction and
portion of the molecule while eliminating
created mutants using both phage display and
other antigenic regions that may elicit non-
in silico affinity maturation to mutagenize
neutralizing antibodies (61). Since the antigen-
antibody residues in contact with the antigen
ic region of gp120 they targeted was the
and produce designs with high predicted
CD4 binding site, a highly conserved region
affinity. In several cases the in silico suggested
among divergent HIV strains, they used this
mutations matched the mutations seen by
engineered antigen as a vaccine to elicit
directed evolution, and overall the designs
broadly neutralizing antibodies and success-
were able to increase affinity to nanomolar
fully identified two novel antibodies with a
levels. Another case used docking of an
high level of breadth of reactivity (61).
antidengue antibody with an NMR-mapped
epitope to identify and rationally design
Epitope Grafting To Elicit
mutations in the antibody CDR loops (58).
Neutralizing Antibodies
The authors used this information to create
several types of antibody mutations, includ- A complementary task to antigen design is
ing those that abolish binding, those that so-called epitope graftingremoving an
increase affinity for a single target, and those epitope from its native antigen and grafting
that increase the breadth of binding to multi- it onto a protein structure that can present it to
ple serotypes. immune receptors in a way that maximizes
186 SEVY AND MEILER

the immune response. This design task cedure is ineffective. To address this concern,
encompasses several unique challenges, the Azoitei et al. published a backbone grafting
most difficult of which is the selection of an method wherein they select a structurally
appropriate peptide onto which the epitope similar template and, rather than transfer
can be placed, known as the scaffold. This individual epitope side chains to a template,
scaffold must maintain the native conforma- remove the epitope-mimetic region en masse
tion of the desired epitope and have minimal and transplant the entire epitope backbone
immunogenicity in its off-epitope regions, all region from the native antigen (66). This
while maintaining favorable biophysical prop- protocol was benchmarked and shown to
erties. Advances in scaffold selection and produce peptides with higher affinity than
design have made this problem tractable and the previously described side-chain grafting
have shown promise in immunization. method, with the added advantage that back-
Early work in this field focused on bone grafting can be applied to templates with
placing a neutralizing linear epitope on a lower structural similarity.
stable scaffold that could maintain the Another challenge in the pursuit of a
native conformation while enhancing pre- generalizable epitope grafting method is
sentation of this epitope. HIV has been that initial reports focused on linear, contin-
widely used as a test case, since there are uous epitopes, even though many neutralizing
well-characterized broadly neutralizing an- epitopes are discontinuous. Azoitei et al.
tibodies with high-resolution crystal struc- developed a more aggressive scaffold search
tures available (62). Two independent groups and design method to identify scaffolds that
were able to graft the linear epitopes of can accommodate multiple discontinuous
two broadly neutralizing anti-HIV antibodies, epitope peptides while maintaining their con-
4E10 and 2F5, onto scaffolds to enhance their formations and relative orientations and min-
presentation and affinity for the desired anti- imizing steric clashes (67). The result of this
bodies (63, 65). These groups used similar protocol was an epitope scaffold that accu-
approaches of searching the PDB for scaffolds rately recapitulates the conformation of the
with a region of backbone conformation with epitope and interactions necessary for anti-
high structural similarity to the epitope and body-antigen interactions. McLellan et al.
using Rosetta Design to place epitope side pursued a similar strategy to create a discon-
chains onto the backbone scaffold and mini- tinuous epitope-presenting scaffold with a
mize the energy of side chain packing. This respiratory syncytial virus (RSV) epitope
protocol resulted in several designed peptides (68). In addition to the selection of an
with nanomolar affinity for their respective appropriate scaffold, there has also been
antibody targets and the ability to elicit work in redesigning a scaffold to reduce
broadly neutralizing sera upon immunization unwanted, nonepitope immunogenicity.
(6365). These earlier studies provided an Correia et al. showed that epitope scaffolds
important proof-of-principle for vaccine- can be further optimized by flexible backbone
based antigen design. remodeling and resurfacing to enhance ther-
One major limitation of initial reports on mostability, increase binding affinity, and
epitope grafting is the nature of the scaffold reduce immune reactivity (64).
selection process. Since it relies on placing To circumvent the problem of scaffold
epitope side chains on a backbone with a high selection, Correia et al. recently described a
degree of similarity to the epitope backbone, method to build a de novo scaffold for optimal
success is highly dependent on the presence of epitope presentation of an RSV epitope (42).
a suitable scaffold among structurally deter- They selected a three-helix bundle as a
mined proteins. In cases where there is no template topology and used extensive rounds
template with structural similarity, this pro- of sequence optimization and minimization
CHAPTER 10 Computer-Aided Prediction of Structure 187

to create an optimized conformation for experimentally is imperative. Computational


presentation. These scaffolds were purified design of the tightest or most broadly neutral-
and shown to be thermodynamically stable, izing antibodies is not only important for the
with affinities in the picomolar range (42). The development of optimal therapeutic antibodies
purified scaffolds were then used to immunize or the development of vaccines. Antibody
macaques and were shown to induce neutraliz- design is also the most stringent test of our
ing titers comparable to natural RSV infection, understanding of the rules that govern anti-
a significant landmark for the epitope-grafting body structure and interaction.
methodology.

ACKNOWLEDGMENT
CONCLUSION Conflicts of interest: We disclose no conflicts.

Computational approaches to predict the


structure of antibodies and antibody-anti- CITATION
gen complexes are of critical importance, as Sevy AM, Meiler J. 2014. Antibodies: computer-
the large number of naturally occurring aided prediction of structure and design of
antibodies restricts experimental character- function. Microbiol Spectrum 2(6):AID-0024-
ization to the most important cases. While 2014.
computational methods are already suffi-
ciently accurate to be useful, it remains a
focus of future research to develop better REFERENCES
sampling methods and more accurate ener- 1. Harris LJ, Larson SB, Hasel KW, McPherson
gy functions. The existing limitations in A. 1997. Refined structure of an intact IgG2a
monoclonal antibody. Biochemistry 36:15811597.
modeling antibodies make computational
2. Chothia C, Lesk AM. 1987. Canonical struc-
methods most useful when applied in a tures for the hypervariable regions of immuno-
tight feedback loop with experimental data, a globulins. J Mol Biol 196:901917.
situation that is expected to continue for the 3. Martin AC, Thornton JM. 1996. Structural
foreseeable future. The increasing throughput families in loops of homologous proteins: auto-
in methods to collect limited experimental matic classification, modelling and application to
antibodies. J Mol Biol 263:800815.
data on antibodies and antibody-antigen com- 4. Al-Lazikani B, Lesk AM, Chothia C. 1997.
plexes will further increase the need for Standard conformations for the canonical struc-
computational methods to add atomic detail tures of immunoglobulins. J Mol Biol 273:927948.
not present in these datasets. The rapidly 5. North B, Lehmann A, Dunbrack RL Jr. 2011.
increasing availability of high-resolution A new clustering of antibody CDR loop con-
formations. J Mol Biol 406:228256.
crystal structures of antibodies and antibody- 6. Briney BS, Willis JR, Crowe JE Jr. 2012.
antigen complexes is expected to improve Location and length distribution of somatic
computational prediction algorithms, for ex- hypermutation-associated DNA insertions and
ample, through increasing the accuracy of deletions reveals regions of antibody structural
plasticity. Genes Immun 13:523529.
knowledge-based potentials and through
7. Morea V, Tramontano A, Rustici M, Chothia
further completing the conformational clusters C, Lesk AM. 1998. Conformations of the third
of CDR loop conformations. Ultimately, the hypervariable region in the VH domain of immu-
reliable computational design of antibodies noglobulins. J Mol Biol 275:269294.
that recognize a target epitope is a long-term 8. Kuroda D, Shirai H, Kobori M, Nakamura
goal for computational structural biology. H. 2008. Structural classification of CDR-H3
revisited: a lesson in antibody modeling. Proteins
Given the even larger number of theoretically 73:608620.
possible antibody sequences, computational 9. Sivasubramanian A, Sircar A, Chaudhury S,
prioritization of the ones to characterize Gray JJ. 2009. Toward high-resolution homology
188 SEVY AND MEILER

modeling of antibody Fv regions and application Vajda S, Kozakov D. 2012. Application of asym-
to antibody-antigen docking. Proteins 74:497514. metric statistical potentials to antibody-protein
10. Zhu K, Day T. 2013. Ab initio structure prediction docking. Bioinformatics 28:26082614.
of the antibody hypervariable H3 loop. Proteins 24. Gray JJ, Moughon S, Wang C, Schueler-
81:10811089. Furman O, Kuhlman B, Rohl CA, Baker D.
11. Teplyakov A, Luo J, Obmolova G, Malia TJ, 2003. Protein-protein docking with simultaneous
Sweet R, Stanfield RL, Kodangattil S, Almagro optimization of rigid-body displacement and side-
JC, Gilliland GL. 2014. Second antibody modeling chain conformations. J Mol Biol 331:281299.
assessment. II. Structures and models. Proteins 25. Sircar A, Gray JJ. 2010. SnugDock: paratope
82:15631582. doi:10.1002/prot.24554. structural optimization during antibody-antigen
12. Almagro JC, Beavers MP, Hernandez-Guzman docking compensates for errors in antibody hom-
F, Maier J, Shaulsky J, Butenhof K, Labute P, ology models. PLoS Comput Biol 6:e1000644.
Thorsteinson N, Kelly K, Teplyakov A, Luo J, doi:10.1371/journal.pcbi.1000644.
Sweet R, Gilliland GL. 2011. Antibody modeling 26. Simonelli L, Beltramello M, Yudina Z, Macagno
assessment. Proteins 79:30503066. A, Calzolai L, Varani L. 2010. Rapid structural
13. Narayanan A, Sellers BD, Jacobson MP. characterization of human antibody-antigen com-
2009. Energy-based analysis and prediction plexes through experimentally validated compu-
of the orientation between light- and heavy- tational docking. J Mol Biol 396:14911507.
chain antibody variable domains. J Mol Biol 27. Sevy AM, Healey JF, Deng W, Spiegel PC,
388:941953. Meeks SL, Li R. 2013. Epitope mapping of
14. Marcatili P, Rosi A, Tramontano A. 2008. inhibitory antibodies targeting the C2 domain
PIGS: automatic prediction of antibody struc- of coagulation factor VIII by hydrogen-deute-
tures. Bioinformatics 24:19531954. rium exchange mass spectrometry. J Thromb
15. Whitelegg NR, Rees AR. 2000. WAM: an Haemost 11:21282136.
improved algorithm for modelling antibodies 28. Coales SJ, Tuske SJ, Tornasso JC, Hamuro
on the WEB. Protein Eng 13:819824. Y. 2009. Epitope mapping by amide hydrogen/
16. Maier JKY, Labute P. 2014. Assessment of deuterium exchange coupled with immobili-
fully automated antibody homology modeling zation of antibody, on-line proteolysis, liquid
protocols in MOE. Proteins 82:15991610. chromatography and mass spectrometry. Rapid
17. Sircar A, Kim ET, Gray JJ. 2009. Rosetta- Commun Mass Spectrom 23:639647.
Antibody: antibody variable region homology 29. Meeks SL, Healey JF, Parker ET, Barrow RT,
modeling server. Nucleic Acids Res 37:W474 Lollar P. 2007. Antihuman factor VIIIC2 domain
W479. antibodies in hemophilia A mice recognize a
18. Almagro JC, Teplyakov A, Luo J, Sweet RW, functionally complex continuous spectrum of
Kodangattil S, Hernandez-Guzman F, Gilliland epitopes dominated by inhibitors of factor VIII
GL. 2014. Second antibody modeling assessment activation. Blood 110:42344242.
(AMA-II). Proteins 82:15531562. 30. Chaves RC, Teulon JM, Odorico M, Parot P,
19. Sundberg EJ, Urrutia M, Braden BC, Isern J, Chen SWW, Pellequer JL. 2013. Conforma-
Tsuchiya D, Fields BA, Malchiodi EL, Tormo tional dynamics of individual antibodies using
J, Schwarz FP, Mariuzza RA. 2000. Estima- computational docking and AFM. J Mol Recognit
tion of the hydrophobic effect in an antigen- 26:596604.
antibody protein-protein interface. Biochemistry 31. Gogolinska A, Nowak W. 2013. Molecular basis
39:1537515387. of lateral force spectroscopy nano-diagnostics:
20. Moreira IS, Fernandes PA, Ramos MJ. 2007. computational unbinding of autism related che-
Hot spot computational identification: appli- mokine MCP-1 from IgG antibody. J Mol Model
cation to the complex formed between the hen 19:47734780.
egg white lysozyme (HEL) and the antibody 32. Sharon J, Rynkiewicz MJ, Lu ZH, Yang CY.
HyHEL-10. Int J Quantum Chem 107:299310. 2014. Discovery of protective B-cell epitopes
21. Wang X, Singh SK, Kumar S. 2010. Potential for development of antimicrobial vaccines and
aggregation-prone regions in complementarity- antibody therapeutics. Immunology 142:123.
determining regions of antibodies and their 33. Thornburg NJ, Nannemann DP, Blum DL,
contribution towards antigen recognition: a com- Belser JA, Tumpey TM, Deshpande S, Fritz
putational analysis. Pharm Res 27:15121529. GA, Sapparapu G, Krause JC, Lee JH, Ward AB,
22. Vajda S. 2005. Classification of protein complexes Lee DE, Li S, Winarski KL, Spiller BW, Meiler J,
based on docking difficulty. Proteins 60:176180. Crowe JE Jr. 2013. Human antibodies that
23. Brenke R, Hall DR, Chuang GY, Comeau SR, neutralize respiratory droplet transmissible H5N1
Bohnuud T, Beglov D, Schueler-Furman O, influenza viruses. J Clin Invest 123:44054409.
CHAPTER 10 Computer-Aided Prediction of Structure 189

34. McKinney BA, Kallewaard NL, Crowe JE Jr, 44. Igawa T, Tsunoda H, Tachibana T, Maeda A,
Meiler J. 2007. Using the natural evolution of Mimoto F, Moriyama C, Nanami M, Sekimori
a rotavirus-specific human monoclonal anti- Y, Nabuchi Y, Aso Y, Hattori K. 2010. Reduced
body to predict the complex topography of a elimination of IgG antibodies by engineering the
viral antigenic site. Immunome Res 3:8. variable region. Protein Eng Des Sel 23:385392.
35. Schneider S, Zacharias M. 2012. Atomic 45. Kanduc D, Lucchese A, Mittelman A. 2001.
resolution model of the antibody Fc interac- Individuation of monoclonal anti-HPV16 E7 anti-
tion with the complement C1q component. Mol body linear peptide epitope by computational
Immunol 51:6672. biology. Peptides 22:19811985.
36. Percy AJ, Rey M, Burns KM, Schriemer DC. 46. Tsurushita N, Hinton PR, Kumar S. 2005.
2012. Probing protein interactions with hydro- Design of humanized antibodies: from anti-Tac
gen/deuterium exchange and mass spectrom- to Zenapax. Methods 36:6983.
etry: a review. Anal Chim Acta 721:721. 47. Teng G, Papavasiliou FN. 2007. Immunoglobulin
37. Baerga-Ortiz A, Hughes CA, Mandell JG, somatic hypermutation. Annu Rev Genet 41:107120.
Komives EA. 2002. Epitope mapping of a 48. Filpula D. 2007. Antibody engineering and mod-
monoclonal antibody against human thrombin ification technologies. Biomol Eng 24:201215.
by R/D-exchange mass spectrometry reveals 49. Pantazes RJ, Maranas CD. 2010. OptCDR: a
selection of a diverse sequence in a highly general computational method for the design of
conserved protein. Protein Sci 11:13001308. antibody complementarity determining regions
38. Aiyegbo MS, Sapparapu G, Spiller BW, Eli for targeted epitope binding. Protein Eng Des
IM, Williams DR, Kim R, Lee DE, Liu T, Li S, Sel 23:849858.
Woods VL Jr, Nannemann DP, Meiler J, 50. Leaver-Fay A, Jacak R, Stranges PB, Kuhlman
Stewart PL, Crowe JE Jr. 2013. Human B. 2011. A generic program for multistate protein
rotavirus VP6-specific antibodies mediate intra- design. PloS One 6:e20937. doi:10.1371/journal.
cellular neutralization by binding to a quaternary pone.0020937.
structure in the transcriptional pore. PloS One 8: 51. Havranek JJ, Harbury PB. 2003. Automated
e61101. doi:10.1371/journal.pone.0061101. design of specificity in molecular recognition.
39. Bublil EM, Yeger-Azuz S, Gershoni JM. Nat Struct Biol 10:4552.
2006. Computational prediction of the cross- 52. Willis JR, Briney BS, DeLuca SL, Crowe JE
reactive neutralizing epitope corresponding to Jr, Meiler J. 2013. Human germline antibody
the monoclonal antibody b12 specific for HIV-1 gene segments encode polyspecific antibodies.
gp120. FASEB J 20:17621774. PLoS Comput Biol 9:e1003045. doi:10.1371/
40. Chuang GY, Acharya P, Schmidt SD, Yang Y, journal.pcbi.1003045.
Louder MK, Zhou T, Kwon YD, Pancera M, 53. Babor M, Kortemme T. 2009. Multi-con-
Bailer RT, Doria-Rose NA, Nussenzweig MC, straint computational design suggests that
Mascola JR, Kwong PD, Georgiev IS. 2013. native sequences of germline antibody H3
Residue-level prediction of HIV-1 antibody loops are nearly optimal for conformational
epitopes based on neutralization of diverse viral flexibility. Proteins 75:846858.
strains. J Virol 87:1004710058. 54. Clark LA, Boriack-Sjodin PA, Eldredge J, Fitch
41. Chuang GY, Liou D, Kwong PD, Georgiev IS. C, Friedman B, Hanf KJ, Jarpe M, Liparoto SF,
2014. NEP: web server for epitope prediction based Li Y, Lugovskoy A, Miller S, Rushe M, Sherman
on antibody neutralization of viral strains with W, Simon K, Van Vlijmen H. 2006. Affinity
diverse sequences. Nucleic Acids Res 42:W64W71. enhancement of an in vivo matured therapeutic
42. Correia BE, Bates JT, Loomis RJ, Baneyx G, antibody using structure-based computational
Carrico C, Jardine JG, Rupert P, Correnti C, design. Protein Sci 15:949960.
Kalyuzhniy O, Vittal V, Connell MJ, Stevens 55. Lippow SM, Wittrup KD, Tidor B. 2007.
E, Schroeter A, Chen M, Macpherson S, Computational design of antibody-affinity im-
Serra AM, Adachi Y, Holmes MA, Li Y, provement beyond in vivo maturation. Nat
Klevit RE, Graham BS, Wyatt RT, Baker D, Biotechnol 25:11711176.
Strong RK, Crowe JE Jr, Johnson PR, Schief 56. Farady CJ, Sellers BD, Jacobson MP, Craik
WR. 2014. Proof of principle for epitope- CS. 2009. Improving the species cross-reactivity
focused vaccine design. Nature 507:201206. of an antibody using computational design. Bioorg
43. Hinton PR, Johlfs MG, Xiong JM, Hanestad Med Chem Lett 19:37443747.
K, Ong KC, Bullock C, Keller S, Tang MT, Tso 57. Barderas R, Desmet J, Timmerman P,
JY, Vasquez M, Tsurushita N. 2004. Engineered Meloen R, Casal JI. 2008. Affinity maturation
human IgG antibodies with longer serum half- of antibodies assisted by in silico modeling.
lives in primates. J Biol Chem 279:62136216. Proc Natl Acad Sci USA 105:90299034.
190 SEVY AND MEILER

58. Simonelli L, Pedotti M, Beltramello M, Livoti 64. Correia BE, Ban YE, Friend DJ, Ellingson K,
E, Calzolai L, Sallusto F, Lanzavecchia A, Xu H, Boni E, Bradley-Hewitt T, Bruhn-
Varani L. 2013. Rational engineering of a human Johannsen JF, Stamatatos L, Strong RK,
anti-dengue antibody through experimentally Schief WR. 2010. Computational protein de-
validated computational docking. PloS One 8: sign using flexible backbone remodeling and
e55561. doi:10.1371/journal.pone.0055561. resurfacing: case studies in structure-based
59. Giles BM, Ross TM. 2011. A computationally antigen design. J Mol Biol [Epub ahead of
optimized broadly reactive antigen (COBRA) print.] doi:10.1016/j.jmb.2010.09.061.
based H5N1 VLP vaccine elicits broadly reac- 65. Ofek G, Guenaga FJ, Schief WR, Skinner
tive antibodies in mice and ferrets. Vaccine J, Baker D, Wyatt R, Kwong PD. 2010.
29:30433054. Elicitation of structure-specific antibodies by
60. Giles BM, Bissel SJ, DeAlmeida DR, Wiley epitope scaffolds. Proc Natl Acad Sci USA 107:
CA, Ross TM. 2012. Antibody breadth and 1788017887.
protective efficacy are increased by vaccina- 66. Azoitei ML, Ban YE, Julien JP, Bryson S,
tion with computationally optimized hemaggluti- Schroeter A, Kalyuzhniy O, Porter JR, Adachi
nin but not with polyvalent hemagglutinin-based Y, Baker D, Pai EF, Schief WR. 2012. Computa-
H5N1 virus-like particle vaccines. Clin Vaccine tional design of high-affinity epitope scaffolds by
Immunol 19:128139. backbone grafting of a linear epitope. J Mol Biol
61. Wu X, Yang ZY, Li Y, Hogerkorp CM, Schief 415:175192.
WR, Seaman MS, Zhou T, Schmidt SD, Wu L, 67. Azoitei ML, Correia BE, Ban YE, Carrico C,
Xu L, Longo NS, McKee K, ODell S, Louder Kalyuzhniy O, Chen L, Schroeter A, Huang
MK, Wycuff DL, Feng Y, Nason M, Doria- PS, McLellan JS, Kwong PD, Baker D, Strong
Rose N, Connors M, Kwong PD, Roederer M, RK, Schief WR. 2011. Computation-guided
Wyatt RT, Nabel GJ, Mascola JR. 2010. backbone grafting of a discontinuous motif
Rational design of envelope identifies broadly onto a protein scaffold. Science 334:373376.
neutralizing human monoclonal antibodies to 68. McLellan JS, Pancera M, Carrico C, Gorman
HIV-1. Science 329:856861. J, Julien JP, Khayat R, Louder R, Pejchal R,
62. Stamatatos L, Morris L, Burton DR, Mascola Sastry M, Dai K, ODell S, Patel N, Shahzad-
JR. 2009. Neutralizing antibodies generated ul-Hussan S, Yang Y, Zhang B, Zhou T, Zhu
during natural HIV-1 infection: good news for J, Boyington JC, Chuang GY, Diwanji D,
an HIV-1 vaccine? Nat Med 15:866870. Georgiev I, Kwon YD, Lee D, Louder MK,
63. Correia BE, Ban YE, Holmes MA, Xu H, Moquin S, Schmidt SD, Yang ZY, Bonsignori
Ellingson K, Kraft Z, Carrico C, Boni E, M, Crump JA, Kapiga SH, Sam NE, Haynes
Sather DN, Zenobia C, Burke KY, Bradley- BF, Burton DR, Koff WC, Walker LM,
Hewitt T, Bruhn-Johannsen JF, Kalyuzhniy Phogat S, Wyatt R, Orwenyo J, Wang LX,
O, Baker D, Strong RK, Stamatatos L, Schief Arthos J, Bewley CA, Mascola JR, Nabel GJ,
WR. 2010. Computational design of epitope- Schief WR, Ward AB, Wilson IA, Kwong PD.
scaffolds allows induction of antibodies spe- 2011. Structure of HIV-1 gp120 V1/V2 domain
cific for a poorly immunogenic HIV vaccine with broadly neutralizing antibody PG9. Nature
epitope. Structure 18:11161126. 480:336343.
PATHOGEN-SPECIFIC
ANTIBODIES
Antibodies Targeting the Envelope
of HIV-1

LUZIA M. MAYR1 and SUSAN ZOLLA-PAZNER2,3


11
INTRODUCTION

HIV continues to be a major global public health issue, with an estimated 35


million people living with the virus and more than 2 million new infections
occurring yearly (1). As part of the natural immune response, antibodies
(Abs) exert immune pressure on HIV and play a key role both in controlling
the virus and in driving escape mutations in the viral envelope glycopro-
teins. Therefore, and because the elicitation of Abs is believed to be crucial
for an effective vaccine against HIV, Abs targeting HIV have been the focus
of intense research in the past years.

ANTIBODY RESPONSE TO HIV

Upon infection with HIV, a strong Ab response occurs in essentially all


infected individuals. These Abs are directed against several viral proteins,
including the gp120 and gp41 envelope proteins that are found on the
surface of the virus particles. While in many viral diseases, such as influenza
and polio, the Abs against surface antigens can establish protective

1
INSERM U1109, Universit de Strasbourg, 67000 Strasbourg, France; 2New York Veterans Affairs Harbor
Healthcare System, New York, NY 10010; 3New York University School of Medicine, New York, NY 10016.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0025-2014

193
194 MAYR AND ZOLLA-PAZNER

immunity, many HIV envelope-specific Abs the letters A, B, C, D, F, G, H, J, and K. Overall,


have little neutralizing capacity due to the the amino acid sequence of the different
many complex escape mechanisms employed subtypes can vary by as much as 30% from
by the surface viral glycoproteins that occur as one clade to another, but in certain genomic
sparse trimeric spikes in the virus envelope regions the genetic variability can be as high as
(2). These virus escape mechanisms include 42%. Further complicating the HIV landscape
the following. are recombinant forms of the virus in which
heterologous virus strains combine segments
of their genome during reverse transcription
Glycan Shield
to produce genetically divergent new virus
One of the reasons why HIV is a difficult forms. Countless recombinants of virtually
target for Abs that prevent infection is the every virus strain combination have been
dense glycosylation of the envelope proteins identified worldwide, and their prevalence
gp120 and gp41. With approximately 25 N- differs by geographic regions (10). A protective
linked glycosylation sites, gp120 is one of vaccine will need to induce Abs that target this
the most heavily glycosylated viral proteins tremendous viral diversity.
described. These glycans are large, complex
carbohydrate structures that shield vulner-
Conformational Masking
able epitopes on the surface of HIV, leading
to viral escape from Abs (3). The function of The envelope spike is a heterotrimer con-
glycans can be demonstrated by the removal sisting of three gp120 molecules noncovalently
of certain glycosylation sites, leading to a anchored to the viral membrane via three gp41
significant increase in neutralization sensi- molecules. In an attempt to shield neutraliza-
tivity of the virus (4, 5). tion-sensitive domains, the elements of the
envelope spike adopt a quaternary conforma-
tion where domains of neighboring gp120
High Mutation Rate
subunits can interact. For example, it was
The high mutation rate of HIV-1 is another suggested that in the apex of the three-
obstacle for the development of immune dimensional envelope spike, intersubunit con-
protection by neutralizing Abs. With an tact between the V1, V2, and V3 loops occurs,
in vitro mutation rate of approximately protecting adjacent regions from recognition
2.2 x 105 to 5.4 x 105 per nucleotide base by Abs (11).
per replication cycle, the virus continuously To enter its target cells, HIV must bind
and rapidly evolves to escape neutralization sequentially to cellular receptors, including
(6). In an attempt to recognize the mutated CD4 and one of two chemokine receptors
virus, neutralizing Abs (nAbs) evolve in (CXCR4 or CCR5). The regions of gp120
tandem with the virus, resulting in succes- that are involved with binding to these
sive waves of nAb maturation followed by receptors must, therefore, be conserved to
viral escape. This leads to the phenomenon maintain the infectious potential of the
of nAbs often being able to neutralize virus virus. The CD4 binding site (CD4bs) and
from months earlier, but not concurrent chemokine receptor binding sites on gp120
circulating variants (79). would thus appear to be good targets for
The high mutation rate and the fact that Abs; however, while able to bind to some
billions of new viral particles are produced exceptional Abs (9, 12, 13), the CD4bs is
daily within each patient leads to an enormous partially obscured by glycans and variable
diversity of HIV-1 variants. Based on their regions and undergoes conformational reor-
sequence, the variants have been divided into ganization, allowing it to evade neutraliza-
nine distinct subtypes, or clades, designated by tion by conventional CD4bs-specific Abs.
Chapter 11 Antibodies Targeting the Envelope of HIV-1 195

Thus, the virus places an energetic barrier to are neutralized with low levels of nAbs (<<1 to
Ab binding (14). 10 mg/ml), often >10 g/ml are required to
It is believed that dynamic conformational neutralize tier 2 pseudoviruses (21, 22).
changes also play a role in masking conserved The Thai clinical HIV-1 vaccine trial
epitopes on chemokine receptor binding sites RV144 provided additional support for
(15); regions of gp120 that are involved in the role of conventional Abs in protection:
binding to the chemokine receptorthe V3 high levels of V1V2 and V3 IgG Ab levels,
loop and the b20/b21 strands of the bridging especially those of the IgG3 subclass, were
sheetform an exposed surface only after found to be significantly associated with the
binding to CD4 and are thus exposed to Abs reduced infection rate (estimated 31% vac-
only transiently (16). cine efficacy) in vaccine recipients (2328).
Monoclonal Abs isolated from the RV144
vaccinees showed very low mutation rates
TYPES OF ANTIBODIES
from germline genes encoding the variable
region of the heavy chain (VH)mutation
Conventional Abs
rate range of 1.5 to 4.5% (29, 30), which is
Abs that commonly occur during HIV even lower than that noted after influenza
infection and that are present in the major- immunization (mean VH mutation rate of
ity of infected individuals are here defined 8.1% [31]). These data further uphold the
as conventional Abs. These Abs do not concept that conventional Abs can be asso-
exhibit unusual structural or genetic charac- ciated with reduced infection rates.
teristics, and their immunoglobulin genes Thus, even though conventional nAbs are
undergo relatively little somatic hypermuta- limited in their breadth and potency, they
tion from germline (17, 18). Conventional Abs are commonly made by HIV-positive indi-
have long been known to protect against viduals (32), by immunized humans (24),
infection in animal systems. This was estab- and by immunized animals (33, 34), and the
lished with passive immunization of chim- previously mentioned studies suggest that
panzees using IgG preparations from the blood they can reduce infection rates. Therefore,
of HIV-infected individuals (19). Thus, the such Abs, induced by vaccines, have sub-
proof of principle was established more than stantial potential for influencing the course
two decades ago that Abs alone from unselect- of the HIV epidemic.
ed HIV-infected individuals could provide
sterilizing immunity.
Exceptional, Broadly Neutralizing Abs
Conventional Abs are elicited as early as
two weeks after seroconversion in acutely Unlike conventional HIV Abs, broadly neu-
infected individuals; however, they display tralizing Abs (bNAbs) are found in relatively
very limited neutralization breadth (20): using rare HIV-infected subjects (35), with the most
a standardized reference pseudovirus panel broad and potent serum Abs being identified
that represents genetically diverse subsets of in only 1% of elite neutralizers (36). Most
viruses, conventional Abs targeting gp120 of the broadly neutralizing monoclonal Abs
were shown to neutralize up to 50% of tier 1 (bNmAbs) have been developed in the last few
pseudoviruses, but 9% of tier 2 pseudo- years (3741). In addition to wide breadth
viruses, suggesting that these Abs target (neutralization of 100% of tier 1 viruses and
epitopes exposed on a minority of viruses 72 to 100% of tier 2 viruses), bNmAbs are very
and tend to be specific for the virus infecting potent, and in vitro assays have shown that
the host. Conventional neutralizing Abs very small amounts are sufficient for neutral-
(nAbs) also have a limited neutralizing poten- ization of tier 1 and tier 2 viruses (<1 mg/ml and
cy in vitro, and while most tier 1 pseudoviruses 0.02 to 27.0 mg/ml, respectively) (17).
196 MAYR AND ZOLLA-PAZNER

For bNAbs to acquire their impressive completely protect against infection with
breadth and potency, extensive somatic simian/HIV when these Abs are present
hypermutation is requiredparticularly in at serum concentrations as low as 30 mg/ml
the variable heavy chain (VH) genes and in (5355). Broadly neutralizing mAbs are also
some framework regionsa process that being developed for treatment of established
usually requires more than a year of exposure HIV infection, a process in which bNmAbs
to the virus after HIV infection (42, 43). will be infused into HIV-infected individuals
Several additional unique characteristics of to decrease and/or eliminate virus. Success
bNmAbs have been described, such as fre- has already been achieved in this area as
quent auto- and/or poly-reactivity for host demonstrated by treatment of humanized
antigens and long heavy chain complementar- mice and macaques infected with HIV and
ity-determining region 3 (HCDR3) sequences simian/HIV, respectively (5658).
composed of 20 to 34 residues, which stand in There is also great interest in whether pas-
stark contrast to the length of HCDR3s sively transferred bNmAbs can contribute to
produced by human B cells, which averages the prevention of mother-to-child-transmission
16 residues (4447). These features contribute of HIV-1, the continuing cause of a signifi-
to the rarity of bNAbs and pose a major cant percentage of new infections in the
challenge for the development of an HIV-1 developing world. During pregnancy, HIV-
vaccine designed to induce such Abs. specific Abs can pass from an HIV-infected
While bNmAbs are exceptionally potent mother to the fetus through the placenta.
and provide protection in animal models These Abs, however, are not effective against
(4851), all attempts to elicit bNAb responses later HIV infection, for example, during the
by vaccination have so far been unsuccessful, breast feeding phase. Moreover, the trans-
and it is believed that a panel of special mitted virus variants have fewer potential N-
immunogens which will both stimulate suit- linked glycosylation sites, a fact that could
able nave B cells and guide them through impact positively on the interaction of glycan-
lineage maturation will be required to achieve dependent bNAb with transmitter/founder
this goal (52); this process of Ab maturation viruses recognition (59). Indeed, transmitted
appears to be long and complex. HIV variants were shown to be susceptible to
To design these immunogens, bNmAbs various bNmAbs, but a combination of potent
from infected donors are being isolated and bNmAbs targeting diverse epitopes might be
sequenced to reconstruct the lineage of the needed to successfully protect against HIV
Abs, including the sequence of the probable infection in the mother-to-child-transmission
common unmutated ancestor of a given context (60).
bNmAb. The envelope glycoproteins rec-
ognized by the Abs in the bNmAb lineage
are being expressed, and these will serve as VACCINE DEVELOPMENT AND
immunogens to be used sequentially to IMMUNOGEN DESIGN
engage the nave B cell receptor and to
stimulate and guide the evolution of the nAb As a result of the extensive data summarized
response (44). above, the elicitation of Abs is clearly indicated
While the ultimate success of this path as a requirement for a successful HIV vaccine.
toward the design of a prophylactic vaccine As noted, this is a challenging task. The effort
is unknowable at this point, there is, never- to develop vaccine candidates capable of
theless, great potential for the use of bNmAbs inducing protective Abs has accelerated dra-
for passive immunization to protect against matically since the Thai vaccine trial RV144
and treat HIV infection. Thus, treatment of revealed that Abs were associated with a
macaques with bNmAbs has been shown to reduced rate of infection (2426, 28).
Chapter 11 Antibodies Targeting the Envelope of HIV-1 197

Several envelope-based immunogens sites of vulnerability (74) defined as the


have been tested for their ability to induce glycan-dependent V2 epitope (see V2q epi-
nAbs. Monomeric gp120 is relatively easy to tope in the list below), the glycan-dependent
produce and has been widely used in animal epitope at the base of V3, the CD4 binding site
studies. Initial human vaccine trials elicited in gp120, and the membrane proximal external
only weak nAbs, and no protection against region (MPER) in gp41.
HIV infection was achieved (61, 62). The
RV144 vaccine trial, however, demonstrated Abs Targeting Variable
a beneficial effect of monomeric gp120 in Loops 1 and 2 (V1V2)
combination with a canarypox prime (63). It
Electron tomography, cryo-electron micro-
was suggested that the epitopes exposed on
scopy, and biochemical studies have shown
monomeric gp120 are poor neutralization
that the V1V2 domain is localized at the
targets because they are occluded on the
apex of the trimeric HIV-1 Env structures,
native envelope trimer (64), and therefore
and therefore at least some of the V1V2
immunogens were designed that better
epitopes are accessible to Abs (75, 76). V2
mimic the native envelope spike. Since the
loop sequences differ in length, but the
trimeric envelope is highly unstable and
majority of amino acids are highly conserved,
difficult to produce, a variety of different
suggesting conserved structural elements
immunogens using gp140, the ectodomain
(77). The V1V2 region forms four antiparallel
of trimeric gp160, have been made (6567).
b-strands (A, B, C, and D) which are linked
To date, immunizations using trimeric en-
via disulfide bonds (78). Via a conserved tri-
velope immunogens have not been success-
peptide, the 179LDV/I181 binding motif, V2
ful in the induction of potent nAbs (68, 69).
can bind to a4b7, an integrin expressed on
Recent studies, monitoring the coevolution
activated CD4+ T cells that is required for
of virus and bNAbs in patients during natural
the homing of CD4+ T cells to the gut mu-
infection, demonstrated that a tight interplay
cosa (79).
between Ab maturation and subsequent viral
Abs targeting the V1V2 region were
escape drives the development of bNAb
associated with a lower risk of infection
responses (9, 70, 71). These studies provide
in the RV144 clinical vaccine trial, thus
important new insights into the elicitation of
making this area a promising target for vaccine
bNAbs and will advance the development of
development and the focus of intense re-
better immunogens.
search (2426). To date, three different epi-
Rather than using whole Env monomers
tope types have been defined in the V1V2
or trimers, another approach to vaccine design
region:
is the use of recombinant immunogens that
target the Ab response to particular epitopes. V2i epitope. A group of seven human mAbs
Computationally designed vaccines that recognizes a conformation-dependent
mimic viral and bacterial epitopes have been epitope, designated V2i since these
shown to induce potent conventional protec- Abs target the disordered region in V2
tive nAbs against various viruses and bacterial that connects the C and D strands and
pathogens (72, 73). This same approach is includes the a4b7-integrin binding site
being applied to the design of recombinant (hence the term V2i) (80, 81). The
vaccines that target specific HIV epitopes. structure of this region has so far not
At the present time, the type of epitopes to been solved, suggesting that it is highly
be targeted by conventional Abs include flexible and dynamic. Abs targeting V2i
the glycan-independent V1V2 and V3 regions are highly cross-reactive in binding to
of the HIV envelope, whereas the type of monomeric gp120 but do not neutral-
epitopes to be targeted by bNAbs include ize HIV well (18), suggesting that the
198 MAYR AND ZOLLA-PAZNER

epitope is mostly occluded from Ab tion sites at its N- and C-terminal ends, and
recognition in the trimeric envelope. several conserved structural features. The
V2p epitope. This epitope is defined by two V3 loop can be divided into three structural
mAbs (CH58 and CH59) that were regions: (i) a base region that is located
isolated from an RV144 vaccinee. The in the gp120 core and includes a disulfide
epitope is glycan-independent, and bond, (ii) a flexible stem region, and (iii) a
these mAbs bind V2 peptides (thus, distal crown that contains the highly con-
V2p) and selected monomeric gp120, served GPGR/Q motif at its tip. The recog-
recognizing an epitope composed of nition of conserved V3 elements contributes
helical or helical-coil structures in the to the broad cross-reactivity of V3-specific
C strand of V1V2 (30, 82). Abs (89, 90).
V2q epitope. This is a quaternary epitope, V3 Abs are present in essentially all
which is preferentially expressed on the infected individuals (91), and V3 Abs have
trimeric structure of the gp120 spike. been elicited by several types of vaccines (4,
Crystallographic studies with a V1V2- 92, 93). Moreover, the first demonstration
fusion protein show that broad and of the successful use of reverse vaccinology,
potently neutralizing V2q mAbs bind to i.e., the design of vaccines based on epitopes
relatively conserved residues within V2 as recognized by biologically active mAbs, was
well as to N-linked glycansmost impor- achieved using V3-scaffold immunogens
tantly the N160 glycan. Earlier studies which targeted the immune response to this
also showed that the binding of V2q- single epitope of the HIV envelope. For this,
specific mAbs was influenced by residues the V3 loop was spliced into a conforma-
in V3 (37, 83, 84). V2q-specific bNmAbs, tionally correct site on the highly immuno-
including PG9, PG16, and CH01, are genic protein, cholera toxin subunit B, a
extremely potent and broad in their protein which forms a pentameric structure
reactivity and have long CDRH3 loops and therefore presents five copies of V3 (94),
that interact with N-linked glycans and serving as a particularly strong antigen for
reach around them to contact amino acids induction of Abs (95). High anti-V3 Ab titers
of V2. These V2q mAbs are highly were elicited in rabbits with one or a combi-
mutated from germline (44). nation of V3-cholera toxin subunit B immu-
nogens, and these immune sera were able
to neutralize numerous diverse HIV strains
Abs Targeting Variable Loop 3 (V3)
(33, 94).
The V3 loop is located in close proximity Just as Abs to V2 target three regions
to the V1V2 domain at the apex of the (V2i, V2p, and V2q), three types of V3 Abs
envelope trimer (85) and is involved in have been described.
CCR5 or CXCR4 coreceptor tropism and
binding. It thus plays an important role in Glycan-independent ladle-like V3 Abs
virus entry into the host cell, and it is The V3 crown is an immunodominant
required for infectivity since V3-deleted region, and Abs targeting the epitopes in
mutants are noninfectious (86). the crown are made by essentially all HIV-
While, by definition, there is consider- infected individuals (91). Abs to this region
able amino acid sequence variation in V3, are glycan-independent. The ladle-like anti-
about 60% of the amino acids are conserved, V3 mAbs bind to the tip of the V3 crown
and the variation occurs at restricted posi- which sits in the bowl of the ladle while
tions (87, 88). The region is characterized the N-terminal V3 beta-strand adheres to
by a conserved length of 34 to 35 amino the handle of the ladle. Representative
acids, the presence of N-linked glycosyla- mAbs of this type include 447-52D, where
Chapter 11 Antibodies Targeting the Envelope of HIV-1 199

the long CDRH3 forms the handle of the like and the PGT128-like Ab families, are
ladle that interacts with the main chain of extremely potent and broadly reactive.
the N-terminal beta-chain of the V3 crown. These mAbs are highly somatically mutated
and require specific glycan interactions,
Glycan-independent cradle-like V3 Abs particularly at position N332 (38, 104). The
The second type of V3 Ab uses an antigen- crystal structure of PGT128 in complex
binding mode typified by mAb 2557. In such with an engineered outer domain of gp120
cradle-like Abs the antigen binding site recently showed that this Ab also interacts
consists of a groove in the Fab fragment, with the N301 oligomannose glycan, a
and the epitope lies in this groove, resem- position that is not recognized by PGT121-
bling a baby in a cradle; in this case, the like Abs (104). Even though the two Ab
major binding site is the hydrophobic core families are suggested to approach gp120
of the V3 crown, usually composed of from different angles, both block HIV-1
hydrophobic, conserved residues 307, 309, infection by interfering with CD4 binding
and 317 (89, 96, 97). through allosteric mechanisms (105).
Both types of glycan-independent Abs
specific for the V3 crown can neutralize
Abs Targeting the CD4 Binding
most laboratory-adapted HIV-1 strains and
Site (CD4bs)
tier 1 viruses but neutralize relatively few
tier 2 viruses using standard neutralization The CD4bs is functionally highly conserved
assays (21, 98, 99). This is largely due to and thus seems to be an ideal target for Abs.
masking of the V3 loop by glycans (100) and However, it is well hidden by surrounding
by the V1V2 domain that is situated atop the glycans and variable regions (74), and Abs
trimer. Deletion of V1V2 leads to a better are obstructed from binding due to steric
exposure of V3 epitopes and thus better neu- and conformational hindrance (see section
tralization by V3 mAbs (101, 102). Import- on conformational masking, above). Studies
antly, it was recently shown that anti-V3 Abs have shown that many CD4bs mAbs can
are effective against tier 2 viruses if the Ab and bind with high affinity to recombinant
virus are coincubated for several hours rather gp120 but cannot access the CD4bs on the
than for 1 to 2 hours, which is the norm in envelope trimer. The mAb b12 was the first
standard neutralization assays (103). These neutralizing mAb discovered to successfully
results suggest that the V3 loop is meta-stable target the CD4bs, but its breadth and
on the virus surface, flickering between a potency are restricted due to amino acid
cryptic and exposed conformation, the latter variation both within and outside of the
being both required for interaction with the CD4bs (44, 106, 107).
chemokine receptor and available for Ab bind- More recently, several bNmAbs have
ing leading to neutralization. Additionally, been isolated that mimic binding of CD4
CD4 binding induces a conformational change to gp120 and as a result neutralize HIV-1
in gp120, releasing the V3 crown from the potently and with broad reactivity. These
surface of the envelope trimer and thus CD4bs-specific bNmAbs, isolated from var-
augmenting V3 epitope exposure and sensi- ious HIV-infected individuals, share several
tivity to V3 Ab neutralization (90). genetic and structural characteristics. First,
their heavy chains all derive from the VH1-2
Glycan-dependent V3 Abs or the closely related VH1-46 germline
The base of V3 is poorly immunogenic, genes. These Abs are also highly somatically
eliciting Abs in a relatively small proportion mutated, with 20 to 30% of nucleotide
of infected individuals. Nonetheless, mAbs changes in their heavy chains compared
that target this region, such as the PGT121- to germline. However, attempts to induce
200 MAYR AND ZOLLA-PAZNER

these CD4bs bNAbs have been problematic, to mimic self antigens, and therefore the
even using engineered gp120 and recom- responses of B cells with MPER specificity
binant designer immunogens modeled on are down-modulated (46).
the structure of the epitopes contacted by
the very effective bNmAbs (108110). Cross-
clade nAbs were induced in rabbits via the CONCLUSIONS
reverse vaccinology approach. Immunogens
were designed based on the epitope recog- Three decades of study have established the
nized by the mAb IgG1b12. Thus, fragments important role of Abs in protecting against
of gp120 containing 70% of the b12 epitope HIV infection. However, it has become quite
were used for priming of rabbits. The clear that HIV uses many mechanisms to
animals received a boost with full-length protect itself from the biologic effects of Abs
gp120 after 16 and 51 weeks. Cross-clade that would block infectivity. Design of an
neutralizing HIV-specific Abs were elicited effective vaccine must take into account the
in the rabbits, which neutralized tier 1, 2, presence of glycans and masking phenomena
and 3 viruses (111), providing a maximum to induce Abs that can penetrate or circum-
geometric mean of IC50 titers against five vent these protective shields employed by the
tier 2 and 3 viruses of 1:134. virion. Current immunogen design is affected
profoundly by whether the aim is to induce
conventional Abs or exceptional broadly neu-
Abs Targeting the Membrane-Proximal
tralizing Abs. Induction of exceptional Abs
External Region (MPER)
with a vaccine may require the use of a series
The MPER consists of the last 24 C-terminal of immunogens that guide the immune
amino acids of the gp41 ectodomain. Its response through the mutations required for
sequence is highly conserved, contains many the specificities displayed by broad and potent
hydrophobic residues, and is usually rich in neutralizing Abs. However desirable this
tryptophans. It is believed that the MPER goal is, whether it is achievable has yet to
undergoes significant conformational changes be established. Alternatively, conventional
during viral entry (112114). Abs, while not as broad or as potent as
Different epitopes have been described exceptional Abs have already been elicited by
in the MPER: the very potent human mAb vaccine trials and are correlated with a reduced
10E8 recognizes an a-helix in this region, rate of infection in the RV144 phase III
while other human mAbs, including 2F5 clinical vaccine trial. Induction of conventional
and 4E10, target an overlapping region that protective Abs is therefore possible. The vac-
additionally includes residues of the trans- cine regimens and reagents to be used in
membrane spanning domain. As opposed vaccine development are many, ranging from
to mAbs 2F5 and 4E10, bNmAb 10E8 can DNA and viral vector priming immunogens to
neutralize 95% of viruses tested and lacks proteins representing the trimeric envelope
detectable reactivity with self-antigens, a fea- proteins or portions thereof spliced onto immu-
ture of the less potent 2F5 and 4E10 (40, 44). nogenic scaffolds. Many more clinical trials
Like the bNAbs targeting gp120, bNAbs for safety, immunogenicity, and protection are
specific for the MPER of gp41 have been required to establish which of these many
extremely difficult to induce with vaccines regimens and reagents will result in a prophy-
(115). They are present in only a minority of lactic vaccine.
HIV-infected individuals (35, 116), sug-
gesting that this is a poorly immunogenic
ACKNOWLEDGMENT
region. This is supported by the finding that
MPER mAbs such as 2F5 and 4E10 appear Conflicts of interest: We disclose no conflicts.
Chapter 11 Antibodies Targeting the Envelope of HIV-1 201

CITATION Mascola JR, Haynes BF. 2013. Co-evolution of


a broadly neutralizing HIV-1 antibody and
Mayr LM, Zolla-Pazner S. 2015. Antibodies founder virus. Nature 496:469476.
targeting the envelope of HIV-1. Microbiol 10. Hemelaar J. 2012. Implications of HIV diver-
sity for the HIV-1 pandemic. J Infect 66:391400.
Spectrum 3(1):AID-0025-2014.
11. Rusert P, Krarup A, Magnus C, Brandenberg
OF, Weber J, Ehlert AK, Regoes RR,
Gunthard HF, Trkola A. 2011. Interaction of
REFERENCES
the gp120 V1V2 loop with a neighboring gp120 unit
1. UNAIDS. 2013. UNAIDS report on the global shields the HIV envelope trimer against cross-
AIDS epidemic. http://www.unaids.org/sites/ neutralizing antibodies. J Exp Med 208:14191433.
default/files/en/media/unaids/contentassets/ 12. Zhou T, Georgiev I, Wu X, Yang ZY, Dai K,
documents/epidemiology/2013/gr2013/ Finzi A, Kwon YD, Scheid JF, Shi W, Xu L,
UNAIDS_Global_Report_2013_en.pdf. Yang Y, Zhu J, Nussenzweig MC, Sodroski J,
2. Mascola JR, Montefiori DC. 2003. HIV-1: Shapiro L, Nabel GJ, Mascola JR, Kwong PD.
natures master of disguise. Nat Med 9:393 2010. Structural basis for broad and potent
394. neutralization of HIV-1 by antibody VRC01.
3. Wei X, Decker JM, Wang S, Hui H, Kappes Science 329:811817.
JC, Wu X, Salazar-Gonzalez JF, Salazar MG, 13. Wu X, Zhou T, Zhu J, Zhang B, Georgiev I,
Kilby JM, Saag MS, Komarova NL, Nowak Wang C, Chen X, Longo NS, Louder M,
MA, Hahn BH, Kwong PD, Shaw GM. 2003. McKee K, ODell S, Perfetto S, Schmidt SD,
Antibody neutralization and escape by HIV-1. Shi W, Wu L, Yang Y, Yang ZY, Yang Z,
Nature 422:307312. Zhang Z, Bonsignori M, Crump JA, Kapiga
4. Kumar R, Tuen M, Liu J, Nadas A, Pan R, SH, Sam NE, Haynes BF, Simek M, Burton
Kong X, Hioe CE. 2013. Elicitation of broadly DR, Koff WC, Doria-Rose NA, Connors M,
reactive antibodies against glycan-modulated Mullikin JC, Nabel GJ, Roederer M, Shapiro
neutralizing V3 epitopes of HIV-1 by immune L, Kwong PD, Mascola JR. 2011. Focused
complex vaccines. Vaccine 31:54135421. evolution of HIV-1 neutralizing antibodies
5. Hioe CE, Kumar R, Hu S-L. 2014. The revealed by structures and deep sequencing.
influence of HIV envelope glycosylation on adap- Science 333:15931602.
tive immune response, p 5983. In Pantophlet R 14. Kwong PD, Doyle ML, Casper DJ, Cicala C,
(ed), HIV Glycans in Infection and Immunity. Leavitt SA, Majeed S, Steenbeke TD, Venturi
Springer Science + Business Media, New York, NY. M, Chaiken I, Fung M, Katinger H, Parren
6. Robertson DL, Sharp PM, McCutchan FE, PW, Robinson J, Van Ryk D, Wang L, Burton
Hahn BH. 1995. Recombination in HIV-1. DR, Freire E, Wyatt R, Sodroski J, Hendrickson
Nature 374:124126. WA, Arthos J. 2002. HIV-1 evades antibody-
7. Richman DD, Wrin T, Little SJ, Petropoulos mediated neutralization through conformational
CJ. 2003. Rapid evolution of the neutralizing masking of receptor-binding sites. Nature
antibody response to HIV type 1 infection. Proc 420:678682.
Natl Acad Sci USA 100:41444149. 15. Korkut A, Hendrickson WA. 2012. Structural
8. Moore PL, Ranchobe N, Lambson BE, Gray plasticity and conformational transitions of
ES, Cave E, Abrahams MR, Bandawe G, HIV envelope glycoprotein gp120. PloS One 7:
Mlisana K, Abdool Karim SS, Williamson C, e52170. doi:10.1371/journal.pone.0052170.
Morris L. 2009. Limited neutralizing antibody 16. Shrivastava IH, Wendel K, LaLonde JM.
specificities drive neutralization escape in 2012. Spontaneous rearrangement of the beta20/
early HIV-1 subtype C infection. PLoS Pathog beta21 strands in simulations of unliganded HIV-1
5:e1000598. doi:10.1371/journal.ppat.1000598. glycoprotein, gp120. Biochemistry 51:77837793.
9. Liao HX, Lynch R, Zhou T, Gao F, Alam SM, 17. Zolla-Pazner S. 2014. A critical question for
Boyd SD, Fire AZ, Roskin KM, Schramm CA, HIV vaccine development: which antibodies to
Zhang Z, Zhu J, Shapiro L, Mullikin JC, induce? Science 345:167168.
Gnanakaran S, Hraber P, Wiehe K, Kelsoe G, 18. Gorny MK, Pan R, Williams C, Wang XH,
Yang G, Xia SM, Montefiori DC, Parks R, Volsky B, ONeal T, Spurrier B, Sampson JM,
Lloyd KE, Scearce RM, Soderberg KA, Cohen Li L, Seaman MS, Kong XP, Zolla-Pazner S.
M, Kamanga G, Louder MK, Tran LM, Chen 2012. Functional and immunochemical cross-
Y, Cai F, Chen S, Moquin S, Du X, Joyce MG, reactivity of V2-specific monoclonal antibodies
Srivatsan S, Zhang B, Zheng A, Shaw GM, from HIV-1-infected individuals. Virology
Hahn BH, Kepler TB, Korber BT, Kwong PD, 427:198207.
202 MAYR AND ZOLLA-PAZNER

19. Prince AM, Reesink H, Pascual D, Horowitz analysis of an HIV-1 vaccine efficacy trial. N Engl
B, Hewlett I, Murthy KK, Cobb KE, Eichberg J Med 366:12751286.
JW. 1991. Prevention of HIV infection by 25. Zolla-Pazner S, deCamp AC, Cardozo T,
passive immunization with HIV immunoglob- Karasavvas N, Gottardo R, Williams C,
ulin. AIDS Res Hum Retroviruses 7:971973. Morris DE, Tomaras G, Rao M, Billings E,
20. Bar KJ, Tsao CY, Iyer SS, Decker JM, Yang Berman P, Shen X, Andrews C, OConnell RJ,
Y, Bonsignori M, Chen X, Hwang KK, Ngauy V, Nitayaphan S, de Souza M, Korber
Montefiori DC, Liao HX, Hraber P, Fischer B, Koup R, Bailer RT, Mascola JR, Pinter A,
W, Li H, Wang S, Sterrett S, Keele BF, Montefiori D, Haynes BF, Robb ML, Rerks-
Ganusov VV, Perelson AS, Korber BT, Ngarm S, Michael NL, Gilbert PB, Kim JH.
Georgiev I, McLellan JS, Pavlicek JW, Gao 2013. Analysis of V2 antibody responses in-
F, Haynes BF, Hahn BH, Kwong PD, Shaw duced in vaccinees in the ALVAC/AIDSVAX
GM. 2012. Early low-titer neutralizing anti- HIV-1 vaccine efficacy trial. PloS One 8:
bodies impede HIV-1 replication and select for e53629. doi:10.1371/journal.pone.0053629.
virus escape. PLoS Pathog 8:e1002721. doi:10.1371/ 26. Zolla-Pazner S, deCamp A, Gilbert PB,
journal.ppat.1002721. Williams C, Yates NL, Williams WT,
21. Hioe CE, Wrin T, Seaman MS, Yu X, Wood B, Howington R, Fong Y, Morris DE, Soderberg
Self S, Williams C, Gorny MK, Zolla-Pazner KA, Irene C, Reichman C, Pinter A, Parks R,
S. 2010. Anti-V3 monoclonal antibodies display Pitisuttithum P, Kaewkungwal J, Rerks-Ngarm
broad neutralizing activities against multiple S, Nitayaphan S, Andrews C, OConnell RJ,
HIV-1 subtypes. PloS One 5:e10254. doi:10.1371/ Yang ZY, Nabel GJ, Kim JH, Michael NL,
journal.pone.0010254. Montefiori DC, Liao HX, Haynes BF, Tomaras
22. Li L, Wang XH, Banerjee S, Volsky B, GD. 2014. Vaccine-induced IgG antibodies
Williams C, Virland D, Nadas A, Seaman to V1V2 regions of multiple HIV-1 subtypes
MS, Chen X, Spearman P, Zolla-Pazner S, correlate with decreased risk of HIV-1 infection.
Gorny MK. 2012. Different pattern of immu- PloS One 9:e87572. doi:10.1371/journal.pone.
noglobulin gene usage by HIV-1 compared to 0087572.
non-HIV-1 antibodies derived from the same 27. Yates NL, Liao HX, Fong Y, deCamp A,
infected subject. PloS One 7:e39534. doi:10.1371/ Vandergrift NA, Williams WT, Alam SM,
journal.pone.0039534. Ferrari G, Yang ZY, Seaton KE, Berman
23. Karasavvas N, Billings E, Rao M, Williams C, PW, Alpert MD, Evans DT, OConnell RJ,
Zolla-Pazner S, Bailer RT, Koup RA, Madnote S, Francis D, Sinangil F, Lee C, Nitayaphan S,
Arworn D, Shen X, Tomaras GD, Currier JR, Rerks-Ngarm S, Kaewkungwal J, Pitisuttithum
Jiang M, Magaret C, Andrews C, Gottardo R, P, Tartaglia J, Pinter A, Zolla-Pazner S, Gilbert
Gilbert P, Cardozo TJ, Rerks-Ngarm S, PB, Nabel GJ, Michael NL, Kim JH, Montefiori
Nitayaphan S, Pitisuttithum P, Kaewkungwal DC, Haynes BF, Tomaras GD. 2014. Vaccine-
J, Paris R, Greene K, Gao H, Gurunathan S, induced Env V1-V2 IgG3 correlates with lower
Tartaglia J, Sinangil F, Korber BT, Montefiori HIV-1 infection risk and declines soon after
DC, Mascola JR, Robb ML, Haynes BF, vaccination. Science Trans Med 6:228239.
Ngauy V, Michael NL, Kim JH, de Souza MS. 28. Gottardo R, Bailer RT, Korber BT, Gnanakaran
2012. The Thai Phase III HIV Type 1 Vaccine S, Phillips J, Shen X, Tomaras GD, Turk E,
trial (RV144) regimen induces antibodies that Imholte G, Eckler L, Wenschuh H, Zerweck J,
target conserved regions within the V2 loop Greene K, Gao H, Berman PW, Francis D,
of gp120. AIDS Res Hum Retroviruses 28:1444 Sinangil F, Lee C, Nitayaphan S, Rerks-Ngarm
1457. S, Kaewkungwal J, Pitisuttithum P, Tartaglia J,
24. Haynes BF, Gilbert PB, McElrath MJ, Zolla- Robb ML, Michael NL, Kim JH, Zolla-Pazner S,
Pazner S, Tomaras GD, Alam SM, Evans DT, Haynes BF, Mascola JR, Self S, Gilbert P,
Montefiori DC, Karnasuta C, Sutthent R, Montefiori DC. 2013. Plasma IgG to linear
Liao HX, DeVico AL, Lewis GK, Williams C, epitopes in the V2 and V3 regions of HIV-1
Pinter A, Fong Y, Janes H, DeCamp A, Huang gp120 correlate with a reduced risk of infection
Y, Rao M, Billings E, Karasavvas N, Robb in the RV144 vaccine efficacy trial. PloS One 8:
ML, Ngauy V, de Souza MS, Paris R, Ferrari e75665. doi:10.1371/journal.pone.0075665.
G, Bailer RT, Soderberg KA, Andrews C, 29. Montefiori DC, Karnasuta C, Huang Y,
Berman PW, Frahm N, De Rosa SC, Alpert MD, Ahmed H, Gilbert P, de Souza MS, McLinden
Yates NL, Shen X, Koup RA, Pitisuttithum P, R, Tovanabutra S, Laurence-Chenine A,
Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Sanders-Buell E, Moody MA, Bonsignori M,
Michael NL, Kim JH. 2012. Immune-correlates Ochsenbauer C, Kappes J, Tang H, Greene K,
Chapter 11 Antibodies Targeting the Envelope of HIV-1 203

Gao H, LaBranche CC, Andrews C, Polonis 35. Sather DN, Armann J, Ching LK, Mavrantoni
VR, Rerks-Ngarm S, Pitisuttithum P, A, Sellhorn G, Caldwell Z, Yu X, Wood B, Self
Nitayaphan S, Kaewkungwal J, Self SG, S, Kalams S, Stamatatos L. 2009. Factors
Berman PW, Francis D, Sinangil F, Lee C, associated with the development of cross-reactive
Tartaglia J, Robb ML, Haynes BF, Michael neutralizing antibodies during human immu-
NL, Kim JH. 2012. Magnitude and breadth nodeficiency virus type 1 infection. J Virol
of the neutralizing antibody response in the 83:757769.
RV144 and Vax003 HIV-1 vaccine efficacy 36. Simek MD, Rida W, Priddy FH, Pung P,
trials. J Infect Dis 206:431441. Carrow E, Laufer DS, Lehrman JK, Boaz M,
30. Liao HX, Bonsignori M, Alam SM, McLellan Tarragona-Fiol T, Miiro G, Birungi J,
JS, Tomaras GD, Moody MA, Kozink DM, Pozniak A, McPhee DA, Manigart O, Karita
Hwang KK, Chen X, Tsao CY, Liu P, Lu X, E, Inwoley A, Jaoko W, Dehovitz J, Bekker
Parks RJ, Montefiori DC, Ferrari G, Pollara LG, Pitisuttithum P, Paris R, Walker LM,
J, Rao M, Peachman KK, Santra S, Letvin NL, Poignard P, Wrin T, Fast PE, Burton DR,
Karasavvas N, Yang ZY, Dai K, Pancera M, Koff WC. 2009. Human immunodeficiency
Gorman J, Wiehe K, Nicely NI, Rerks-Ngarm S, virus type 1 elite neutralizers: individuals
Nitayaphan S, Kaewkungwal J, Pitisuttithum P, with broad and potent neutralizing activity
Tartaglia J, Sinangil F, Kim JH, Michael NL, identified by using a high-throughput neutral-
Kepler TB, Kwong PD, Mascola JR, Nabel GJ, ization assay together with an analytical selec-
Pinter A, Zolla-Pazner S, Haynes BF. 2013. tion algorithm. J Virol 83:73377348.
Vaccine induction of antibodies against a struc- 37. Walker LM, Phogat SK, Chan-Hui PY,
turally heterogeneous site of immune pressure Wagner D, Phung P, Goss JL, Wrin T,
within HIV-1 envelope protein variable regions 1 Simek MD, Fling S, Mitcham JL, Lehrman
and 2. Immunity 38:176186. JK, Priddy FH, Olsen OA, Frey SM,
31. Moody MA, Zhang R, Walter EB, Woods CW, Hammond PW, Kaminsky S, Zamb T, Moyle
Ginsburg GS, McClain MT, Denny TN, Chen M, Koff WC, Poignard P, Burton DR. 2009.
X, Munshaw S, Marshall DJ, Whitesides JF, Broad and potent neutralizing antibodies from
Drinker MS, Amos JD, Gurley TC, Eudailey an African donor reveal a new HIV-1 vaccine
JA, Foulger A, DeRosa KR, Parks R, target. Science 326:285289.
Meyerhoff RR, Yu JS, Kozink DM, Barefoot 38. Walker LM, Huber M, Doores KJ, Falkowska
BE, Ramsburg EA, Khurana S, Golding H, E, Pejchal R, Julien JP, Wang SK, Ramos A,
Vandergrift NA, Alam SM, Tomaras GD, Chan-Hui PY, Moyle M, Mitcham JL,
Kepler TB, Kelsoe G, Liao HX, Haynes BF. Hammond PW, Olsen OA, Phung P, Fling S,
2011. H3N2 influenza infection elicits more Wong CH, Phogat S, Wrin T, Simek MD, Koff
cross-reactive and less clonally expanded anti- WC, Wilson IA, Burton DR, Poignard P. 2011.
hemagglutinin antibodies than influenza vaccina- Broad neutralization coverage of HIV by
tion. PloS One 6:e25797. doi:10.1371/journal. multiple highly potent antibodies. Nature
pone.0025797. 477:466470.
32. Seaman MS, Janes H, Hawkins N, Grandpre 39. Wu X, Yang ZY, Li Y, Hogerkorp CM, Schief
LE, Devoy C, Giri A, Coffey RT, Harris L, WR, Seaman MS, Zhou T, Schmidt SD, Wu L,
Wood B, Daniels MG, Bhattacharya T, Xu L, Longo NS, McKee K, ODell S, Louder
Lapedes A, Polonis VR, McCutchan FE, MK, Wycuff DL, Feng Y, Nason M, Doria-
Gilbert PB, Self SG, Korber BT, Montefiori Rose N, Connors M, Kwong PD, Roederer M,
DC, Mascola JR. 2010. Tiered categorization Wyatt RT, Nabel GJ, Mascola JR. 2010.
of a diverse panel of HIV-1 Env pseudoviruses Rational design of envelope identifies broadly
for assessment of neutralizing antibodies. J neutralizing human monoclonal antibodies to
Virol 84:14391452. HIV-1. Science 329:856861.
33. Zolla-Pazner S, Kong XP, Jiang X, Cardozo 40. Huang J, Ofek G, Laub L, Louder MK, Doria-
T, Nadas A, Cohen S, Totrov M, Seaman MS, Rose NA, Longo NS, Imamichi H, Bailer RT,
Wang S, Lu S. 2011. Cross-clade HIV-1 neu- Chakrabarti B, Sharma SK, Alam SM, Wang
tralizing antibodies induced with V3-scaffold T, Yang Y, Zhang B, Migueles SA, Wyatt R,
protein immunogens following priming with Haynes BF, Kwong PD, Mascola JR, Connors
gp120 DNA. J Virol 85:98879898. M. 2012. Broad and potent neutralization of
34. Cardozo T, Wang S, Jiang X, Kong XP, Hioe HIV-1 by a gp41-specific human antibody.
C, Krachmarov C. 2014. Vaccine focusing to Nature 491:406412.
cross-subtype HIV-1 gp120 variable loop 41. Morris L, Chen X, Alam M, Tomaras G,
epitopes. Vaccine 32:49164924. Zhang R, Marshall DJ, Chen B, Parks R,
204 MAYR AND ZOLLA-PAZNER

Foulger A, Jaeger F, Donathan M, Bilska M, serum levels giving complete neutralization


Gray ES, Abdool Karim SS, Kepler TB, in vitro. J Virol 75:83408347.
Whitesides J, Montefiori D, Moody MA, 50. Hessell AJ, Rakasz EG, Poignard P, Hangartner
Liao HX, Haynes BF. 2011. Isolation of a L, Landucci G, Forthal DN, Koff WC, Watkins
human anti-HIV gp41 membrane proximal region DI, Burton DR. 2009. Broadly neutralizing
neutralizing antibody by antigen-specific single human anti-HIV antibody 2G12 is effective in
B cell sorting. PloS One 6:e23532. doi:10.1371/ protection against mucosal SHIV challenge even
journal.pone.0023532. at low serum neutralizing titers. PLoS Pathog 5:
42. Klein F, Diskin R, Scheid JF, Gaebler C, e1000433. doi:10.1371/journal.ppat.1000433.
Mouquet H, Georgiev IS, Pancera M, Zhou T, 51. Hessell AJ, Rakasz EG, Tehrani DM, Huber
Incesu RB, Fu BZ, Gnanapragasam PN, M, Weisgrau KL, Landucci G, Forthal DN,
Oliveira TY, Seaman MS, Kwong PD, Koff WC, Poignard P, Watkins DI, Burton
Bjorkman PJ, Nussenzweig MC. 2013. Somat- DR. 2010. Broadly neutralizing monoclonal
ic mutations of the immunoglobulin frame- antibodies 2F5 and 4E10 directed against the
work are generally required for broad and human immunodeficiency virus type 1 gp41
potent HIV-1 neutralization. Cell 153:126138. membrane-proximal external region protect
43. Sok D, Laserson U, Laserson J, Liu Y, against mucosal challenge by simian-human
Vigneault F, Julien JP, Briney B, Ramos A, immunodeficiency virus SHIVBa-L. J Virol
Saye KF, Le K, Mahan A, Wang S, Kardar M, 84:13021313.
Yaari G, Walker LM, Simen BB, St John EP, 52. Haynes BF, Verkoczy L. 2014. AIDS/HIV.
Chan-Hui PY, Swiderek K, Kleinstein SH, Host controls of HIV neutralizing antibodies.
Alter G, Seaman MS, Chakraborty AK, Koller Science 344:588589.
D, Wilson IA, Church GM, Burton DR, 53. Pegu A, Yang ZY, Boyington JC, Wu L, Ko
Poignard P. 2013. The effects of somatic SY, Schmidt SD, McKee K, Kong WP, Shi W,
hypermutation on neutralization and binding Chen X, Todd JP, Letvin NL, Huang J, Nason
in the PGT121 family of broadly neutralizing MC, Hoxie JA, Kwong PD, Connors M, Rao
HIV antibodies. PLoS Pathog 9:e1003754. SS, Mascola JR, Nabel GJ. 2014. Neutralizing
doi:10.1371/journal.ppat.1003754. antibodies to HIV-1 envelope protect more
44. Mascola JR, Haynes BF. 2013. HIV-1 neutral- effectively in vivo than those to the CD4
izing antibodies: understanding natures pathways. receptor. Sci Trans Med 6:243288.
Immunol Rev 254:225244. 54. Shingai M, Donau OK, Plishka RJ, Buckler-
45. Yu L, Guan Y. 2014. Immunologic basis for White A, Mascola JR, Nabel GJ, Nason MC,
long HCDR3s in broadly neutralizing anti- Montefiori D, Moldt B, Poignard P, Diskin R,
bodies against HIV-1. Front Immunol 5:250. Bjorkman PJ, Eckhaus MA, Klein F, Mouquet
46. Verkoczy L, Diaz M. 2014. Autoreactivity in H, Cetrulo Lorenzi JC, Gazumyan A, Burton
HIV-1 broadly neutralizing antibodies: impli- DR, Nussenzweig MC, Martin MA, Nishimura
cations for their function and induction by Y. 2014. Passive transfer of modest titers of potent
vaccination. Curr Opin HIV AIDS 9:224234. and broadly neutralizing anti-HIV monoclonal
47. Haynes BF, Fleming J, St Clair EW, Katinger antibodies block SHIV infection in macaques.
H, Stiegler G, Kunert R, Robinson J, Scearce J Exp Med 211:20612074.
RM, Plonk K, Staats HF, Ortel TL, Liao HX, 55. Watkins JD, Siddappa NB, Lakhashe SK,
Alam SM. 2005. Cardiolipin polyspecific Humbert M, Sholukh A, Hemashettar G,
autoreactivity in two broadly neutralizing HIV-1 Wong YL, Yoon JK, Wang W, Novembre
antibodies. Science 308:19061908. FJ, Villinger F, Ibegbu C, Patel K, Corti D,
48. Moldt B, Rakasz EG, Schultz N, Chan-Hui Agatic G, Vanzetta F, Bianchi S, Heeney JL,
PY, Swiderek K, Weisgrau KL, Piaskowski Sallusto F, Lanzavecchia A, Ruprecht RM.
SM, Bergman Z, Watkins DI, Poignard P, 2011. An anti-HIV-1 V3 loop antibody fully
Burton DR. 2012. Highly potent HIV-specific protects cross-clade and elicits T-cell immunity
antibody neutralization in vitro translates into in macaques mucosally challenged with an R5
effective protection against mucosal SHIV clade C SHIV. PloS One 6:e18207. doi:10.1371/
challenge in vivo. Proc Natl Acad Sci USA journal.pone.0018207.
109:1892118925. 56. Halper-Stromberg A, Lu CL, Klein F,
49. Parren PW, Marx PA, Hessell AJ, Luckay A, Horwitz JA, Bournazos S, Nogueira L,
Harouse J, Cheng-Mayer C, Moore JP, Eisenreich TR, Liu C, Gazumyan A, Schaefer
Burton DR. 2001. Antibody protects macaques U, Furze RC, Seaman MS, Prinjha R,
against vaginal challenge with a pathogenic R5 Tarakhovsky A, Ravetch JV, Nussenzweig
simian/human immunodeficiency virus at MC. 2014. Broadly neutralizing antibodies and
Chapter 11 Antibodies Targeting the Envelope of HIV-1 205

viral inducers decrease rebound from HIV-1 64. Burton DR, Desrosiers RC, Doms RW, Koff
latent reservoirs in humanized mice. Cell WC, Kwong PD, Moore JP, Nabel GJ,
158:989999. Sodroski J, Wilson IA, Wyatt RT. 2004.
57. Barouch DH, Whitney JB, Moldt B, Klein F, HIV vaccine design and the neutralizing
Oliveira TY, Liu J, Stephenson KE, Chang antibody problem. Nat Immunol 5:233236.
HW, Shekhar K, Gupta S, Nkolola JP, Sea- 65. Sanders RW, Vesanen M, Schuelke N, Master
man MS, Smith KM, Borducchi EN, Cabral C, A, Schiffner L, Kalyanaraman R, Paluch M,
Smith JY, Blackmore S, Sanisetty S, Perry Berkhout B, Maddon PJ, Olson WC, Lu M,
JR, Beck M, Lewis MG, Rinaldi W, Moore JP. 2002. Stabilization of the soluble,
Chakraborty AK, Poignard P, Nussenzweig cleaved, trimeric form of the envelope glyco-
MC, Burton DR. 2013. Therapeutic efficacy of protein complex of human immunodeficiency
potent neutralizing HIV-1-specific monoclonal virus type 1. J Virol 76:88758889.
antibodies in SHIV-infected rhesus monkeys. 66. Binley JM, Sanders RW, Clas B, Schuelke N,
Nature 503:224228. Master A, Guo Y, Kajumo F, Anselma DJ,
58. Klein F, Nogueira L, Nishimura Y, Phad G, Maddon PJ, Olson WC, Moore JP. 2000. A
West AP Jr, Halper-Stromberg A, Horwitz recombinant human immunodeficiency virus
JA, Gazumyan A, Liu C, Eisenreich TR, type 1 envelope glycoprotein complex stabilized
Lehmann C, Fatkenheuer G, Shingai M, by an intermolecular disulfide bond between the
Martin M, Bjorkman PJ, Seaman MS, Zolla- gp120 and gp41 subunits is an antigenic mimic of
Pazner S, Hedestam G, Nussenzweig MC. the trimeric virion-associated structure. J Virol
2014. Enhanced HIV-1 immunotherapy by 74:627643.
naturally arising antibodies targeting resistant 67. Yang X, Lee J, Mahony EM, Kwong PD,
variants. J Exp Med 211:23612372. Wyatt R, Sodroski J. 2002. Highly stable
59. Wu X, Parast AB, Richardson BA, Nduati R, trimers formed by human immunodeficiency
John-Stewart G, Mbori-Ngacha D, Rainwater virus type 1 envelope glycoproteins fused with
SM, Overbaugh J. 2006. Neutralization escape the trimeric motif of T4 bacteriophage fibritin.
variants of human immunodeficiency virus type 1 J Virol 76:46344642.
are transmitted from mother to infant. J Virol 68. Spearman P, Lally MA, Elizaga M, Montefiori
80:835844. D, Tomaras GD, McElrath MJ, Hural J, De Rosa
60. Mabuka J, Goo L, Omenda MM, Nduati R, SC, Sato A, Huang Y, Frey SE, Sato P, Donnelly
Overbaugh J. 2013. HIV-1 maternal and infant J, Barnett S, Corey LJ. 2011. A trimeric, V2-
variants show similar sensitivity to broadly neu- deleted HIV-1 envelope glycoprotein vaccine
tralizing antibodies, but sensitivity varies by elicits potent neutralizing antibodies but limited
subtype. AIDS 27:15351544. breadth of neutralization in human volunteers.
61. Flynn NM, Forthal DN, Harro CD, Judson J Infect Dis 203:11651173.
FN, Mayer KH, Para MF. 2005. Placebo- 69. Forsell MN, Schief WR, Wyatt RT. 2009.
controlled phase 3 trial of a recombinant Immunogenicity of HIV-1 envelope glycoprotein
glycoprotein 120 vaccine to prevent HIV-1 oligomers. Curr Opin HIV AIDS 4:380387.
infection. J Infect Dis 191:654665. 70. Sather DN, Carbonetti S, Malherbe D,
62. Pitisuttithum P, Gilbert P, Gurwith M, Pissani F, Stuart AB, Hessell AJ, Gray MD,
Heyward W, Martin M, van Griensven F, M ik ell I, Kalam s S A , Haigw o o d NL ,
Hu D, Tappero JW, Choopanya K. 2006. Stamatatos L. 2014. Emergence of broadly
Randomized, double-blind, placebo-controlled neutralizing antibodies and viral co-evolution
efficacy trial of a bivalent recombinant glyco- in two subjects during the early stages of infection
protein 120 HIV-1 vaccine among injection with the human immunodeficiency virus type 1.
drug users in Bangkok, Thailand. J Infect Dis J Virol 88:1296812981.
194:16611671. 71. Doria-Rose NA, Schramm CA, Gorman J,
63. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan Moore PL, Bhiman JN, DeKosky BJ,
S, Kaewkungwal J, Chiu J, Paris R, Premsri Ernandes MJ, Georgiev IS, Kim HJ, Pancera
N, Namwat C, de Souza M, Adams E, M, Staupe RP, Altae-Tran HR, Bailer RT,
Benenson M, Gurunathan S, Tartaglia J, Crooks ET, Cupo A, Druz A, Garrett NJ, Hoi
McNeil JG, Francis DP, Stablein D, Birx KH, Kong R, Louder MK, Longo NS, McKee
DL, Chunsuttiwat S, Khamboonruang C, K, Nonyane M, ODell S, Roark RS, Rudicell
Thongcharoen P, Robb ML, Michael NL, RS, Schmidt SD, Sheward DJ, Soto C,
Kunasol P, Kim JH. 2009. Vaccination with Wibmer CK, Yang Y, Zhang Z, Mullikin JC,
ALVAC and AIDSVAX to prevent HIV-1 infection Binley JM, Sanders RW, Wilson IA, Moore
in Thailand. N Engl J Med 361:22092220. JP, Ward AB, Georgiou G, Williamson C,
206 MAYR AND ZOLLA-PAZNER

Abdool Karim SS, Morris L, Kwong PD, Shapiro 79. Arthos J, Cicala C, Martinelli E, Macleod K, Van
L, Mascola JR. 2014. Developmental pathway for Ryk D, Wei D, Xiao Z, Veenstra TD, Conrad TP,
potent V1V2-directed HIV-neutralizing anti- Lempicki RA, McLaughlin S, Pascuccio M,
bodies. Nature 509:5562. Gopaul R, McNally J, Cruz CC, Censoplano N,
72. Correia BE, Bates JT, Loomis RJ, Baneyx G, Chung E, Reitano KN, Kottilil S, Goode DJ,
Carrico C, Jardine JG, Rupert P, Correnti C, Fauci AS. 2008. HIV-1 envelope protein binds to
Kalyuzhniy O, Vittal V, Connell MJ, Stevens and signals through integrin alpha4beta7, the gut
E, Schroeter A, Chen M, Macpherson S, mucosal homing receptor for peripheral T cells.
Serra AM, Adachi Y, Holmes MA, Li Y, Nat Immunol 9:301309.
Klevit RE, Graham BS, Wyatt RT, Baker D, 80. Gorny MK, Moore JP, Conley AJ, Karwowska
Strong RK, Crowe JE Jr, Johnson PR, Schief S, Sodroski J, Williams C, Burda S, Boots LJ,
WR. 2014. Proof of principle for epitope- Zolla-Pazner S. 1994. Human anti-V2 monoclo-
focused vaccine design. Nature 507:201206. nal antibody that neutralizes primary but not
73. Delany I, Rappuoli R, Seib KL. 2013. Vaccines, laboratory isolates of human immunodeficiency
reverse vaccinology, and bacterial pathogenesis. virus type 1. J Virol 68:83128320.
Cold Spring Harbor Perspect Med 3:a012476. 81. Mayr LM, Cohen S, Spurrier B, Kong XP,
74. Zhou T, Xu L, Dey B, Hessell AJ, Van Ryk D, Zolla-Pazner S. 2013. Epitope mapping of
Xiang SH, Yang X, Zhang MY, Zwick MB, conformational V2-specific anti-HIV human mono-
Arthos J, Burton DR, Dimitrov DS, Sodroski clonal antibodies reveals an immunodominant
J, Wyatt R, Nabel GJ, Kwong PD. 2007. site in V2. PloS One 8:e70859. doi:10.1371/journal.
Structural definition of a conserved neutralization pone.0070859.
epitope on HIV-1 gp120. Nature 445:732737. 82. Bonsignori M, Pollara J, Moody MA, Alpert
75. Cimbro R, Gallant TR, Dolan MA, Guzzo MD, Chen X, Hwang KK, Gilbert PB, Huang
C, Zhang P, Lin Y, Miao H, Van Ryk D, Y, Gurley TC, Kozink DM, Marshall DJ,
Arthos J, Gorshkova I, Brown PH, Hurt DE, Whitesides JF, Tsao CY, Kaewkungwal J,
Lusso P. 2014. Tyrosine sulfation in the second Nitayaphan S, Pitisuttithum P, Rerks-Ngarm
variable loop (V2) of HIV-1 gp120 stabilizes S, Kim JH, Michael NL, Tomaras GD, Montefiori
V2-V3 interaction and modulates neutraliza- DC, Lewis GK, DeVico A, Evans DT, Ferrari G,
tion sensitivity. Proc Natl Acad Sci USA Liao HX, Haynes BF. 2012. Antibody-dependent
111:31523157. cellular cytotoxicity-mediating antibodies from
76. White TA, Bartesaghi A, Borgnia MJ, Meyerson an HIV-1 vaccine efficacy trial target multiple
JR, de la Cruz MJ, Bess JW, Nandwani R, Hoxie epitopes and preferentially use the VH1 gene
JA, Lifson JD, Milne JL, Subramaniam S. 2010. family. J Virol 86:1152111532.
Molecular architectures of trimeric SIV and HIV- 83. Gorny MK, Stamatatos L, Volsky B, Revesz
1 envelope glycoproteins on intact viruses: strain- K , W i ll i a m s C , W a n g X H , C o h e n S ,
dependent variation in quaternary structure. Staudinger R, Zolla-Pazner S. 2005. Identifi-
PLoS Pathog 6:e1001249. doi:10.1371/journal. cation of a new quaternary neutralizing epi-
ppat.1001249. tope on human immunodeficiency virus type 1
77. Zolla-Pazner S, Cardozo T. 2011. Structure- virus particles. J Virol 79:52325237.
function relationships of HIV-1 envelope se- 84. Bonsignori M, Hwang KK, Chen X, Tsao CY,
quence-variable regions refocus vaccine design. Morris L, Gray E, Marshall DJ, Crump JA,
Nat Rev 10:527535. Kapiga SH, Sam NE, Sinangil F, Pancera M,
78. McLellan JS, Pancera M, Carrico C, Gorman Yongping Y, Zhang B, Zhu J, Kwong PD, ODell
J, Julien JP, Khayat R, Louder R, Pejchal R, S, Mascola JR, Wu L, Nabel GJ, Phogat S,
Sastry M, Dai K, ODell S, Patel N, Shahzad- Seaman MS, Whitesides JF, Moody MA, Kelsoe
ul-Hussan S, Yang Y, Zhang B, Zhou T, Zhu G, Yang X, Sodroski J, Shaw GM, Montefiori
J, Boyington JC, Chuang GY, Diwanji D, DC, Kepler TB, Tomaras GD, Alam SM, Liao
Georgiev I, Kwon YD, Lee D, Louder MK, HX, Haynes BF. 2011. Analysis of a clonal lineage
Moquin S, Schmidt SD, Yang ZY, Bonsignori of HIV-1 envelope V2/V3 conformational epi-
M, Crump JA, Kapiga SH, Sam NE, Haynes tope-specific broadly neutralizing antibodies
BF, Burton DR, Koff WC, Walker LM, and their inferred unmutated common ancestors.
Phogat S, Wyatt R, Orwenyo J, Wang LX, J Virol 85:999810009.
Arthos J, Bewley CA, Mascola JR, Nabel GJ, 85. Julien JP, Cupo A, Sok D, Stanfield RL,
Schief WR, Ward AB, Wilson IA, Kwong PD. Lyumkis D, Deller MC, Klasse PJ, Burton
2011. Structure of HIV-1 gp120 V1/V2 domain DR, Sanders RW, Moore JP, Ward AB, Wilson
with broadly neutralizing antibody PG9. Nature IA. 2013. Crystal structure of a soluble cleaved
480:336343. HIV-1 envelope trimer. Science 342:14771483.
Chapter 11 Antibodies Targeting the Envelope of HIV-1 207

86. Ivanoff LA, Dubay JW, Morris JF, Roberts can mimic the antigen-binding modes of anti-
SJ, Gutshall L, Sternberg EJ, Hunter E, bodies derived from HIV-1-infected humans.
Matthews TJ, Petteway SR Jr. 1992. V3 loop J Virol 87:1022110231.
region of the HIV-1 gp120 envelope protein is 97. Burke V, Williams C, Sukumaran M, Kim SS,
essential for virus infectivity. Virology 187:423432. Li H, Wang XH, Gorny MK, Zolla-Pazner S,
87. Zolla-Pazner S. 2004. Identifying epitopes of Kong XP. 2009. Structural basis of the cross-
HIV-1 that induce protective antibodies. Nat reactivity of genetically related human anti-HIV-1
Rev 4:199210. mAbs: implications for design of V3-based immu-
88. Zolla-Pazner S, Cardozo T. 2010. Structure- nogens. Structure 17:15381546.
function relationships of HIV-1 envelope se- 98. Mouquet H, Klein F, Scheid JF, Warncke M,
quence-variable regions refocus vaccine design. Pietzsch J, Oliveira TY, Velinzon K, Seaman
Nat Rev 10:527535. MS, Nussenzweig MC. 2011. Memory B cell
89. Jiang X, Burke V, Totrov M, Williams C, antibodies to HIV-1 gp140 cloned from individu-
Cardozo T, Gorny MK, Zolla-Pazner S, Kong als infected with clade Aand B viruses. PloS One 6:
XP. 2010. Conserved structural elements in the e24078. doi:10.1371/journal.pone.0024078.
V3 crown of HIV-1 gp120. Nat Struct Mol Biol 99. Corti D, Langedijk JP, Hinz A, Seaman MS,
17:955961. Vanzetta F, Fernandez-Rodriguez BM, Silacci
90. Huang CC, Tang M, Zhang MY, Majeed S, C, Pinna D, Jarrossay D, Balla-Jhagjhoorsingh
Montabana E, Stanfield RL, Dimitrov DS, S, Willems B, Zekveld MJ, Dreja H, OSullivan
Korber B, Sodroski J, Wilson IA, Wyatt R, E, Pade C, Orkin C, Jeffs SA, Montefiori DC,
Kwong PD. 2005. Structure of a V3-containing Davis D, Weissenhorn W, McKnight A, Heeney
HIV-1 gp120 core. Science 310:10251028. JL, Sallusto F, Sattentau QJ, Weiss RA,
91. Zolla-Pazner S. 2005. Improving on nature: Lanzavecchia A. 2010. Analysis of memory B
focusing the immune response on the V3 loop. cell responses and isolation of novel mono-
Hum Antibodies 14:6972. clonal antibodies with neutralizing breadth from
92. Crooks ET, Moore PL, Franti M, Cayanan HIV-1-infected individuals. PloS One 5:e8805.
CS, Zhu P, Jiang P, de Vries RP, Wiley C, doi:10.1371/journal.pone.0008805.
Zharkikh I, Schulke N, Roux KH, Montefiori 100. McCaffrey RA, Saunders C, Hensel M,
DC, Burton DR, Binley JM. 2007. A compar- Stamatatos L. 2004. N-linked glycosylation
ative immunogenicity study of HIV-1 virus-like of the V3 loop and the immunologically silent
particles bearing various forms of envelope face of gp120 protects human immunodefi-
proteins, particles bearing no envelope and solu- ciency virus type 1 SF162 from neutralization
ble monomeric gp120. Virology 366:245262. by anti-gp120 and anti-gp41 antibodies. J Virol
93. Visciano ML, Tagliamonte M, Stewart-Jones 78:32793295.
G, Heyndrickx L, Vanham G, Jansson M, 101. Wyatt R, Moore J, Accola M, Desjardin E,
Fomsgaard A, Grevstad B, Ramaswamy M, Robinson J, Sodroski J. 1995. Involvement
Buonaguro FM, Tornesello ML, Biswas P, of the V1/V2 variable loop structure in the
Scarlatti G, Buonaguro L. 2013. Characteriza- exposure of human immunodeficiency virus
tion of humoral responses to soluble trimeric type 1 gp120 epitopes induced by receptor
HIV gp140 from a clade A Ugandan field isolate. binding. J Virol 69:57235733.
J Transl Med 11:165. 102. Gzyl J, Bolesta E, Wierzbicki A, Kmieciak D,
94. Totrov M, Jiang X, Kong XP, Cohen S, Naito T, Honda M, Komuro K, Kaneko Y,
Krachmarov C, Salomon A, Williams C, Kozbor D. 2004. Effect of partial and complete
Seaman MS, Abagyan R, Cardozo T, Gorny variable loop deletions of the human immuno-
MK, Wang S, Lu S, Pinter A, Zolla-Pazner S. deficiency virus type 1 envelope glycoprotein
2010. Structure-guided design and immunological on the breadth of gp160-specific immune
characterization of immunogens presenting the responses. Virology 318:493506.
HIV-1 gp120 V3 loop on a CTB scaffold. Virology 103. Upadhyay C, Mayr LM, Zhang J, Kumar R,
405:513523. Gorny MK, Nadas A, Zolla-Pazner S, Hioe CE.
95. Zolla-Pazner S, Cohen S, Pinter A, Krachmarov 2014. Distinct mechanisms regulate exposure of
C, Wrin T, Wang S, Lu S. 2009. Cross-clade neutralizing epitopes in the V2 and V3 loops of
neutralizing antibodies against HIV-1 induced in HIV-1 envelope. J Virol 88:1285312865.
rabbits by focusing the immune response on a 104. Pejchal R, Doores KJ, Walker LM, Khayat R,
neutralizing epitope. Virology 392:8293. Huang PS, Wang SK, Stanfield RL, Julien JP,
96. Pan R, Sampson JM, Chen Y, Vaine M, Wang Ramos A, Crispin M, Depetris R, Katpally U,
S, Lu S, Kong XP. 2013. Rabbit anti-HIV-1 Marozsan A, Cupo A, Maloveste S, Liu Y,
monoclonal antibodies raised by immunization McBride R, Ito Y, Sanders RW, Ogohara C,
208 MAYR AND ZOLLA-PAZNER

Paulson JC, Feizi T, Scanlan CN, Wong CH, HIV-1 Env trimers, uncleaved Env proteins,
Moore JP, Olson WC, Ward AB, Poignard P, and monomeric subunits. Retrovirology 11:41.
Schief WR, Burton DR, Wilson IA. 2011. A 111. Bhattacharyya S, Singh P, Rathore U, Purwar M,
potent and broad neutralizing antibody recog- Wagner D, Arendt H, DeStefano J, LaBranche
nizes and penetrates the HIV glycan shield. CC, Montefiori DC, Phogat S, Varadarajan R.
Science 334:10971103. 2013. Design of an Escherichia coli expressed HIV-1
105. Julien JP, Sok D, Khayat R, Lee JH, Doores gp120 fragment immunogen that binds to b12 and
KJ, Walker LM, Ramos A, Diwanji DC, induces broad and potent neutralizing antibodies.
Pejchal R, Cupo A, Katpally U, Depetris RS, J Biol Chem 288:98159825.
Stanfield RL, McBride R, Marozsan AJ, 112. Montero M, van Houten NE, Wang X, Scott
Paulson JC, Sanders RW, Moore JP, Burton JK. 2008. The membrane-proximal external
DR, Poignard P, Ward AB, Wilson IA. 2013. region of the human immunodeficiency virus type
Broadly neutralizing antibody PGT121 alloste- 1 envelope: dominant site of antibody neutraliza-
rically modulates CD4 binding via recognition tion and target for vaccine design. Microbiol Mol
of the HIV-1 gp120 V3 base and multiple Biol Rev 72:5484.
surrounding glycans. PLoS Pathog 9:e1003342. 113. Kim M, Sun ZY, Rand KD, Shi X, Song L,
doi:10.1371/journal.ppat.1003342. Cheng Y, Fahmy AF, Majumdar S, Ofek G,
106. Wu X, Zhou T, ODell S, Wyatt RT, Kwong Yang Y, Kwong PD, Wang JH, Engen JR,
PD, Mascola JR. 2009. Mechanism of human Wagner G, Reinherz EL. 2011. Antibody
immunodeficiency virus type 1 resistance to mechanics on a membrane-bound HIV seg-
monoclonal antibody B12 that effectively ment essential for GP41-targeted viral neutral-
targets the site of CD4 attachment. J Virol ization. Nat Struct Mol Biol 18:12351243.
83:1089210907. 114. Song L, Sun ZY, Coleman KE, Zwick MB,
107. Chen L, Kwon YD, Zhou T, Wu X, ODell S, Gach JS, Wang JH, Reinherz EL, Wagner G,
Cavacini L, Hessell AJ, Pancera M, Tang M, Kim M. 2009. Broadly neutralizing anti-HIV-1
Xu L, Yang ZY, Zhang MY, Arthos J, Burton antibodies disrupt a hinge-related function of
DR, Dimitrov DS, Nabel GJ, Posner MR, gp41 at the membrane interface. Proc Natl
Sodroski J, Wyatt R, Mascola JR, Kwong Acad Sci USA 106:90579062.
PD. 2009. Structural basis of immune evasion 115. Dennison SM, Sutherland LL, Jaeger FH,
at the site of CD4 attachment on HIV-1 gp120. Anasti KM, Parks R, Stewart S, Bowman C,
Science 326:11231127. Xia SM, Zhang R, Shen X, Scearce RM, Ofek G,
108. Selvarajah S, Puffer B, Pantophlet R, Law M, Yang Y, Kwong PD, Santra S, Liao HX, Tomaras
Doms RW, Burton DR. 2005. Comparing G, Letvin NL, Chen B, Alam SM, Haynes BF.
antigenicity and immunogenicity of engineered 2011. Induction of antibodies in rhesus macaques
gp120. J Virol 79:1214812163. that recognize a fusion-intermediate conforma-
109. Ahmed FK, Clark BE, Burton DR, Pantophlet tion of HIV-1 gp41. PloS One 6:e27824. doi:10.1371/
R. 2012. An engineered mutant of HIV-1 gp120 journal.pone.0027824.
formulated with adjuvant Quil A promotes 116. Shen X, Parks RJ, Montefiori DC, Kirchherr
elicitation of antibody responses overlapping JL, Keele BF, Decker JM, Blattner WA, Gao
the CD4-binding site. Vaccine 30:922930. F, Weinhold KJ, Hicks CB, Greenberg ML,
110. Yasmeen A, Ringe R, Derking R, Cupo A, Hahn BH, Shaw GM, Haynes BF, Tomaras
Julien JP, Burton DR, Ward AB, Wilson IA, GD. 2009. In vivo gp41 antibodies targeting the
Sanders RW, Moore JP, Klasse PJ. 2014. 2F5 monoclonal antibody epitope mediate
Differential binding of neutralizing and non- human immunodeficiency virus type 1 neutral-
neutralizing antibodies to native-like soluble ization breadth. J Virol 83:36173625.
Committing the Oldest Sins in the
Newest Kind of WaysAntibodies
Targeting the Inuenza Virus Type A
Hemagglutinin Globular Head

JENS C. KRAUSE1 and JAMES E. CROWE, JR.2


12
INFLUENZA HEMAGGLUTININ IS A TRIMERIC GLYCOPROTEIN
OF THREE IDENTICAL SUBUNITS AND THE MAJOR PROTECTIVE
ANTIGEN OF THE VIRUS

Influenza hemagglutinin (HA) is the major glycoprotein on the surface


of influenza virions. It mediates receptor binding and fusion. The surface
glycoprotein neuraminidase (NA) is a receptor-destroying enzyme. Even though
humoral immunity to NA and other proteins and cellular immunity to several
viral proteins contribute to protection against influenza infection, neutralizing
antibodies directed against influenza HA are sufficient to protect against disease.
The H3 HA crystal structure was solved in 1981 at 3- resolution (1). Since then,
the crystal structures of HA molecules from H2, H5, H7, and several different H1
strains including the pandemic 1918 H1 and the pandemic 2009 H1 (2) have been
determined. In brief, HA is a trimeric type I membrane glycoprotein made of
three identical subunits (Fig. 1). Each subunit is synthesized as an HA0 precursor
and cleaved proteolytically into an HA1 subunit that composes the membrane-
distal globular head and part of the membrane-proximal stem region, and an
HA2 subunit that only contributes to the stem region (Fig. 1).

1
Childrens Hospital, University of Freiburg Medical Center, 79106 Freiburg, Germany; 2Vanderbilt Vaccine
Center, Vanderbilt University Medical Center, Nashville, TN 37232-0417.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0021-2014

209
210 KRAUSE AND CROWE

argue that, in fact, the boosting of antibodies to


conserved epitopes is consistent with the
conventional understanding of B-cell memory.
The original antigenic sin theory suggests that,
after repeated antigenic exposure, the antibody
response is biased toward epitopes that were
present in the original antigenic exposure and
against new related antigens that cause subse-
quent exposures. Some people have hypothe-
sized that such cross-reactive stem antibodies
lead to prepandemic influenza strains becom-
ing extinct (11). However, the frequency of
antibodies to conserved epitopes on the glob-
ular head of HA also appeared to be increased
FIGURE 1 Space-lling model of inuenza hemagglu-
after the 2009 H1N1 pandemic (12, 13, 14, 15).
tinin HA based on PDB 1RD8 (38). The three protomers Such antibodies also could have limited the
of the HA trimer are colored white, gray, or black. circulation of prepandemic H1N1 strains. We
doi:10.1128/microbiolspec.AID-0021-2014.f1 have cloned mAbs to the H2N2 virus that
circulated in humans from 1957 until 1968 from
the peripheral blood of healthy donors of an
The receptor-binding domain (RBD) of appropriate age to have had exposure to H2
HA forms a conserved pocket on the glob- viruses (16). These mAbs were directed against
ular head (Fig. 1). Antibodies that target the highly conserved epitopes on the head of HA
RBD or adjacent hypervariable loops can (16, 17). If such antibodies were present on a
inhibit hemagglutination of red blood cells population level, they probably would limit the
in vitro and binding to sialic acid of respi- circulation of H2 viruses in the human popu-
ratory epithelial cells (Table 1). In contrast, lation even more so than stem antibodies
neutralizing antibodies against the HA stem
domain can prevent the pH-induced confor-
mational change that occurs in the endosome
after binding to and internalization into the TABLE 1 Characteristics of globular head versus
stem HA antibodies
host cell or by interfering with the maturation
Globular head
of HA0 to HA1 and HA2 (3, 4). After the Characteristics antibodies Stem antibodies
discovery of the murine HA stem-specific
Mechanism Inhibition of Inhibition of
monoclonal antibody (mAb) C179 in 1993 (5) of action receptor binding, pH-induced
that neutralizes H1, H2, and H5 viruses, several inhibition of conformational
groups have reported that humans can gener- pH-induced change,
ate mAbs against the influenza HA stem as conformational interference of
well, many of which are encoded by the VH1-69 change (some) HA0 maturation
(some)
germ line gene that specifies amino acids that Inhibition of +++
seem ideally configured to reach into a hydro- hemagglutination
phobic pocket on the stem. The frequency of Neutralization ++ / +++ + / ++
antibodies to the conserved HA stem region activity
may have been increased in infected individu- Cross-reactive + / ++ (RBD) +++
potential
als after the 2009 H1N1 pandemic (6). This Frequency +++ + (higher after
observation may represent a special case of heterologous
original antigenic sin (7, 8, 9, 10), much talked challenge)
about in the influenza field, although one might In vivo activity +++ + / ++
CHAPTER 12 Committing the Oldest Sins in the Newest Kind of Ways 211

because of their vastly superior potency. In reactivity between those two strains (2, 20, 21).
general, HA globular head antibodies seem to Antibodies that recognized multiple antigenic
be more frequent than stem antibodies proba- sites of the globular head of HA had already
bly because their epitope is more readily been identified in the 1980s (19). More recent
accessible on the surface of the virus and can X-ray cocrystallization experiments have
be targeted by a wide diversity of antibodies shown subsequently that antibodies, such as
encoded by a broad variety of antibody germ 2D1 that is oriented above the center of the Sa
line genes (Table 1). site with its large surface area, recognize
A lot of interest has been devoted recently residues outside the Sa epitope and even
toward presenting this conserved HA stem residues within neighboring antigenic sites
region as part of a universal vaccine against (2). Human antibodies that bind a single,
influenza because of the exciting isolation of discrete antigenic loop of a conventionally
cross-neutralizing antibodies directed against defined site on the HA head domain are
the conserved HA stem region (18). With the probably the exception rather than the rule.
recent discovery of several cross-neutralizing The antigenic mapping of murine antibodies
HA globular head antibodies, the HA globular may not translate directly to that of human
head also appears as an attractive target for antibodies, since the antibody repertoires of
vaccine design. This finding seems particularly different species use entirely different germ
important because the access of antibodies line genes. Still, it can helpful to use the
to the globular head is not hindered by steric antigenic mapping terms in some instances
constraints and because the most potent HA to orient influenza scientists to features on
globular head antibodies neutralize more the HA globular head. One could easily use
strongly than HA stem antibodies. Further- terms such as 190-helix instead of Sb
more, the cocrystal structures of HA globular antigenic site, but even epitopes such as the
head antibodies in complex with HA may Sb antigenic site that was supposed to be a
further serve as starting points for the design linear epitope (22) seem like a rather complex
of inhibitors against the RBD. Therefore, it epitope of multiple loops (23). Even antibodies
seems timely to review the literature on HA that target the conserved receptor-binding
globular head antibodies. sites often touch this 190-helix or Sb site
(Table 2).
For H3, antigenic sites designated A to D
THE FIRST ANTIGENIC MAPPING were established (24). Around the year
2000, three cocrystal structures of mouse
The first mouse monoclonal antibodies against H3 globular head antibodies were solved
influenza A HA were generated by hybridoma in complex with their HA (25, 26, 27, 28)
technology in the early 1980s. By testing these in work reviewed in detail by Knossow
mAbs in competition assays and/or through the and Skehel (29). The antigenic structure of
selection and sequencing of escape mutants in each HA subtype is slightly different, and
embryonated egg cells, an antigenic mapping of one should exercise caution when using
the HA globular head could be performed for the antigenic mapping from the HA of one
H1, H2, and H3 viruses. For the H1 antibodies, subtype to describe the topography of the
antigenic sites designated Sa, Sb, Ca1, Ca2, and HA from an entirely different subtype. A
Cb were proposedthe S standing for strain- case in point is the H2 HA. In the initial
specific and the C for cross-reactive (19). antigenic mapping, the authors were unable
Somewhat ironically, the preservation of the to establish discrete antigenic sites on H2
supposedly strain-specific Sa antigenic site HA, since most epitopes overlapped (30).
of the 1918 H1N1 HA in the pandemic 2009 Also, about half of the antibodies cloned
virus was a major reason for antibody cross- were influenced by receptor specificity (30).
212 KRAUSE AND CROWE

TABLE 2 Select cross-reactive globular head HA antibodies


Characteristics 2D1 1F1 5J8 CH65/CH67 2G1, 8M2 S139/1 C05

Origin Human Human Human Human Human Mouse Human phage


hybridoma hybridoma hybridoma plasmablast hybridoma hybridoma display
VH gene VH2-70 VH3-30 VH4-b VH1-2 VH1-69 Not VH3-23
assignment described
Bind(s) Sites Sa, Sb, site Ca2 RBD, sites RBD, site Sb RBD RBD, RBD, site Sb
Sb, Ca1 Sb, Ca2 sites A, B, D
Mimics sialic n/a + + + +
acid
Binds and/or Pandemic H1 1918 H1 Most H1N1, Most H1N1 Most H2N2 Select H1, Select H1, H2,
neutralizes and select including since 1977 H2, H3, H3, H9, H12
20th pandemic H1 and H13
century
strains
Breadth Not tested Not tested + + Not tested + +
extended by
avidity
Note Lead mAb Binds Three escape Rigidied Lead mAbs Fab by itself Cross-
for 1918/ residues mutations, heavy chain for VH1-69 protects reactivity
2009 mediating all outside interactions globular against H3 mediated by
cross- receptor- conventional increase head HA viruses single antibody
reactivity specicity antigenic afnity recognition loop
sites
PDB(s) of 3LZF, 3QHZ, 4GXU, 4M5Y, 4M5Z 3SM5, 4HG4, 4GMS, 4FQR, 4FP8,
crystal 3QHF 4GXV 4HKB, 4HFU 4GMT 4FNL
structure 4KH3, 4KH0
References 2, 20, 32, 33 20, 23, 33 13, 45 15 16, 17 47, 48 49

We now know that receptor specificity of H2 that have infected humans: H1, H2, H5) and
viruses is mediated by residues 226 and 228 group 2 HA viruses (which includes additional
(31), two residues that lie on the edge of the subtypes that have infected humans: H3, H7),
RBD. Why the murine immune response to respectively, some stem antibodies can neu-
H2 HA is so focused on the conserved RBD is tralize multiple strains across subtypes. How-
not clear (16). However, human monoclonal ever, because of hypervariability of loops
antibodies against H2 HA also seem to be surrounding the RBD, the cross-reactive po-
focused on this very same pocket (16, 17). tential of globular head antibodies is generally
Thus, such antibodies, if present on a popula- thought to be more limited. Cross-reactive
tion level, may have limited the duration of antibodies to the globular head HA can bind
H2N2 circulation in humans to 1957 to 1968, a outside of the conserved RBD. Some particu-
relatively brief period in comparison with that larly well-studied examples are the human
of other subtypes, for example, H1N1 that has pandemic H1N1 mAbs 2D1 (2, 20, 32, 33) (Table
been in human circulation from 1918 until 1957 2), 2B12 (20, 33), and related Sa site-specific
and again from 1977 through the present time. antibodies (14). Such antibodies neutralize the
pandemic 1918 and 2009 H1N1 viruses and
CROSS-REACTIVITY OF GLOBULAR HEAD swine H1N1 viruses, because residues in the Sa
HA ANTIBODIES site epitope of the 1918 H1N1 are almost
unchanged in the 2009 H1N1 virus (2, 20).
Since the HA stem is conserved within group 1 Richard Shope discovered in the 1930s that
HA viruses (which includes several subtypes variants of the pandemic 1918 H1N1 virus
CHAPTER 12 Committing the Oldest Sins in the Newest Kind of Ways 213

continued to circulate in pigs (34), probably ANTIBODIES THAT TARGET RESIDUES


because antigenic pressure in those short-lived THAT MEDIATE RECEPTOR SPECIFICITY
animals is very low. Parts of those viruses were
reintroduced in 2009 into human circulation We cloned both mAbs 1F1 and 1I20 from
through a triple reassortant virus event, so one survivor of the 1918 H1N1 pandemic
elderly people that had immunity to the 1918 (33). The heavy chains of those antibodies
H1N1 virus or swine viruses such as the New are genetically related, but clonally inde-
Jersey/1976 vaccine strain in the United States pendent. MAb 1I20 is strain specific, but
may have been partially protected against the mAb 1F1 is able to neutralize select H1N1
2009 H1N1 virus through Sa site-specific strains across the 20th century (33), includ-
antibodies. There was nothing inherently ing 1943, 1947, and 1977 viruses, but not the
cross-reactive about these antibodies; the pandemic 2009 H1N1 virus (20). These
supposedly hypervariable epitope of such antibodies selected for escape mutations in
antibodies was simply conserved between position P186 adjacent to the Sb antigenic
the two H1 viruses 1918 and 2009. MAb 2D1 site (33). The cocrystal structure of 1F1 in
did have an interesting feature, a 3-amino-acid complex with 1918 HA shows that 1F1 does
insertion, that reconfigures that antibody- indeed make multiple contacts within the
combining site by distorting and removing 190-helix of the Sb antigenic site, but also
the CDR-H1 loop away from the antigen- touches on the Ca 2 antigenic site, and
antibody interface (32). reaches into the RBD (23) (Table 2). This
Sa site-specific antibodies such as 2D1 or finding supports our hypothesis that most
2B12 do not recognize seasonal H1N1 strains human antibodies contact multiple conven-
that circulated between the H1N1 pandemics tional antigenic sites, although the number
(33). Strains such as A/USSR/1977 H1N1 of influenza antibody cocrystal structures
possess three predicted N-linked glycosylation is too limited so far to state that this is a
sites within its Sa antigenic site (20). It is universal characteristic of human neutral-
generally believed that glycosylation at these izing antibodies to HA head domain. In
sites shields HA from neutralization (20, 35). addition to those conventionally defined
However, we were able to clone a panel of antigenic sites, 1F1 contacts several HA
neutralizing antibodies that bound the Sa site residues that typically interact with the
based on escape mutations of the Sa site sialoglycan receptor, including 135, 153,
residue K166, but that were still able to 183, 190, 194, 222, and 225 (23). Residues
neutralize A/USSR/1977 H1N1 despite the D190 and D225 mediate H1 HA receptor
presence of said glycosylation sites (14). We specificity (31, 37, 38, 39, 40, 41, 42). Reverting
presently do not understand the structural these residues to D190E and D225G of avian
basis for the fact that glycosylation within the H1N1 viruses significantly reduces binding
Sa site does not always confer escape from Sa- for both 1F1 and 1I20 (23). While it is not
specific mAbs. Unexpectedly, this panel of surprising that two antibodies with similar
independent antibody clones that was derived genetic backgrounds recognize the same epi-
of the VH3-7/JH6 germ line heavy chain genes tope (as was seen also for the VH3-7/JH6 germ
converged toward similar mutations (14). line antibodies mentioned above [14]), we
Interestingly, even essentially unmutated were surprised to see that the epitope of
states of those antibodies were found in the 1F1 on H1 HA is similar to the murine mAb
peripheral blood (14). Persistent low-affinity HC63 epitope on H3 HA (28, 43), despite a
memory populations may aid in the immu- rotation of the heavy chains by 20, resulting
nologic response toward a related HA that in completely different interactions for the
the individual subsequently encounters (8, 14, light chains of 1F1 and HC63. There may be
36). only a limited number of preferred antibody-
214 KRAUSE AND CROWE

binding modes even across subtypes (23). (13). It seems likely that H1N1 cannot easily
HC63 can not only inhibit receptor binding, change such residues without the loss of
but also inhibits the pH-induced conforma- replicative capacity (13).
tional change that happens upon receptor The human H2N2 mAbs 8M2, 2G1, and 8F8
binding, because HC63 binds multiple HA are also directed against the RBD (16, 17). 8M2
monomers (28, 43); 1F1 Fab does not share and 2G1 are encoded by the VH1-69 germ line
these features (23). gene that was conventionally thought of as
being stem specific, but it is also ideally suited
for recognition of another structurally con-
ANTIBODIES THAT MIMIC SIALIC ACID, served influenza hydrophobic pocket, the HA
THE INFLUENZA RECEPTOR RBD (17). Even though 8M2 and 2G1 were
cloned from two different individuals, they
Whittle et al. crystallized the human mAb both insert germ line Phe54 of CDR-H2 into
CH65 that was cloned by plasmablast tech- the RBD, and thus provide further evidence of
nology from the peripheral blood of a seasonal sequence convergence across antibodies (14).
influenza vaccine recipient in complex with In the H2-antibody complexes, the aromatic
A/Solomon Islands/3/2006 HA (15). CH65 rings of Phe54 are situated for optimal p-p
neutralized a broad range of seasonal H1N1 interaction with the universally conserved HA
viruses from 1977 until 2006 (15) (Table 2). Trp153 and are surrounded by further highly
The CH65 CDR-H3 inserts into the RBD on conserved influenza A residues (17). MAb 8F8,
HA1 where it mimics the physiologic receptor, a VH3-33-encoded antibody, interacts with HA
sialic acid (15). CH65-resistant H1N1 strains Trp153 through the insertion of aromatic
seemed to have an insertion in position 133A of residue Tyr100 of CDR-H3. Escape is mediated
the HA, either an arginine or a lysine (15), but through residues that are on the edge of the
Schmidt et al. later found exceptions to this RBD (16).
assumption (44). Similarly, we reported the MAb 2G1 showed hemagglutination-inhibi-
H1N1 cross-reactive mAb 5J8 that neutralizes tion (HAI) activity against H2N2 viruses, but
a broad range of H1N1 strains of the 20th also against the pandemic H3N2 virus from
century and the pandemic strains of H1N1, a 1968 (16). The human VH1-69 encoded mAb
spectrum of activity that seemed complemen- F045-092 that was derived of phage display
tary to the mAb CH65 (13). Indeed, mAb 5J8- technology has been reported to cross-neutral-
resistant H1N1 strains did not have an inser- ize select strains from an even broader spec-
tion in position 133A of the HA (13). Also, a trum of influenza subtypes (H1, H2, H3, and
cocrystal structure of 5J8 in complex with the H13) (46). S139/1 was the first head-specific
globular HA1 head domain of A/California/ mAb to be described to neutralize select strains
07/2009 HA revealed that 5J8 also uses from different influenza subtypes (H1, H2, H3,
receptor mimicry, but both antibodies have and H13; Table 2) (47). The crystal structure of
distinct footprints and different angles of the S139/1 Fab in complex with the HA from
approach (45). Antibodies such as 5J8 may the A/Victoria/3/1975 (H3N2) virus reveals
have also contributed to the preexisting that the mAb targets residues within the RBD
immunity toward the pandemic 2009 virus and contacts antigenic sites A, B, and D (48).
(13), although we speculate that such anti- Although S139/1 Fab is sufficient for neutrali-
bodies were less frequent than Sa site-specific zation of most H3N2 viruses, it depends on the
antibodies such as 2D1 (14, 20, 32, 33). MAb avidity mediated by the bivalent IgG for its
5J8 elicited escape mutations in position 133A, heterosubtypic neutralization (48). The ex-
137, and 222, none of which is part of a tension of breadth through avidity is also a
conventionally defined H1N1 antigenic site feature for other mAbs that target the RBD
and all of which are relatively well-conserved (45, 49).
CHAPTER 12 Committing the Oldest Sins in the Newest Kind of Ways 215

The footprint of an antibody on HA is piratory-droplet-transmissible avian H5N1


larger than the RBD, so an antibody will viruses could be used as postexposure pro-
have to bind to at least some hypervariable phylaxis (50). Computational designs of pro-
residues and can compensate for this fact by teins against the stem region of HA have
avoiding high-affinity interactions depen- yielded novel proteins inhibitors (51) that can
dent on these positions (48). An antibody be further optimized by creating comprehen-
features three hypervariable loops each on sive sequence-function maps obtained by deep
the heavy and light chain, but not all of sequencing (52). The wealth of cross-reactive
those loops typically participate in the antibodies against the RBD could serve as
antigen-antibody interaction. An antibody templates to design novel influenza therapeu-
with a very long CDR-H3 can minimize its tics directed against the RBD of HA by using
footprint on the antigen (49). Ekiert et al. similar approaches. Because only few residues
reported such an antibody, designated mAb are universally conserved across all HAs, and
C05; it binds and/or neutralizes selected such residues will likely not be the only
strains from H1, H2, H3, H9, and H12 sub- contact residues of novel therapeutics, escape
types (Table 2). Apart from CDR-H3, only mutations will invariably occur. Based on
CDR-H1 makes additional minor interactions existing structures and naturally occurring
with HA, and only 550 2 of surface area is variability in those residues, it seems possible
buried on the HA by C05 (49). As with other to rationally predict such escape mutations
RBD antibodies, insertions at position 133A (49). One single protein or small molecule
seem to abrogate binding (49). And unlike might not be sufficient to neutralize all
other RBD antibodies, C05 interaction with influenza strains. It might be possible to
the RBD differs considerably from sialic acid develop multiple different inhibitors that
binding to HA (49). Insertions and deletions could be tailored toward specific strains or
at other positions explain why other HA sub- used in conjunction with other inhibitors
types are not bound and/or neutralized (49). (including neuraminidase inhibitors) as an
inhibitor cocktail.
The challenge remains how to elicit
CONCLUSIONS AND AREAS FOR reliably broadly cross-neutralizing human
FUTURE STUDIES mAb through vaccination. Studies suggest
that individuals may have a greater chance of
The antibodies described here have impor- mounting a robust cross-neutralizing antibody
tant potential diagnostic and therapeutic response if primed with DNA vaccines (53) or
applications. Antibodies such as 1F1 or 8M2 immunized with adjuvants (54). The con-
that recognize viruses with human-receptor served stem epitopes seem to be predominant-
specificity only, but not avian-receptor speci- ly encoded by the VH1-69 germ line gene.
ficity, could be used to screen for viruses with Given that there is variability in VH1-69 alleles
supposedly greater human infectivity. A col- and the number of VH1-69 alleles a person
lection of broadly neutralizing monoclonal carries, some individuals may mount a more
antibodies can be used for comparative stud- robust immune response toward the stem
ies, for example, to facilitate the development than others. Even though we (16, 17) and
of universal vaccines directed against the others (46) have shown that the VH1-69 gene
RBD. Passive administration of such anti- element plays an important role for antibodies
bodies to humans could provide protective against the RBD as well, the still limited
immunity, for example, for immunocompro- number of globular head antibodies suggests
mised individuals that would not be expected a more diverse genetic heritage of RBD mAbs.
to mount a protective immune response after Although antibodies such as C05 with its 24-
active immunization. Antibodies against res- amino-acid CDR-H3 and 5-amino-acid inser-
216 KRAUSE AND CROWE

tion in CDR-H1 stand out (49), unlike HIV preexisting immunity to the 2009 H1N1 pan-
antibodies, most cross-reactive human influ- demic influenza virus. Science 328:357360.
3. Corti D, Voss J, Gamblin SJ, Codoni G, Macagno
enza antibodies analyzed so far do not seem to A, Jarrossay D, Vachieri SG, Pinna D, Minola A,
need unusual structural features or an extra- Vanzetta F, Silacci C, Fernandez-Rodriguez
ordinarily high number of somatic mutations BM, Agatic G, Bianchi S, Giacchetto-Sasselli I,
to achieve cross-reactivity. This observation Calder L, Sallusto F, Collins P, Haire LF,
suggests that it may be easier to induce Temperton N, Langedijk JP, Skehel JJ,
Lanzavecchia A. 2011. A neutralizing antibody
influenza cross-reactive antibodies than selected from plasma cells that binds to group 1
cross-reactive HIV antibodies. It has been and group 2 influenza A hemagglutinins. Science
proposed to design immunogens to guide 333:850856.
precursors of mature B cells in multiple steps 4. Ekiert DC, Friesen RH, Bhabha G, Kwaks T,
toward a desired response (55). We believe Jongeneelen M, Yu W, Ophorst C, Cox F, Korse
HJ, Brandenburg B, Vogels R, Brakenhoff JP,
that H2 influenza may be a key for the design
Kompier R, Koldijk MH, Cornelissen LA, Poon
of a universal vaccine against influenza since LL, Peiris M, Koudstaal W, Wilson IA, Goudsmit
its RBD seems to be particularly immunogenic. J. 2011. A highly conserved neutralizing epitope
The molecular or structural basis for this on group 2 influenza A viruses. Science 333:843
phenomenon is not immediately apparent 850.
5. Okuno Y, Isegawa Y, Sasao F, Ueda S. 1993. A
from structures of H2 HA by itself or in
common neutralizing epitope conserved between
complex with antibodies (17), although the the hemagglutinins of influenza A virus H1 and H2
mechanism may be related to the paucity of strains. J Virol 67:25522558.
glycosylation sites on the globular head of H2 6. Wrammert J, Koutsonanos D, Li GM,
HA. Glycosylation can modulate the ability of Edupuganti S, Sui J, Morrissey M, McCausland
HA to induce cross-reactive antibodies (56). M, Skountzou I, Hornig M, Lipkin WI, Mehta A,
Razavi B, Del Rio C, Zheng NY, Lee JH, Huang
Increasing the immunogenicity of the RBD in M, Ali Z, Kaur K, Andrews S, Amara RR, Wang
influenza vaccines by modifying select resi- Y, Das SR, ODonnell CD, Yewdell JW,
dues of the HA globular head would be major Subbarao K, Marasco WA, Mulligan MJ,
step toward developing an additional compo- Compans R, Ahmed R, Wilson PC. 2011. Broadly
nent of a universal vaccine. cross-reactive antibodies dominate the human B
cell response against 2009 pandemic H1N1 influ-
enza virus infection. J Exp Med 208:181193.
ACKNOWLEDGMENT 7. Davenport FM, Hennessy AV, Francis T, Jr.
1953. Epidemiologic and immunologic signifi-
Conflicts of interest: We declare no conflicts. cance of age distribution of antibody to antigenic
variants of influenza virus. J Exp Med 98:641656.
8. Fish S, Zenowich E, Fleming M, Manser T. 1989.
CITATION Molecular analysis of original antigenic sin. I.
Clonal selection, somatic mutation, and isotype
Krause JC, Crowe JE, Jr. 2014. Committing switching during a memory B cell response. J Exp
the oldest sins in the newest kind of ways Med 170:11911209.
antibodies targeting the influenza virus type A 9. Krause R. 2006. The swine flu episode and the
hemagglutinin globular head. Microbiol Spec- fog of epidemics. Emerg Infect Dis 12:4043.
10. Wrammert J, Smith K, Miller J, Langley WA,
trum 2(5):AID-0021-2014. Kokko K, Larsen C, Zheng NY, Mays I, Garman
L, Helms C, James J, Air GM, Capra JD, Ahmed
R, Wilson PC. 2008. Rapid cloning of high-
REFERENCES affinity human monoclonal antibodies against
1. Wilson IA, Skehel JJ, Wiley DC. 1981. Struc- influenza virus. Nature 453:667671.
ture of the haemagglutinin membrane glyco- 11. Palese P, Wang TT. 2011. Why do influenza virus
protein of influenza virus at 3 A resolution. subtypes die out? A hypothesis. MBio doi:10.1128/
Nature 289:366373. mBio.00150-11.
2. Xu R, Ekiert DC, Krause JC, Hai R, Crowe 12. Wrammert J, Onlamoon N, Akondy RS, Perng
JEJ, Wilson IA. 2010. Structural basis of GC, Polsrila K, Chandele A, Kwissa M,
CHAPTER 12 Committing the Oldest Sins in the Newest Kind of Ways 217

Pulendran B, Wilson PC, Wittawatmongkol O, enza A virus by 1918-like and classical swine
Yoksan S, Angkasekwinai N, Pattanapanyasat H1N1 based vaccines. PLoS Pathog 6:e1000745.
K, Chokephaibulkit K, Ahmed R. 2012. Rapid and doi: 10.1371/journal.ppat.1000745.
massive virus-specific plasmablast responses dur- 22. Brownlee GG, Fodor E. 2001. The predicted
ing acute dengue virus infection in humans. antigenicity of the haemagglutinin of the 1918
J Virol 86:29112918. Spanish influenza pandemic suggests an avian
13. Krause JC, Tsibane T, Tumpey TM, Huffman origin. Philos Trans R Soc Lond B Biol Sci
CJ, Basler CF, Crowe JE. 2011. A broadly 356:18711876.
neutralizing human monoclonal antibody that rec- 23. Tsibane T, Ekiert DC, Krause JC, Martinez
ognizes a conserved, novel epitope on the glob- O, Crowe JE, Wilson IA, Basler CF. 2012.
ular head of the influenza H1N1 virus hemagglutinin. Influenza human monoclonal antibody 1F1
J Virol 85:1090510908. interacts with three major antigenic sites and
14. Krause JC, Tsibane T, Tumpey TM, Huffman residues mediating human receptor specificity
CJ, Briney BS, Smith SA, Basler CF, Crowe JE, in H1N1 viruses. PLoS Pathog 8:e1003067.
Jr. 2011. Epitope-specific human influenza anti- doi:10.1371/journal.ppat.1003067.
body repertoires diversify by B cell intraclonal 24. Wiley DC, Wilson IA, Skehel JJ. 1981. Struc-
sequence divergence and interclonal conver- tural identification of the antibody-binding
gence. J Immunol 187:37043711. sites of Hong Kong influenza haemagglutinin
15. Whittle JR, Zhang R, Khurana S, King LR, and their involvement in antigenic variation.
Manischewitz J, Golding H, Dormitzer PR, Nature 289:373378.
Haynes BF, Walter EB, Moody MA, Kepler 25. Fleury D, Wharton SA, Skehel JJ, Knossow
TB, Liao HX, Harrison SC. 2011. Broadly M, Bizebard T. 1998. Antigen distortion allows
neutralizing human antibody that recognizes influenza virus to escape neutralization. Nat
the receptor-binding pocket of influenza virus Struct Biol 5:119123.
hemagglutinin. Proc Natl Acad Sci USA 108: 26. Fleury D, Barrere B, Bizebard T, Daniels RS,
1421614221. Skehel JJ, Knossow M. 1999. A complex of
16. Krause JC, Tsibane T, Tumpey TM, Huffman influenza hemagglutinin with a neutralizing
CJ, Albrecht R, Blum DL, Ramos I, Fernandez- antibody that binds outside the virus receptor
Sesma A, Edwards KM, Garca-Sastre A, Basler binding site. Nat Struct Biol 6:530534.
CF, Crowe JE. 2012. Human monoclonal anti- 27. Fleury D, Daniels RS, Skehel JJ, Knossow M,
bodies to pandemic 1957 H2N2 and pandemic Bizebard T. 2000. Structural evidence for
1968 H3N2 influenza viruses. J Virol 86:6334 recognition of a single epitope by two distinct
6340. antibodies. Proteins 40:572578.
17. Xu R, Krause JC, McBride R, Paulson JC, 28. Barbey-Martin C, Gigant B, Bizebard T,
Crowe JE, Wilson IA. 2013. A recurring motif Calder LJ, Wharton SA, Skehel JJ, Knossow
for antibody recognition of the receptor-binding M. 2002. An antibody that prevents the
site of influenza hemagglutinin. Nat Struct Mol hemagglutinin low pH fusogenic transition.
Biol 20:363370. Virology 294:7074.
18. National Institute of Allergy and Infectious 29. Knossow M, Skehel JJ. 2006. Variation and
Diseases, National Institutes of Health, US infectivity neutralization in influenza. Immunology
Food and Drug Administration. 2012. Universal 119:17.
Influenza Vaccines Meeting Summary. 19 to 20 30. Yamada A, Brown LE, Webster RG. 1984.
June 2012. Characterization of H2 influenza virus hemag-
19. Caton AJ, Brownlee GG, Yewdell JW, glutinin with monoclonal antibodies: influence of
Gerhard W. 1982. The antigenic structure of receptor specificity. Virology 138:276286.
the influenza virus A/PR/8/34 hemagglutinin 31. Connor RJ, Kawaoka Y, Webster RG,
(H1 subtype). Cell 31:417427. Paulson JC. 1994. Receptor specificity in
20. Krause JC, Tumpey TM, Huffman CJ, human, avian, and equine H2 and H3 influenza
McGraw PA, Pearce MB, Tsibane T, Hai R, virus isolates. Virology 205:1723.
Basler CF, Crowe JE. 2010. Naturally occurring 32. Krause JC, Ekiert DC, Tumpey TM, Smith PB,
human monoclonal antibodies neutralize both Wilson IA, Crowe JE Jr. 2011. An insertion
1918 and 2009 pandemic influenza A (H1N1) mutation that distorts antibody binding site archi-
viruses. J Virol 84:31273130. tecture enhances function of a human antibody.
21. Manicassamy B, Medina RA, Hai R, Tsibane MBio 2:e00345-10. doi:10.1128/mBio.00345-10.
T, Stertz S, Nistal-Villan E, Palese P, Basler 33. Yu X, Tsibane T, McGraw PA, House FS,
CF, Garcia-Sastre A. 2010. Protection of mice Keefer CJ, Hicar MD, Tumpey TM, Pappas C,
against lethal challenge with 2009 H1N1 influ- Perrone LA, Martinez O, Stevens J, Wilson
218 KRAUSE AND CROWE

IA, Aguilar PV, Altschuler EL, Basler CF, H, Suphaphiphat P, Carfi A, Settembre EC,
Crowe JE, Jr. 2008. Neutralizing antibodies Dormitzer PR, Kepler TB, Zhang R, Moody
derived from the B cells of 1918 influenza MA, Haynes BF, Liao H-X, Shaw DE, Harrison
pandemic survivors. Nature 455:532536. SC. 2013. Preconfiguration of the antigen-binding
34. Shope RE. 1931. Swine influenza. I. Experi- site during affinity maturation of a broadly
mental transmission and pathology. J Exp Med neutralizing influenza virus antibody. Proc Natl
54:349359. Acad Sci USA 110:264269.
35. Wei CJ, Boyington JC, Dai K, Houser KV, 45. Hong M, Lee PS, Hoffman RMB, Zhu X, Krause
Pearce MB, Kong WP, Yang ZY, Tumpey JC, Laursen NS, Yoon S-I, Song L, Tussey L,
TM, Nabel GJ. 2010. Cross-neutralization of Crowe JE, Ward AB, Wilson IA. 2013. Antibody
1918 and 2009 influenza viruses: role of glycans recognition of the pandemic H1N1 Influenza virus
in viral evolution and vaccine design. Sci Transl hemagglutinin receptor binding site. J Virol 87:
Med 2:24ra21. doi:10.1126/scitranslmed.3000799. 1247112480.
36. Herzenberg LA, Black SJ, Tokuhisa T. 1980. 46. Ohshima N, Iba Y, Kubota-Koketsu R, Asano
Memory B cells at successive stages of differ- Y, Okuno Y, Kurosawa Y. 2011. Naturally
entiation. Affinity maturation and the role of occurring antibodies in humans can neutralize
IgD receptors. J Exp Med 151:10711087. a variety of influenza virus strains, including
37. Gamblin SJ, Haire LF, Russell RJ, Stevens H3, H1, H2, and H5. J Virol 85:1104811057.
DJ, Xiao B, Ha Y, Vasisht N, Steinhauer DA, 47. Yoshida R, Igarashi M, Ozaki H, Kishida N,
Daniels RS, Elliot A, Wiley DC, Skehel JJ. 2004. Tomabechi D, Kida H, Ito K, Takada A. 2009.
The structure and receptor binding properties Cross-protective potential of a novel monoclonal
of the 1918 influenza hemagglutinin. Science antibody directed against antigenic site B of the
303:18381842. hemagglutinin of influenza A viruses. PLoS Pathog
38. Stevens J, Corper AL, Basler CF, Taubenberger 5:e1000350. doi:10.1371/journal.ppat.1000350.
JK, Palese P, Wilson IA. 2004. Structure of the 48. Lee PS, Yoshida R, Ekiert DC, Sakai N,
uncleaved human H1 hemagglutinin from the Suzuki Y, Takada A, Wilson IA. 2012.
extinct 1918 influenza virus. Science 303:18661870. Heterosubtypic antibody recognition of the
39. Glaser L, Stevens J, Zamarin D, Wilson IA, influenza virus hemagglutinin receptor binding
Garcia-Sastre A, Tumpey TM, Basler CF, site enhanced by avidity. Proc Natl Acad Sci
Taubenberger JK, Palese P. 2005. A single USA 109:1704017045.
amino acid substitution in 1918 influenza virus 49. Ekiert DC, Kashyap AK, Steel J, Rubrum A,
hemagglutinin changes receptor binding spec- Bhabha G, Khayat R, Lee JH, Dillon MA,
ificity. J Virol 79:1153311536. ONeil RE, Faynboym AM, Horowitz M,
40. Stevens J, Blixt O, Glaser L, Taubenberger Horowitz L, Ward AB, Palese P, Webby R,
JK, Palese P, Paulson JC, Wilson IA. 2006. Lerner RA, Bhatt RR, Wilson IA. 2012. Cross-
Glycan microarray analysis of the hemagglutinins neutralization of influenza A viruses mediated
from modern and pandemic influenza viruses by a single antibody loop. Nature 489:526532.
reveals different receptor specificities. J Mol Biol 50. Thornburg NJ, Nannemann DP, Blum DL, Belser
355:11431155. JA, Tumpey TM, Desphande S, Fritz GA,
41. Colman PM, Tulip WR, Varghese JN, Tulloch Sapparapu G, Krause JC, Lee JH, Warm AB,
PA, Baker AT, Laver WG, Air GM, Webster RG. Lee DE, Li S, Winarski KL, Spiller BW, Meiler J,
1989. Three-dimensional structures of influenza Crowe JE, Jr. 2013. Human antibodies that
virus neuraminidase-antibody complexes. Philos neutralize respiratory droplet transmissible H5N1
Trans R Soc Lond B Biol Sci 323:511518. influenza viruses. J Clin Invest 123:44054409.
42. Naeve CW, Hinshaw VS, Webster RG. 1984. 51. Fleishman SJ, Whitehead TA, Ekiert DC,
Mutations in the hemagglutinin receptor- Dreyfus C, Corn JE, Strauch EM, Wilson IA,
binding site can change the biological proper- Baker D. 2011. Computational design of proteins
ties of an influenza virus. J Virol 51:567569. targeting the conserved stem region of influenza
43. Daniels PS, Jeffries S, Yates P, Schild GC, hemagglutinin. Science 332:816821.
Rogers GN, Paulson JC, Wharton SA, 52. Whitehead TA, Chevalier A, Song Y, Dreyfus
Douglas AR, Skehel JJ, Wiley DC. 1987. The C, Fleishman SJ, De Mattos C, Myers CA,
receptor-binding and membrane-fusion prop- Kamisetty H, Blair P, Wilson IA, Baker D.
erties of influenza virus variants selected using 2012. Optimization of affinity, specificity and
anti-haemagglutinin monoclonal antibodies. function of designed influenza inhibitors using
EMBO J 6:14591465. deep sequencing. Nat Biotechnol 30:543548.
44. Schmidt AG, Xu H, Khan AR, ODonnell T, 53. Wei C-J, Yassine HM, McTamney PM, Gall
Khurana S, King LR, Manischewitz J, Golding JG, Whittle JR, Boyington JC, Nabel GJ.
CHAPTER 12 Committing the Oldest Sins in the Newest Kind of Ways 219

2012. Elicitation of broadly neutralizing influenza 55. Haynes BF, Kelsoe G, Harrison SC, Kepler
antibodies in animals with previous influenza TB. 2012. B-cell-lineage immunogen design
exposure. Sci Transl Med 4:147ra114. doi:10.1126/ in vaccine development with HIV-1 as a case
scitranslmed.3004273. study. Nat Biotechnol 30:423433.
54. Khurana S, Chearwae W, Castellino F, 56. Medina RA, Stertz S, Manicassamy B,
Manischewitz J, King LR, Honorkiewicz A, Zimmermann P, Sun X, Albrecht RA, Uusi-
Rock MT, Edwards KM, Del Giudice G, Kerttula H, Zagordi O, Belshe RB, Frey SE,
Rappuoli R, Golding H. 2010. Vaccines with Tumpey TM, Garcia-Sastre A. 2013. Glyco-
MF59 adjuvant expand the antibody repertoire sylations in the globular head of the hemagglutinin
to target protective sites of pandemic avian protein modulate the virulence and antigenic
H5N1 influenza virus. Sci Transl Med 2:15ra5. properties of the H1N1 influenza viruses. Sci Transl
doi:10.1126/scitranslmed.3000624. Med 5:187ra70. doi:10.1126/scitranslmed.3005996.
Prevention of Respiratory
Syncytial Virus Infection:
From Vaccine to Antibody

KELLY HUANG1 and HERREN WU2


13
RESPIRATORY SYNCYTIAL VIRUS

Respiratory syncytial virus (RSV) poses a serious and significant health


problem. RSV was discovered in 1956 and quickly became recognized as the
leading cause of lower respiratory tract disease in infants and young
children (1, 2). Preterm infants and young children with bronchopulmonary
dysplasia (BPD) or congenital heart disease (CHD) are at high risk of serious
RSV infection and may require hospitalizations and stays in the pediatric
intensive care unit (3, 4). Although these high-risk groups experience an
increased incidence of RSV disease, it is important to note that the majority
of infants hospitalized for RSV are previously healthy, nonpremature
children (5). In children less than 5 years of age, RSV infections account
for 50 to 80% of winter bronchiolitis hospitalizations and 30 to 60% of
pneumonia hospitalizations (6, 7). RSV bronchiolitis is reported as the
leading cause of hospitalization for infants less than 12 months of age (8, 9,
10, 11). Hospitalization for RSV can reach rates of 1 to 20 per 1,000 infants
less than 1 year of age in developed countries (12). Although not common,
RSV can be fatal, as 140 to 500 infant deaths are attributed to RSV each year
in the United States (13, 14). In addition to infants and young children,

1
Department of Infectious Disease, MedImmune, LLC., Gaithersburg, MD 20878; 2Department of Antibody
Discovery and Protein Engineering, MedImmune, LLC., Gaithersburg, MD 20878.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0014-2014

221
222 HUANG AND WU

another risk group for RSV disease is the 34, 35). These spheroid and filamentous
elderly. In fact, while mortality in children structures are decorated with three enve-
due to RSV disease has decreased over the lope glycoproteins including G (glycosyla-
years, mortality due to RSV disease among ted) protein, F (fusion) protein, and SH
the elderly is still a significant problem (15, (small hydrophobic) protein. There is one
16, 17, 18, 19, 20). Also, immunosuppressed serotype of RSV and two antigenic subtypes
leukemia patients or patients receiving stem designated as A and B. The sequence diversity
cell transplant therapy experience as much of the G protein defines the A and B sub-
as 80 to 100% mortality upon RSV infection types, with 53% sequence identity between
and are therefore a high-risk group for RSV A and B subtypes; whereas, F-protein se-
disease (21). quence identity between subtypes is as high
RSV is highly infectious and thus highly as 89% (36). The G protein and F protein
prevalent; however, RSV disease does not are the major targets for antibody-mediated
occur in most cases of infection. Two-thirds virus neutralization.
of infants are seropositive for RSV by their The first steps of virus replication in-
first birthday, and nearly all children have clude attachment to the target cell followed
been infected by their second birthday (22). by viral and cellular membrane fusion. The
Outbreaks of RSV are seasonal, occurring G protein mediates the majority of the
during the colder months of the year and attachment process to target cells, and the
varying from year to year in peak months. F protein mediates the process of fusion.
In certain settings during seasonal RSV out- Recombinant viruses with ablated G-protein
breaks, such as daycare centers, the attack rate and SH-protein expression are still infec-
of RSV approaches 100% (23). In addition, tious in vitro (in some cell lines), suggesting
RSV is considered a major nosocomial prob- that the F protein alone can mediate both
lem for already hospitalized infants (24). attachment and fusion; however, viruses
Reinfection is quite common throughout life; with ablated G protein are highly attenu-
however, it is typically less severe than the ated in vivo (37). The contribution of the SH
primary infection (22, 23, 25, 26). It is reported protein to virus replication is unclear. Upon
that reinfection can occur even with a highly fusion, the ribonucleoprotein (RNP) com-
similar virus strain (26, 27), which raises plex, consisting of the viral genome coated
questions about the adaptive immunological by the viral N (nucleocapsid) protein along
response to RSV and about RSV seasonality in with the P (phosphoprotein) protein, L
that acquisition of herd immunity typically (RNA-dependent RNA polymerase) protein,
slows the rate of infection until a new strain and M2-1 protein (transcription factor), are
emerges and starts the next seasonal outbreak, released into the cytoplasm where viral
as is observed for influenza. However, the genome replication, transcription, and trans-
two subtypes of RSV tend to alternate in lation occur (36). Viral genomes and proteins
circulation by 1- to 2-year intervals, which associate at the cell surface to form new virus
suggests that there is at least some lasting herd particles that bud from the plasma membrane
immunity (28, 29, 30, 31). (36). In vitro, the F protein can mediate cell-to-
Respiratory syncytial virus is classified as cell fusion, called syncytium formation, which
part of the Pneumovirinae subfamily of the is the major cytopathic effect of RSV and can
Paramyxoviridae family of viruses. RSV is be easily observed by light microscopy. How-
an enveloped virus with a single-stranded, ever, the observation of syncytium formation
nonsegmented, and negative sense RNA in vivo is minimal and does not appear to play
genome. By electron microscopy, RSV is a a major role in viral spread. A general RSV
pleomorphic virus that adopts both spher- infection process of epithelial cells is shown
oid and long filamentous structures (32, 33, in Fig. 1.
CHAPTER 13 Prevention of RSV Infection: From Vaccine to Antibody 223

The pathogenesis of RSV begins with infection impairs cilia function leading to a
infection of the epithelial layer of the upper reduced ability to clear the necrotic debris
respiratory tract, resulting in rhinorrhea, (41). Infant bronchioles have a more restricted
possibly fever and otitis media 4 to 5 days airway resistance, resulting in greater dif-
later, which generally lasts 4 to 6 days (38). ficulty dealing with the debris and mucous
Viral shedding typically lasts 3 to 8 days, but brought on by an RSV infection of the lower
can be longer in immune-compromised respiratory tract. This might explain, at least in
individuals (38). In 25 to 40% of infections, part, why prematurity and young age are risk
the lower respiratory tract becomes infected, factors for RSV disease (42). The abnormali-
which can result in bronchiolitis or pneumo- ties in pulmonary function from RSV disease
nia (22). RSV infection causes necrosis of the can persist for 10 years and longer (43),
epithelial layer of the respiratory tract, infil- although it is unclear if the patients with
tration of leukocytes, edema, and excessive persistent pulmonary dysfunction had an
mucous, all of which lead to bronchial obs- underlying condition that predisposed them
truction (39, 40). To make matters worse, RSV to RSV disease.

FIGURE 1 The illustration depicts the mechanism of action of RSV-neutralizing mAbs, palivizumab and
motavizumab. An RSV virion is illustrated at the top left corner showing that the F and G proteins are
located on the surface of the virion and the drawing below depicts G-protein-mediated attachment to
cells followed by F protein mediated virus-cell fusion. The steps of fusion are shown on the top right.
F protein also mediates cell-to-cell fusion resulting in syncytium formation, depicted in the middle
drawing. In the nal drawing on the bottom, an RSV-neutralizing antibody, either palivizumab or
motavizumab, binds to F protein and blocks virus replication. It was determined that palivizumab and
motavizumab do not inhibit RSV attachment, but rather F-protein-mediated virus-cell fusion and
syncytia formation as reported recently (99). doi:10.1128/microbiolspec.AID-0014-2014.f1
224 HUANG AND WU

The standard of care or treatment of RSV Another example, as discussed in detail later in
disease is an evolving paradigm. One goal in this article, is that high-titer anti-RSV intra-
treatment is to reduce viral load. To this venous immunoglobulin (IVIG) and anti-RSV
end, ribavirin, a nonspecific antiviral was monoclonal antibody administration provides
used historically as a treatment for RSV. sterilizing immunity to RSV. Finally, there is a
However, ribavirin is no longer recommended direct correlation between the level of neu-
since it does not appear to provide a clinical tralizing antibody in the serum and reduced
benefit (44, and American Academy of Pedi- risk and severity of RSV disease (25, 26, 51, 52,
atrics). Another approach to treating RSV 53, 54, 55, 56, 57, 58).
infection is to treat the pathological results
of RSV, particularly inflammation. However,
treatment with corticosteroids alone has RSV VACCINE
proven to be ineffective (45). Because of
the clinical similarity between RSV disease Despite advances in understanding the
and asthma, bronchial dilators were explored immunological correlates to protection, a
as a treatment strategy for RSV infection, but vaccine for RSV is not available. RSV vaccine
were not efficacious (46). One success story in efforts were greatly stifled by the use of a
the treatment of RSV disease is the use of formalin-inactivated RSV vaccine that
cysteinyl leukotrienes that effectively treat resulted in a greater number of vaccinees
persistent wheezing that can follow bron- hospitalized upon infection with RSV in
chiolitis (47). comparison with the placebo control group
RSV is unique in its ability to cause (80% versus 5%, respectively) due to serious
reinfections in persons of all ages (22, 23, pulmonary dysfunction, and this resulted
25, 26, 27). This observation has led to the in some fatalities (59). Upon RSV infection,
hypothesis that RSV infection does not vaccinees experienced exaggerated lympho-
induce a durable immune response. There cyte proliferation that resulted in a high Th2
is evidence that supports the role of both response, which is speculated to have contrib-
cell-mediated and humoral immunity in uted to the high degree of lung inflammation
clearing an RSV infection. In terms of cell- (59). The formalin-inactivated RSV vaccine
mediated immunity, most RSV proteins will results were recapitulated in animal models
stimulate a cytotoxic T lymphocyte (CTL) with vaccines consisting of recombinant viral
response in seropositive individuals (48). proteins (60, 61, 62), which may guide current
The observation that RSV tetramer+CD8+ vaccine development. For instance, the use
cells are reduced in the elderly population of recombinant RSV viral protein vaccines
(49), a high-risk group for RSV disease, and might be safer for the elderly population to
that these cells do not have a robust prevent reinfection, but may not be preferred
proliferation response in RSV recall assays for the pediatric population to prevent prima-
in comparison with young patients, suggests ry RSV infection out of concern for provoking
the importance of cell-mediated immunity a high Th2 response. Strategies for pediatric
in clearing RSV (50). The role of humoral vaccine include attenuated RSV or virus vectors
immunity is supported by evidence from expressing RSV antigens. Attenuated virus
numerous studies that show antibodies re- vaccines will need to be sufficiently attenuated
strict RSV replication and aid faster clearance to greatly minimize the chance of reversion to
by cell-mediated immunity (reviewed in ref- wild-type virus. Virus vectored antigens typi-
erence 36). One supportive example of this is cally possess low immunogenicity and will
the observation that maternal antibodies ac- need to be engineered to improve the ability
quired via transplacental passive immuniza- to provoke cell-mediated immune responses.
tion provide a protective role for neonates (12). All pediatric vaccine strategies will need
CHAPTER 13 Prevention of RSV Infection: From Vaccine to Antibody 225

to address the challenges of immunological of anti-RSV antibodies in the presence of RSV


immaturity and maternal antibodies that ham- was not observed in any of these animal
per vaccine efforts (63, 64, 65, 66). Further studies, which is in contrast to the results of
understanding of the correlates of protec- the formalin-inactivated vaccine administered
tion against RSV infection and the correlates to humans.
of risk will greatly benefit vaccine develop-
ment efforts.
IMMUNOPROPHYLAXIS OF RSV
Since the identification of RSV, the need
INFECTION: FROM DISCOVERY
for a vaccine has long been recognized (1).
TO MARKET APPROVAL
However, the efforts to develop a safe and
effective vaccine against RSV have been
Intravenous Immunoglobulin
met with great challenges. This has steered
researchers to seek alternative approaches. Since IVIG was shown to protect animals
Early evidence suggested that passive im- against RSV infection (71, 72), it seemed
munization with antibodies may be a viable likely that passive immunization with
alternative for the prophylaxis of RSV infec- human IVIG would prevent RSV infection
tion. It was found that the severity of RSV- in high-risk children. Human clinical trials
induced pneumonia was inversely related to were conducted to test this hypothesis (73, 74).
the titer of maternal neutralizing antibody in It was concluded that monthly infusions of
infants (53). Moreover, the level of serum standard immune globulin containing RSV-
IgG to RSV F (fusion) protein has correlated neutralizing antibodies could be safely admin-
with the protective effect against RSV rein- istered to high-risk pediatric patients. There
fection and illness severity (51). However, was a trend toward less severe RSV illness
there was controversial view about antibodies in IVIG-treated patients compared with the
in the early days, since vaccine results seemed control group, as measured by the length of
to imply that serum antibodies confer little hospitalization. However, standard IVIG
protection and might even exacerbate dis- lacked sufficient RSV-neutralizing antibody
ease (67). In addition, the incidence of RSV- titer to confer full protection against severe
induced bronchiolitis peaks in infants between RSV illness (74). To improve on efficacy, an
1 and 6 months of age when maternal anti- effective screening assay to identify plasma-
bodies are still present (68). Further studies in yielding immunoglobulin with high RSV-
animal models using monoclonal or polyclonal neutralizing and animal-protective activities
antibodies have helped delineate the protec- was developed (75). Among seven assays
tion role of antibodies and enhanced confi- tested to identify such RSV antibody acti-
dence in the antibody-mediated prophylaxis vity, a microneutralization assay was found to
approach. Two research teams showed that be most useful. Microneutralization-screened
animals (mice and cotton rats) are protected IVIG, so called RSV-IVIG, has 5-fold better
against lung infection by the administration activity to neutralize RSV than standard IVIG.
of mouse anti-RSV monoclonal antibodies Two major clinical trials were conducted
prior to RSV challenge (69, 70). In addition, using RSV-IVIG (76, 77). The results from
it was found that human convalescent antise- these randomized, controlled trials were
rum to RSV administered intraperitoneally very promising; they demonstrated the
to cotton rats provided near-complete pul- safety and efficacy of RSV-IVIG in pre-
monary protection upon RSV challenge (71). venting RSV infection in pediatric patients.
Furthermore, human IVIG was shown to In the National Institute of Allergy and
reduce RSV replication in the lungs of the Infectious Diseases (NIAID) study, 249 child-
RSV-infected monkeys (72). Evidence of en- ren with prematurity, BPD, or CHD were
hanced pathology due to the administration enrolled (76). Monthly infusions at 750 mg/kg
226 HUANG AND WU

resulted in a 63% reduction in RSV hospi- derived from human donors screened for a
talizations and in a 97% reduction in the high titer of RSV-neutralizing activity.
number of days in the intensive care unit. Despite the fact that the production of
The adverse events were generally mild. The RSV-IVIG utilizes modern viral inactivation
PREVENT study sponsored by MedImmune, methods, concerns about transmission of
Inc. (Gaithersburg, MD) was a larger trial, unsuspected blood-borne pathogens have
which enrolled 510 children with BPD and/or remained. RSV-IVIG requires monthly in-
a history of prematurity (77). In this study, it travenous infusion and is time consuming,
was demonstrated that monthly administra- typically lasting several hours with the
tion of 750 mg of RSV-IVIG per kg reduced administration of a total fluid volume of
RSV hospitalization by 41%. The PREVENT 15 ml/kg. This could cause fluid overload in
study reported a 53% reduction in the total some children (76, 77). In addition, RSV-
number of RSV hospitalization days, a 60% IVIG, similar to other IVIGs, may poten-
reduction in the number of RSV days with tially interfere with routine administration
increased oxygen requirement, and a 54% schedules of certain vaccines, like measles,
reduction in the number of RSV hospital mumps, and rubella (80). Furthermore, in a
days with a moderate or severe lower respira- trial in children with CHD, RSV-IVIG did
tory track illness upon RSV-IVIG administra- not show a statistically significant decrease
tion. Similar to the NIAID study results, in RSV hospitalization for all children with
RSV-IVIG was shown to be safe and well CHD (although there was a trend). Also,
tolerated in the PREVENT study, as well, with there was a significantly higher frequency
only 1 to 3% of treated children experiencing of unanticipated cyanotic episodes and poor
medically significant adverse events related outcomes after surgery among children with
to RSV-IVIG administration. The safety profile cyanotic CHD in the RSV-IVIG group than
was similar to other IVIG treatments. Based in the control group (81). The hyperviscosity
on these trial results, the U.S. Food and Drug caused by 750 mg of RSV-IVIG per kg was
Administration (FDA) approved MedImmunes speculated to be one of the potential causes.
RSV-IVIG (RespiGam) on 18 January 1996 for To improve upon RSV-IVIG, researchers
the prevention of serious lower respiratory turned toward recombinant monoclonal
tract infection caused by RSV in children antibodies (mAbs) as a second-generation pro-
<24 months of age with BPD or a history of duct. There were great technology advance-
premature birth (35 weeks gestation). This ments in the antibody field from 1975 to
was a major milestone in the development 1990. Several key technologies were invented,
of effective medicine against severe RSV including hybridoma, chimeric antibody, an-
disease. It validated the hypothesis of passive tibody humanization, and recombinant anti-
immunization with antibodies as an effective body expression in mammalian cell cultures.
approach for the prophylaxis of RSV infec- These technologies allowed researchers to
tion in humans. In separate trials, RSV-IVIG produce highly specific mAbs against RSV
was tested to treat RSV-infected infants and with high affinity. The humanization ap-
young children. Unfortunately, despite giving proach was a key component in the develop-
the very high dose of 1,500 mg/kg, there was ment of the second-generation RSV product.
no significant therapeutic effect (78, 79). This approach was used to reshape hybrid-
oma-derived murine monoclonal antibodies to
be human-like, and enabled evasion of the
Monoclonal Antibody
human immune system to reduce unwanted
Although RSV-IVIG is a safe and effective immunogenicity. Advances in mammalian
immunoprophylaxis against RSV infection, expressing technologies enabled high-quantity
there are some drawbacks. RSV-IVIG is production of RSV mAbs in a defined medium
CHAPTER 13 Prevention of RSV Infection: From Vaccine to Antibody 227

without the concerns of potential blood-borne The second antibody HNK20 is a mouse
pathogens. IgA secreted by hybridomas derived from
During the same period of time, basic lung lymphocytes of RSV-immunized mice
knowledge about RSV also greatly accumul- (86) and was developed by OraVax. The
ated, allowing researchers to identify appro- rationale of using IgA is that it is the dominant
priate RSV antigens as antibody targets. In antibody isotype in the upper airway secre-
animal studies (69, 70), researchers found that tions. It is also less likely to induce inflamma-
mAbs against two RSV glycoproteins, F and G, tory responses at the mucosal surface since it
confer protection; however, antibodies against cannot fix complement factors efficiently.
other RSV proteins, such as N protein and P HNK20 was developed as a nose-drop treat-
protein, have no significant effect. The sub- ment with the intent of protecting the site of
stantially conserved sequence identity of F initial infection, thus preventing infection
protein between subtypes, in comparison with from spreading to the lungs. HNK20 was
G protein (36), suggests that F protein is likely shown to protect the upper and lower respi-
a more ideal target than G protein for ratory tract from RSV infections in mice (86)
developing broad neutralizing mAbs among and in rhesus monkeys (87). In rhesus
different RSV strains. Further study by the use monkeys, HNK20 administered at 0.5 mg/
of a large panel of neutralizing mAbs against kg intranasally once daily for 2 days before
the F protein of RSV A2 strain was conducted RSV challenge and for 4 days after challenge
to construct a detailed topological and opera- reduced viral load in the nose, throat, and
tional map of epitopes involved in neutraliza- lungs by 3 to 4 log10/ml. After treatment,
tion and fusion (82). In this study, researchers HNK20 remained at viral neutralization levels
immunized mice by sequential infection in nasal secretions for >24 h. Encouraged by
with RSV A2 and recombinant vaccinia virus these results, human clinical trials were
expressing RSV F. conducted (88). However, HNK20 did not
In the 1990s, there were three recombi- reduce the RSV hospitalization rate signifi-
nant anti-RSV F-protein mAbs, two IgG1, cantly in a phase III trial during which >600
and one IgA tested in humans. One of the high-risk infants received intranasal pro-
antibodies, a humanized IgG1/k RSHZ19 (SB phylaxis treatment. In a subgroup analysis, a
209763) was developed by GlaxoSmithKline. trend with reduced RSV hospitalization was
RSHZ19 had demonstrated RSV-neutraliza- observed for infants younger than 4 months at
tion ability in mice (83). In an early trial study entry. The overall results were not
in healthy men at single ascending doses of encouraging, and there was no additional
0.025 to 10 mg/kg, RSHZ19 was shown to be clinical development of HNK20 (88).
safe and well tolerated, and immunogenicity The third antibody, palivizumab (also
against RSHZ19 was not detected (84). How- named MEDI-493 and Synagis) was devel-
ever, RSHZ19 administered at repeat doses oped by MedImmune, Inc., as described in
(two intramuscular doses, 8 weeks apart) up to the following section.
10 mg/kg failed in a pediatric trial to protect
infants born prematurely or with BPD against
Palivizumab: the Only Approved mAb
RSV lower respiratory tract infection (85). The
for Preventing RSV Infection
trial showed that RSHZ19 had a mean half-life
of 32.5 days and did not induce detectable The antibody, mAb 1129, is one from a panel
antidrug immunogenicity. The authors sug- of antibodies derived from hybridomas used
gested that higher doses should be considered to characterize the neutralization epitopes
in additional trials. However, no further study of RSV F protein in a study described earlier
was reported, and RSHZ19 was never licensed (82), and it showed neutralization of a broad
in any worldwide market. spectrum of RSV isolates. This antibody
228 HUANG AND WU

was licensed to and further developed by equivalent volume of placebo. Palivizumab


MedImmune, Inc. for potential applications was shown to reduce RSV hospitalization
in humans. incidence by 55% (hospitalization occurrence
Murine mAb 1129 was humanized by a was 10.6% for placebo treated versus 4.8% for
complementarity-determining region (CDR)- palivizumab treated). In addition, palivizumab
grafting approach (Fig. 2) (89). The light-chain treatment resulted in fewer days spent in the
CDRs were transplanted onto the human K102 hospital, less time on oxygen support, reduced
VL/Jk4 framework regions. The heavy-chain moderate/severe lower respiratory tract ill-
CDRs were transplanted onto the human Cor/ ness, and a lower incidence of intensive care
CE-1 VH framework regions. Several murine unit admission due to RSV infection. It was
residues on framework 4 regions of VH and concluded that palivizumab is safe and effec-
VL were retained to potentially maintain the tive for the prevention of serious RSV illness in
structural integrity of the binding site. In premature children and those with BPD.
addition, because of an unintended frameshift Based on these clinical results, palivizumab
during the humanization process, the first four was approved by the FDA in 1998 for
residues of the light chain CDR1, SASS, were immunoprophylaxis of serious RSV respirato-
substituted by four random, nonhuman, ry disease in premature infants and children
nonmouse residues, KCQL. The humanized with BPD. Subsequently, an additional study
antibody (IgG1/k), palivizumab, recognizes a was conducted to demonstrate the efficacy of
conserved neutralizing epitope on the RSV F palivizumab in young children with CHD (n =
protein with a binding affinity 1 to 2 nM in 1,287) (91), and this resulted in FDA approval
Kd, which is similar to that of the chimeric to use palivizumab as immunoprophylaxis in
derivative of the parental antibody (Fig. 1). this patient population. Palivizumab was also
Palivizumab was tested against a panel of approved in Europe in 1999, and in Japan in
57 clinical isolates of RSV consisting of 34 A 2002. Currently, it is licensed in over 60
and 23 B subtypes, and was shown to have countries and has been administered to more
broad neutralization activity against all test than one million high-risk infants and children.
isolates. When compared with RSV-IVIG in
the microneutralization and fusion-inhibition
Motavizumab and Motavizumab-YTE
assays, palivizumab demonstrated a 20- to 30-
fold enhanced potency. In a cotton rat pro- The approval of palivizumab validates the
phylaxis study, palivizumab was able to reduce approach for targeting RSV F protein.
RSV titers in the lung by more than 99% at a However, despite the use of palivizumab,
dose of 2.5 mg/kg. Furthermore, the adminis- there still existed an unmet medical need
tration of palivizumab did not induce in- regarding RSV infection. For example, some
creased RSV infection or pathology (89). infants treated with palivizumab still be-
Supported by the in vitro and in vivo results, come infected by RSV and require hospital-
palivizumab was further evaluated as an ization. In addition, there are no adequate
immunoprophylaxis agent against RSV infec- preventative or treatment measures available
tion in high-risk human infants. for adult immunocompromised patients. A
Several clinical trials were conducted to second-generation antibody, motavizumab,
evaluate the safety and efficacy of pali- was generated (92, 93) and tested in human
vizumab. The most notable trial was the clinical trials by MedImmune. It is an affinity-
multicenter Phase III IMpact study (90). In optimized, humanized mAb derived from
this study, 1,502 children with prematurity palivizumab. A direct-evolution approach
(35 weeks) or BPD were randomly assigned was used to substantially improve the binding
to receive either five monthly intramuscular kinetics, both kon and koff, of the antibody to F
injections of palivizumab (15 mg/kg) or an protein which is mediated by only 13 amino
CHAPTER 13 Prevention of RSV Infection: From Vaccine to Antibody 229

acid substitutions. Motavizumab binds to RSV In 2010, The Antiviral Drugs Advisory Com-
F protein with 70-fold higher affinity than mittee to the FDA voted not to recommend
palivizumab, and exhibits a 20-fold improve- approval of motavizumab, citing a concern on
ment in the viral neutralization potency in vitro. the risk/benefit profile. The FDA requested
In a cotton rat prophylaxis model, motavi- additional safety and efficacy data before
zumab was found to be more potent in reduc- considering motavizumab for approval. Sub-
ing nasal and lung RSV titers than palivizumab. sequently, MedImmune withdrew the BLA
Multiple clinical trials of motavizumab and discontinued further development of
were conducted. In a pivotal phase III, motavizumab for the prophylaxis of serious
noninferiority trial, 6,635 high-risk infants RSV respiratory disease.
and children were enrolled and received Both palivizumab and motavizumab have
15 mg/kg motavizumab or palivizumab a serum half-life of up to 3 weeks and
monthly for 5 months (94). Recognizing that require monthly dosing during the RSV
it may be challenging to show superiority season. With the intent to reduce the dosing
against an effective agent, this trial was frequency, the antibody constant region
designed to evaluate whether motavizumab (Fc) of motavizumab was engineered to
was noninferior and potentially superior to enhance its binding affinity to neonatal Fc
palivizumab in reducing the RSV hospitaliza- receptor (FcRn). Studies have shown that
tion rate and other RSV-associated endpoints. FcRn plays a key role in maintaining the serum
The trial results showed that the motavizumab IgG concentration. IgG binds to FcRn in a pH-
group had a 26% lower RSV hospitalization dependent manner, as it binds tightly at acidic
rate than the palivizumab group, which pH and has almost no binding at neutral pH.
achieved the noninferiority primary endpoint. This differential binding mechanism allows
In addition, motavizumab was shown to be efficient recycling of IgG back to circulation
superior to palivizumab in one of the second- during the pinocytosis event. Three mutations
ary endpoints for reducing RSV-specific out- (M252Y/S254T/T256E) were introduced to
patient medically attended lower respiratory the Fc region of motavizumab, resulting in a
tract infection (MALRI) by 50% compared new molecule named motavizumab-YTE (also
with palivizumab (P = 0.005). Overall, adverse named MEDI-557). These mutations, termed
events were not significantly different be- YTE, increase the binding of antibody to
tween these two groups. However, the inci- human FcRn at pH 6 by 10-fold while main-
dence of cutaneous reactions were higher in taining its very low or no binding ability to
the motavizumab group (7.2% versus 5.1% FcRn at pH 7.4. In pharmacokinetic studies in
with palivizumab; P < 0.001). The overall cynomolgus monkeys, the serum half-life and
results suggest motavizumab may provide an lung bioavailability of motavizumab-YTE
improved alternative in preventing serious were increased by 4-fold compared with mota-
RSV infection in high-risk infants and chil- vizumab (96). A randomized dose-escalation
dren. In a separate trial that compared phase I human trial in healthy adults (n = 31)
motavizumab with palivizumab in children was conducted to evaluate the pharmacokinet-
with CHD (n = 1,236) for safety and tolerabil- ics, tolerability, and safety of motavizumab-
ity, both molecules were shown to have similar YTE (97). A single dose of motavizumab-YTE
safety profiles with the exception of cutaneous or motavizumab (0.3, 3, 15, or 30 mg/kg) was
reactions, which occurred more frequently administered intravenously, and the data were
in motavizumab recipients (95). In 2008, collected for 240 days. It was found that the
MedImmune submitted a Biologics License half-life of motavizumab-YTE was 2- to 4-fold
Application (BLA) for motavizumab to the longer than that of motavizumab. In addition,
FDA for the prevention of serious RSV respir- motavizumab-YTE remains fully functional
atory disease in high-risk pediatric patients. during the course of 240 days in circulation,
230 HUANG AND WU

(Continued)
CHAPTER 13 Prevention of RSV Infection: From Vaccine to Antibody 231

same protection. These potent anti-RSV anti-


bodies are currently licensed to MedImmune
for further development. MedImmune is
applying its half-life extension YTE technolo-
gy to these antibodies. The goal is to provide
patients with a very potent long-lasting anti-
body that can be administered less frequently,
perhaps once per quarter or once per RSV
season. In addition to mAbs, other emerging
drug modalities are being explored. Ablynx
is currently developing an anti-RSV F pro-
tein Nanobody (ALX-0141) to treat RSV infec-
tions. Nanobody is a technology based on
the camelid VHH domain, about one-tenth
the size of mAb. ALX-0141 is a trivalent
Nanobody and is developed for delivery
directly into the lungs by inhalation; it is
currently in phase I human trial. In addition,
Symphogen had once developed a recombi-
FIGURE 2 (A) Palivizumab was generated by CDR- nant oligoclonal antibody approach to prevent
grafting humanization of murine mAb 1129.
RSV infection, Sym003. Sym003 is a mixture
Murine CDR regions are depicted in ball structure.
The remaining regions are human origin. (B) Top of six unique antibodies against RSV and was
view of the six murine CDRs that were grafted to in preclinical development. However, there
human frameworks. doi:10.1128/microbiolspec. are no new reports in recent years, and the
AID-0014-2014.f2 program is no longer shown on the Symphogen
website. It is likely that this project was
as determined by RSV neutralization activity. suspended for further development. Alnylam
Motavizumab-YTE was well tolerated and had has developed an RNAi approach (ALN-
an extended half-life of up to 100 days. RSV01) for the treatment of RSV infection.
Its RNAi therapeutic was designed to target
the nucleocapsid N gene, which is required
FUTURE OPPORTUNITIES FOR for RSV replication. The molecule was tested
TREATMENT AND PREVENTION in a phase IIb trial against progressive
OF RSV INFECTION bronchiolitis syndrome (BOS) in lung trans-
plant patients. A dose of 0.6 mg/kg or placebo
There are continued efforts in searching for was administered by inhalation once daily for
new approaches to treat or prevent RSV 5 days. In all analyses, inhaled ALN-RSV01
infection. Recently, several very potent anti- was associated with a clinically meaningful
RSV antibodies were isolated from peri- reduction in the incidence of BOS, and the
pheral blood memory B cells by genetic drug was safe and well tolerated. However, the
programming (98). In microneutralization study narrowly missed its primary endpoint
assay, these human antibodies were 2 log of reduction in BOS in an intent-to-treat
more potent than palivizumab. One of the analysis of confirmed RSV-infected patients.
most potent antibodies, D25, was tested in a There are no new reports on the status of
cotton rat prophylaxis model and achieved this program.
full prevention of lung viral replication at Palivizumab continues to remain the only
a dose of 0.6 mg/kg compared with the approved prophylaxis drug against RSV. De-
required 2 mg/kg of palivizumab to achieve spite the challenges in developing vaccines
232 HUANG AND WU

or new drugs against RSV, these new delivery system: a retrospective cohort study.
approaches offer promises for RSV interven- Clin Ther 32:22202229.
7. Shay DK, Holman RC, Newman RD, Liu LL,
tion in the future. Stout JW, Anderson LJ. 1999. Bronchiolitis-
associated hospitalizations among US children,
19801996. JAMA 282:14401446.
ACKNOWLEDGMENTS 8. Fryzek JP, Martone WJ, Groothuis JR. 2011.
Trends in chronologic age and infant respira-
We thank Vaheh Oganesyan for providing
tory syncytial virus hospitalization: an 8-year
the antibody model structures used in Fig. 2. cohort study. Adv Ther 28:195201.
We declare a conflict of interest: Both 9. Leader S, Kohlhase K. 2002. Respiratory
authors are employees of MedImmune, syncytial virus-coded pediatric hospitalizations,
which developed and markets palivizumab. 1997 to 1999. Pediatr Infect Dis J 21:629632.
10. Leader S, Kohlhase K. 2003. Recent trends in
severe respiratory syncytial virus (RSV) among
CITATION US infants, 1997 to 2000. J Pediatr 143:S127S132.
11. Stockman LJ, Curns AT, Anderson LJ,
Wu H, Huang K. 2014. Prevention of respira- Fischer-Langley G. 2012. Respiratory syncy-
tory syncytial virus infection: from vaccine tial virus-associated hospitalizations among
to antibody. Microbiol Spectrum 2(4):AID- infants and young children in the United
0014-2014. States, 19972006. Pediatr Infect Dis J 31:59.
12. Glezen WP, Paredes A, Allison JE, Taber LH,
Frank AL. 1981. Risk of respiratory syncytial
REFERENCES virus infection for infants from low-income
families in relationship to age, sex, ethnic group,
1. Chanock R, Roizman B, Myers R. 1957. and maternal antibody level. J Pediatr 98:708715.
Recovery from infants with respiratory illness 13. Shay DK, Holman RC, Roosevelt GE,
of a virus related to chimpanzee coryza agent Clarke MJ, Anderson LJ. 2001. Bronchiolitis-
(CCA). I. Isolation, properties and character- associated mortality and estimates of respira-
ization. Am J Hyg 66:281290. tory syncytial virus-associated deaths among
2. Beem M, Wright FH, Hamre D, Egerer R, US children, 19791997. J Infect Dis 183:1622.
Oehme M. 1960. Association of the chimpan- 14. Thompson WW, Shay DK, Weintraub E,
zee coryza agent with acute respiratory disease Brammer L, Cox N, Anderson LJ, Fukuda
in children. N Engl J Med 263:523530. K. 2003. Mortality associated with influenza
3. Boyce TG, Mellen BG, Mitchel EF, Jr, Wright and respiratory syncytial virus in the United
PF, Griffin MR. 2000. Rates of hospitalization States. JAMA 289:179186.
for respiratory syncytial virus infection among 15. Falsey AR, Walsh EE. 2005. Respiratory
children in Medicaid. J Pediatr 137:865870. syncytial virus infection in elderly adults.
4. Hall CB, Weinberg GA, Iwane MK, Blumkin Drugs Aging 22:577587.
AK, Edwards KM, Staat MA, Auinger P, 16. Walsh EE, Falsey AR, Hennessey PA. 1999.
Griffin MR, Poehling KA, Erdman D, Grijalva Respiratory syncytial and other virus infections
CG, Zhu Y, Szilagyi P. 2009. The burden of in persons with chronic cardiopulmonary disease.
respiratory syncytial virus infection in young Am J Respir Crit Care Med 160:791795.
children. N Engl J Med 360:588598. 17. Hall CB, Long CE, Schnabel KC. 2001.
5. Wang EE, Law BJ, Boucher FD, Stephens D, Respiratory syncytial virus infections in previ-
Robinson JL, Dobson S, Langley JM, ously healthy working adults. Clin Infect Dis
McDonald J, MacDonald NE, Mitchell I. 33:792796.
1996. Pediatric Investigators Collaborative Net- 18. Sorvillo FJ, Huie SF, Strassburg MA, Butsumyo
work on Infections in Canada (PICNIC) study A, Shandera WX, Fannin SL. 1984. An outbreak
of admission and management variation in of respiratory syncytial virus pneumonia in a
patients hospitalized with respiratory syncytial nursing home for the elderly. J Infect 9:252256.
viral lower respiratory tract infection. J Pediatr 19. Vikerfors T, Grandien M, Olcen P. 1987.
129:390395. Respiratory syncytial virus infections in adults.
6. Flaherman V, Li S, Ragins A, Masaquel A, Am Rev Respir Dis 136:561564.
Kipnis P, Escobar GJ. 2010. Respiratory 20. Dowell SF, Anderson LJ, Gary HE, Jr,
syncytial virus testing during bronchiolitis Erdman DD, Plouffe JF, File TM, Jr, Marston
episodes of care in an integrated health care BJ, Breiman RF. 1996. Respiratory syncytial
CHAPTER 13 Prevention of RSV Infection: From Vaccine to Antibody 233

virus is an important cause of community- ment (G) glycoprotein and GM1 within the
acquired lower respiratory infection among hos- envelope of mature respiratory syncytial virus
pitalized adults. J Infect Dis 174:456462. filaments revealed using field emission scanning
21. Whimbey E, Champlin RE, Couch RB, Englund electron microscopy. Virology 306:254267.
JA, Goodrich JM, Raad I, Przepiorka D, Lewis 34. Okamoto Y, Kudo K, Ishikawa K, Ito E,
VA, Mirza N, Yousuf H, Tarrand JJ, Bodey GP. Togawa K, Saito I, Moro I, Patel JA, Ogra
1996. Community respiratory virus infections PL. 1993. Presence of respiratory syncytial
among hospitalized adult bone marrow transplant virus genomic sequences in middle ear fluid
recipients. Clin Infect Dis 22:778782. and its relationship to expression of cytokines
22. Glezen WP, Taber LH, Frank AL, Kasel JA. and cell adhesion molecules. J Infect Dis
1986. Risk of primary infection and reinfection 168:12771281.
with respiratory syncytial virus. Am J Dis Child 35. Roberts SR, Compans RW, Wertz GW. 1995.
140:543546. Respiratory syncytial virus matures at the
23. Henderson FW, Collier AM, Clyde WA Jr, apical surfaces of polarized epithelial cells.
Denny FW. 1979. Respiratory-syncytial-virus J Virol 69:26672673.
infections, reinfections and immunity. A pro- 36. Collins PL, Crowe JE. 2007. Respiratory
spective, longitudinal study in young children. syncytial virus and metapneumovirus, p 1601.
N Engl J Med 300:530534. In Knipe DM, Howley PM (ed), Fields Virol-
24. Groothuis J, Bauman J, Malinoski F, ogy, 5th ed. Lippincott Williams & Wilkins,
Eggleston M. 2008. Strategies for prevention of Philadelphia, PA.
RSV nosocomial infection. J Perinatol 28:319323. 37. Teng MN, Whitehead SS, Collins PL. 2001.
25. Beem M. 1967. Repeated infections with respira- Contribution of the respiratory syncytial virus G
tory syncytial virus. J Immunol 98:11151122. glycoprotein and its secreted and membrane-
26. Hall CB, Walsh EE, Long CE, Schnabel KC. bound forms to virus replication in vitro and in
1991. Immunity to and frequency of reinfection vivo. Virology 289:283296.
with respiratory syncytial virus. J Infect Dis 38. Black CP. 2003. Systematic review of the
163:693698. biology and medical management of respiratory
27. Hall CB, Douglas RG Jr, Geiman JM, Messner syncytial virus infection. Respir Care 48:209233.
MK. 1975. Nosocomial respiratory syncytial virus 39. Aherne W, Bird T, Court SD, Gardner PS,
infections. N Engl J Med 293:13431346. McQuillin J. 1970. Pathological changes in
28. Hall CB, Walsh EE, Schnabel KC, Long CE, virus infections of the lower respiratory tract
McConnochie KM, Hildreth SW, Anderson in children. J Clin Pathol 23:718.
LJ. 1990. Occurrence of groups A and B of 40. Neilson KA, Yunis EJ. 1990. Demonstration of
respiratory syncytial virus over 15 years: associ- respiratory syncytial virus in an autopsy series.
ated epidemiologic and clinical characteristics in Pediatr Pathol 10:491502.
hospitalized and ambulatory children. J Infect 41. Tristram DA, Hicks W, Jr, Hard R. 1998.
Dis 162:12831290. Respiratory syncytial virus and human bron-
29. Peret TC, Hall CB, Schnabel KC, Golub JA, chial epithelium. Arch Otolaryngol Head Neck
Anderson LJ. 1998. Circulation patterns of Surg 124:777783.
genetically distinct group A and B strains of 42. Hogg JC, Williams J, Richardson JB,
human respiratory syncytial virus in a com- Macklem PT, Thurlbeck WM. 1970. Age as a
munity. J Gen Virol 79(Pt 9):22212229. factor in the distribution of lower-airway conduc-
30. Waris M. 1991. Pattern of respiratory syncytial tance and in the pathologic anatomy of obstructive
virus epidemics in Finland: two-year cycles lung disease. N Engl J Med 282:12831287.
with alternating prevalence of groups A and B. 43. Johnston ID. 1999. Effect of pneumonia in
J Infect Dis 163:464469. childhood on adult lung function. J Pediatr
31. White LJ, Waris M, Cane PA, Nokes DJ, 135:3337.
Medley GF. 2005. The transmission dynamics 44. Leyssen P, De Clercq E, Neyts J. 2008.
of groups A and B human respiratory syncytial Molecular strategies to inhibit the replication
virus (hRSV) in England & Wales and Finland: of RNA viruses. Antiviral Res 78:925.
seasonality and cross-protection. Epidemiol Infect 45. Patel H, Platt R, Lozano JM, Wang EE. 2004.
133:279289. Glucocorticoids for acute viral bronchiolitis in
32. Bachi T, Howe C. 1973. Morphogenesis and infants and young children. Cochrane Database
ultrastructure of respiratory syncytial virus. Syst Rev 3:CD004878. doi:10.1002/14651858.
J Virol 12:11731180. CD004878.
33. Jeffree CE, Rixon HW, Brown G, Aitken J, 46. Kellner JD, Ohlsson A, Gadomski AM, Wang
Sugrue RJ. 2003. Distribution of the attach- EE. 2000. Bronchodilators for bronchiolitis.
234 HUANG AND WU

Cochrane Database Syst Rev 2:CD001266. 58. Piedra PA, Jewell AM, Cron SG, Atmar RL,
doi:10.1002/14651858.CD001266. Glezen WP. 2003. Correlates of immunity to
47. Bisgaard H, Study Group on Montelukast respiratory syncytial virus (RSV) associated-
and Respiratory Syncytial Virus. 2003. A hospitalization: establishment of minimum
randomized trial of montelukast in respiratory protective threshold levels of serum neutraliz-
syncytial virus postbronchiolitis. Am J Respir ing antibodies. Vaccine 21:34793482.
Crit Care Med 167:379383. 59. Kim HW, Canchola JG, Brandt CD, Pyles G,
48. Cherrie AH, Anderson K, Wertz GW, Chanock RM, Jensen K, Parrott RH. 1969.
Openshaw PJ. 1992. Human cytotoxic T cells Respiratory syncytial virus disease in infants
stimulated by antigen on dendritic cells recognize despite prior administration of antigenic inac-
the N, SH, F, M, 22K, and 1b proteins of tivated vaccine. Am J Epidemiol 89:422434.
respiratory syncytial virus. J Virol 66:21022110. 60. Connors M, Collins PL, Firestone CY,
49. de Bree GJ, Heidema J, van Leeuwen EM, Sotnikov AV, Waitze A, Davis AR, Hung PP,
van Bleek GM, Jonkers RE, Jansen HM, van Chanock RM, Murphy BR. 1992. Cotton rats
Lier RA, Out TA. 2005. Respiratory syncytial previously immunized with a chimeric RSV FG
virus-specific CD8+ memory T cell responses glycoprotein develop enhanced pulmonary
in elderly persons. J Infect Dis 191:17101718. pathology when infected with RSV, a phenom-
50. Cusi MG, Martorelli B, Di Genova G, Terrosi enon not encountered following immuniza-
C, Campoccia G, Correale P. 2010. Age related tion with vacciniaRSV recombinants or RSV.
changes in T cell mediated immune response Vaccine 10:475484.
and effector memory to Respiratory Syncytial 61. Hancock GE, Speelman DJ, Heers K, Bortell
Virus (RSV) in healthy subjects. Immun Ageing E, Smith J, Cosco C. 1996. Generation of
7:14. doi:10.1186/1742-4933-7-14. atypical pulmonary inflammatory responses
51. Kasel JA, Walsh EE, Frank AL, Baxter BD, in BALB/c mice after immunization with the
Taber LH, Glezen WP. 1987. Relation of native attachment (G) glycoprotein of respira-
serum antibody to glycoproteins of respiratory tory syncytial virus. J Virol 70:77837791.
syncytial virus with immunity to infection in 62. Murphy BR, Sotnikov AV, Lawrence LA,
children. Viral Immunol 1:199205. Banks SM, Prince GA. 1990. Enhanced pul-
52. Bruhn FW, Yeager AS. 1977. Respiratory monary histopathology is observed in cotton rats
syncytial virus in early infancy. Circulating immunized with formalin-inactivated respiratory
antibody and the severity of infection. Am J syncytial virus (RSV) or purified F glycoprotein
Dis Child 131:145148. and challenged with RSV 3-6 months after
53. Lamprecht CL, Krause HE, Mufson MA. immunization. Vaccine 8:497502.
1976. Role of maternal antibody in pneumonia 63. Karron RA, Wright PF, Belshe RB, Thumar
and bronchiolitis due to respiratory syncytial B, Casey R, Newman F, Polack FP, Randolph
virus. J Infect Dis 134:211217. VB, Deatly A, Hackell J, Gruber W, Murphy
54. Ogilvie MM, Vathenen AS, Radford M, Codd BR, Collins PL. 2005. Identification of a
J, Key S. 1981. Maternal antibody and respira- recombinant live attenuated respiratory syncytial
tory syncytial virus infection in infancy. J Med virus vaccine candidate that is highly attenuated
Virol 7:263271. in infants. J Infect Dis 191:10931104.
55. Parrott RH, Kim HW, Arrobio JO, Hodes DS, 64. Murphy BR, Alling DW, Snyder MH, Walsh
Murphy BR, Brandt CD, Camargo E, EE, Prince GA, Chanock RM, Hemming VG,
Chanock RM. 1973. Epidemiology of respi- Rodriguez WJ, Kim HW, Graham BS. 1986.
ratory syncytial virus infection in Washington, Effect of age and preexisting antibody on serum
D.C. II. Infection and disease with respect to antibody response of infants and children to the F
age, immunologic status, race and sex. Am J and G glycoproteins during respiratory syncytial
Epidemiol 98:289300. virus infection. J Clin Microbiol 24:894898.
56. Stensballe LG, Ravn H, Kristensen K, Agerskov 65. Shinoff JJ, OBrien KL, Thumar B, Shaw JB,
K, Meakins T, Aaby P, Simoes EA. 2009. Respir- Reid R, Hua W, Santosham M, Karron RA.
atory syncytial virus neutralizing antibodies in 2008. Young infants can develop protective
cord blood, respiratory syncytial virus hospitali- levels of neutralizing antibody after infection
zation, and recurrent wheeze. J Allergy Clin with respiratory syncytial virus. J Infect Dis
Immunol 123:398403. 198:10071015.
57. Falsey AR, Walsh EE. 1998. Relationship of 66. Yamazaki H, Tsutsumi H, Matsuda K, Nagai
serum antibody to risk of respiratory syncytial K, Ogra PL, Chiba S. 1994. Effect of maternal
virus infection in elderly adults. J Infect Dis antibody on IgA antibody response in naso-
177:463466. pharyngeal secretion in infants and children
CHAPTER 13 Prevention of RSV Infection: From Vaccine to Antibody 235

during primary respiratory syncytial virus HC, Fulton DR, Welliver RC. 1993. Prophy-
infection. J Gen Virol 75(Pt 8):21152119. lactic administration of respiratory syncytial
67. Kapikian AZ, Mitchell RH, Chanock RM, virus immune globulin to high-risk infants and
Shvedoff RA, Stewart CE. 1969. An epidemi- young children. The Respiratory Syncytial
ologic study of altered clinical reactivity to Virus Immune Globulin Study Group. N Engl
respiratory syncytial (RS) virus infection in J Med 329:15241530.
children previously vaccinated with an inacti- 77. The PREVENT Study Group. 1997. Reduction
vated RS virus vaccine. Am J Epidemiol of respiratory syncytial virus hospitalization
89:405421. among premature infants and infants with
68. Chanock RM, Parrott RH, Vargosko AJ, bronchopulmonary dysplasia using respiratory
Kapikian AZ, Knight V, Johnson KM. 1962. syncytial virus immune globulin prophylaxis.
Acute respiratory diseases of viral etiology. IV. Pediatrics 99:9399.
Respiratory syncytial virus. Am J Public Health 78. Rodriguez WJ, Gruber WC, Welliver RC,
52:918925. Groothuis JR, Simoes EA, Meissner HC,
69. Walsh EE, Schlesinger JJ, Brandriss MW. Hemming VG, Hall CB, Lepow ML, Rosas
1984. Protection from respiratory syncytial virus AJ, Robertsen C, Kramer AA. 1997. Respira-
infection in cotton rats by passive transfer of tory syncytial virus (RSV) immune globulin
monoclonal antibodies. Infect Immun 43:756758. intravenous therapy for RSV lower respiratory
70. Taylor G, Stott EJ, Bew M, Fernie BF, Cote tract infection in infants and young children at
PJ, Collins AP, Hughes M, Jebbett J. 1984. high risk for severe RSV infections: Respira-
Monoclonal antibodies protect against respi- tory Syncytial Virus Immune Globulin Study
ratory syncytial virus infection in mice. Immu- Group. Pediatrics 99:454461.
nology 52:137142. 79. Rodriguez WJ, Gruber WC, Groothuis JR,
71. Prince GA, Hemming VG, Horswood RL, Simoes EA, Rosas AJ, Lepow M, Kramer A,
Chanock RM. 1985. Immunoprophylaxis and Hemming V. 1997. Respiratory syncytial virus
immunotherapy of respiratory syncytial virus immune globulin treatment of RSV lower
infection in the cotton rat. Virus Res 3:193206. respiratory tract infection in previously
72. Hemming VG, Prince GA, Horswood RL, healthy children. Pediatrics 100:937942.
London WJ, Murphy BR, Walsh EE, Fischer 80. American Academy of Pediatrics. 1997. Measles,
GW, Weisman LE, Baron PA, Chanock RM. p 353. In Peter G (ed), Red Book: Report of the
1985. Studies of passive immunotherapy for Committee on Infectious Diseases, 24th ed. Amer-
infections of respiratory syncytial virus in the ican Academy of Pediatrics, Elk Grove Village, IL.
respiratory tract of a primate model. J Infect 81. Simoes EA, Sondheimer HM, Top FH, Jr,
Dis 152:10831087. Meissner HC, Welliver RC, Kramer AA,
73. Groothuis JR, Levin MJ, Rodriguez W, Hall Groothuis JR. 1998. Respiratory syncytial
CB, Long CE, Kim HW, Lauer BA, Hemming virus immune globulin for prophylaxis against
VG. 1991. Use of intravenous gamma globulin respiratory syncytial virus disease in infants and
to passively immunize high-risk children children with congenital heart disease. The
against respiratory syncytial virus: safety and Cardiac Study Group. J Pediatr 133:492499.
pharmacokinetics. The RSVIG Study Group. 82. Beeler JA, van Wyke Coelingh K. 1989.
Antimicrob Agents Chemother 35:14691473. Neutralization epitopes of the F glycoprotein
74. Meissner HC, Fulton DR, Groothuis JR, of respiratory syncytial virus: effect of mutation
Geggel RL, Marx GR, Hemming VG, Hougen upon fusion function. J Virol 63:29412950.
T, Snydman DR. 1993. Controlled trial to 83. Tempest PR, Bremner P, Lambert M, Taylor
evaluate protection of high-risk infants against G, Furze JM, Carr FJ, Harris WJ. 1991.
respiratory syncytial virus disease by using Reshaping a human monoclonal antibody to
standard intravenous immune globulin. inhibit human respiratory syncytial virus in-
Antimicrob Agents Chemother 37:16551658. fection in vivo. Biotechnology (N Y) 9:266271.
75. Siber GR, Leszcynski J, Pena-Cruz V, Ferren- 84. Everitt DE, Davis CB, Thompson K, DiCicco
Gardner C, Anderson R, Hemming VG, Walsh R, Ilson B, Demuth SG, Herzyk DJ, Jorkasky
EE, Burns J, McIntosh K, Gonin R. 1992. DK. 1996. The pharmacokinetics, antigenicity,
Protective activity of a human respiratory syncy- and fusion-inhibition activity of RSHZ19, a
tial virus immune globulin prepared from donors humanized monoclonal antibody to respiratory
screened by microneutralization assay. J Infect syncytial virus, in healthy volunteers. J Infect
Dis 165:456463. Dis 174:463469.
76. Groothuis JR, Simoes EA, Levin MJ, Hall CB, 85. Meissner HC, Groothuis JR, Rodriguez WJ,
Long CE, Rodriguez WJ, Arrobio J, Meissner Welliver RC, Hogg G, Gray PH, Loh R,
236 HUANG AND WU

Simoes EA, Sly P, Miller AK, Nichols AI, and binding valence on viral neutralization.
Jorkasky DK, Everitt DE, Thompson KA. J Mol Biol 350:126144.
1999. Safety and pharmacokinetics of an intra- 93. Wu H, Pfarr DS, Johnson S, Brewah YA,
muscular monoclonal antibody (SB 209763) Woods RM, Patel NK, White WI, Young JF,
against respiratory syncytial virus (RSV) in infants Kiener PA. 2007. Development of motavizumab,
and young children at risk for severe RSV disease. an ultra-potent antibody for the prevention
Antimicrob Agents Chemother 43:11831188. of respiratory syncytial virus infection in the
86. Weltzin R, Hsu SA, Mittler ES, Georgakopoulos upper and lower respiratory tract. J Mol Biol
K, Monath TP. 1994. Intranasal monoclonal 368:652665.
immunoglobulin A against respiratory syncytial 94. Carbonell-Estrany X, Simoes EA, Dagan R,
virus protects against upper and lower respiratory Hall CB, Harris B, Hultquist M, Connor EM,
tract infections in mice. Antimicrob Agents Losonsky GA, Motavizumab Study Group.
Chemother 38:27852791. 2010. Motavizumab for prophylaxis of respi-
87. Weltzin R, Traina-Dorge V, Soike K, Zhang ratory syncytial virus in high-risk children: a
JY, Mack P, Soman G, Drabik G, Monath TP. noninferiority trial. Pediatrics 125:e35e51.
1996. Intranasal monoclonal IgA antibody to doi:10.1542/peds.2008-1036.
respiratory syncytial virus protects rhesus 95. Feltes TF, Sondheimer HM, Tulloh RM,
monkeys against upper and lower respiratory Harris BS, Jensen KM, Losonsky GA, Griffin
tract infection. J Infect Dis 174:256261. MP, Motavizumab Cardiac Study Group.
88. Weltzin R, Monath TP. 1999. Intranasal 2011. A randomized controlled trial of mota-
antibody prophylaxis for protection against vizumab versus palivizumab for the prophylaxis
viral disease. Clin Microbiol Rev 12:383393. of serious respiratory syncytial virus disease in
89. Johnson S, Oliver C, Prince GA, Hemming children with hemodynamically significant con-
VG, Pfarr DS, Wang SC, Dormitzer M, genital heart disease. Pediatr Res 70:186191.
OGrady J, Koenig S, Tamura JK, Woods R, 96. Dallacqua WF, Kiener PA, Wu H. 2006.
Bansal G, Couchenour D, Tsao E, Hall WC, Properties of human IgG1s engineered for
Young JF. 1997. Development of a humanized enhanced binding to the neonatal Fc receptor
monoclonal antibody (MEDI-493) with potent (FcRn). J Biol Chem 281:2351423524.
in vitro and in vivo activity against respiratory 97. Robbie GJ, Criste R, Dallacqua WF, Jensen
syncytial virus. J Infect Dis 176:12151224. K, Patel NK, Losonsky GA, Griffin MP. 2013.
90. The IMpact-RSV Study Group. 1998. Palivi- A novel investigational Fc-modified human-
zumab, a humanized respiratory syncytial virus ized monoclonal antibody, motavizumab-YTE,
monoclonal antibody, reduces hospitalization Has an extended half-life in healthy adults.
from respiratory syncytial virus infection in high- Antimicrob Agents Chemother 57:61476153.
risk infants. Pediatrics 102:531537. 98. Kwakkenbos MJ, Diehl SA, Yasuda E, Bakker
91. Feltes TF, Cabalka AK, Meissner HC, Piazza AQ, van Geelen CM, Lukens MV, van Bleek
FM, Carlin DA, Top FH, Jr, Connor EM, GM, Widjojoatmodjo MN, Bogers WM, Mei
Sondheimer HM, Cardiac Synagis Study H, Radbruch A, Scheeren FA, Spits H,
Group. 2003. Palivizumab prophylaxis reduces Beaumont T. 2010. Generation of stable
hospitalization due to respiratory syncytial monoclonal antibody-producing B cell recep-
virus in young children with hemodynamically tor-positive human memory B cells by genetic
significant congenital heart disease. J Pediatr programming. Nat Med 16:123128.
143:532540. 99. Huang K, Incognito L, Cheng X, Ulbrandt
92. Wu H, Pfarr DS, Tang Y, An LL, Patel NK, ND, Wu H. 2010. Respiratory syncytial virus-
Watkins JD, Huse WD, Kiener PA, Young JF. neutralizing monoclonal antibodies mota-
2005. Ultra-potent antibodies against respira- vizumab and palivizumab inhibit fusion. J Virol
tory syncytial virus: effects of binding kinetics 84:81328140.
Human Metapneumovirus

JENNIFER E. SCHUSTER1 and JOHN V. WILLIAMS2,3


14
INTRODUCTION

Human metapneumovirus (HMPV), a paramyxovirus first discovered in


2001, is a significant cause of respiratory tract disease in children and adults
(1). Humoral immunity plays an important role in HMPV infection, and
the study of HMPV antibodies provides important clinical information
including the seroprevalence of HMPV, age of primary infection, serological
cross-protection between HMPV subgroups, evaluation of vaccine immu-
nogenicity, and strategies for prophylaxis and therapy using monoclonal
antibodies (mAbs).

SEROPREVALENCE OF HMPV INFECTION

Primary HMPV infection in children can be determined by seroconversion,


which typically occurs within the first 1 to 2 years of life. In an Israeli
cohort, 80% of children 2 months of age had evidence of HMPV antibodies,
reflective of the broad adult seroprevalence and maternal transmission of

1
Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108-4619; 2Department of Pediatrics,
Vanderbilt University School of Medicine, Nashville, TN 37232-2581; 3Department of Pathology, Microbiology,
and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232-2581.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0020-2014

237
238 SCHUSTER AND WILLIAMS

immunity. Consistent with waning maternally one study, young adults had the highest rates
derived antibodies, only 30% of children of asymptomatic infection at 4% (8).
at 13 months were seropositive. However,
52% of children had HMPV antibodies at
24 months, suggesting that primary infection ANTIBODY SPECIFICITY
had occurred (2). By school age, nearly all
children have been infected with HMPV. In a Avian pneumovirus (APV) is the other mem-
Japanese cohort, 77% of children 2 to 5 years ber of the Metapneumovirus genus, and re-
of age were seropositive and 100% of children searchers postulate that HMPV diverged from
>10 years of age had HMPV antibodies (3). APV type C 200 to 400 years ago (11, 12, 13).
Reinfection occurs throughout childhood and The N protein is 80% conserved between
can be assessed by repeated measurements of APV/A, B, and C, and 100% conserved be-
serology. In one cohort, HMPV-infected chil- tween APV/C and HMPV. An N-protein
dren had both a positive IgM and IgG at the polyclonal antibody, which targets a conserved
time of acute infection, indicating a history of peptide, cross-reacted with all members of
previous infection. These children had a 4-fold APV and HMPV, while an anti-N mAb cross-
IgG rise in convalescent serology, suggesting reacted with both APV/C and HMPV (14). Sera
that a positive IgG is not entirely protective from animals infected with APV/C cross-
(4). In a cohort of Thai children, 99.7% had reacted with HMPV fusion (F) protein, and
positive HMPV serology. As they were sera from HMPV-infected animals cross-
followed longitudinally, 5% had evidence of reacted with APV/C F protein. However,
reinfection, as defined by a 4-fold serology HMPV polyclonal sera did not cross-react
rise, during the 4-year study period (5). HMPV with APV/A or B (15), consistent with the
seroconversion typically occurs later than closer relationship of HMPV to APV/C. Sera
respiratory syncytial virus (RSV). In children from animals infected with RSV, a member of
aged 4 to 11 months, 48% had positive RSV the same subfamily as HMPV, do not neutral-
antibody titers, but only 11% had anti-HMPV ize HMPV in vitro and vice versa (16). Thus,
antibodies. In children over the age of 2 years, HMPV is serologically distinct from other
seroprevalence is similar (6, 7). related pneumoviruses.
Almost all adults have serologic evidence
of prior HMPV infection. Older adults are at
increased risk of severe disease, which some ANTIBODY CROSS-PROTECTION
researchers postulate is due to waning immu-
nity (8). However, in a German cohort, >90% Four genetic subgroups of HMPV (A1, A2,
of people 60 to 89 years of age had neutralizing B1, and B2) circulate with year-to-year vari-
antibodies (9). Similarly, Japanese adults were ability (17) (Fig. 1). The subgroups are
followed longitudinally to determine whether antigenically related with some evidence of
the presence of HMPV antibody was protec- cross-neutralization (18). In hamsters, serum
tive. Nine subjects with baseline positive titers neutralization titers showed a 48% antigenic
by enzyme-linked immunosorbent assay relatedness between subgroups A and B, and,
(ELISA) and neutralization assay became in nonhuman primates, titers were 64 to 99%
infected with HMPV during the study period. related (19). HMPV-infected hamsters were
Infection was associated with a rise in anti- protected against subsequent challenge with a
body titer by both methods, demonstrating heterologous subgroup virus. Sera from in-
that a positive antibody titer is not fully fected hamsters neutralized both homologous
protective (10). and heterologous subgroups in vitro; however,
Last, seroconversion can be used to detect the neutralizing capacity against the heterolo-
asymptomatic HMPV infection in adults. In gous subgroup was reduced by 16-fold. African
CHAPTER 14 Human Metapneumovirus 239

green monkeys and macaques displayed similar Antibody titers at 58 weeks after the initial
cross-protection (20, 21). In humans, sera from infection were much lower, animals were
HMPV-infected children cross-neutralized completely unprotected against heterologous
other subgroups with varying degrees of effi- reinfection, and 2 of 3 animals were not
cacy. For example, sera from an HMPV A2- protected against homologous reinfection.
infected child neutralized A2 and B2 viruses, The remaining animal had partial, but not
but had no activity against B1 in vitro (22). full, protection against infection. Interestingly,
The F protein is highly conserved and infection with NL/1/00, a subgroup A virus,
immunogenic (13, 18, 23). Antigenic map- induced higher neutralizing titers than NL/1/
ping studies using mAbs defined a number 99, a subgroup B virus (21). Thus, animal studies
of antigenic regions on HMPV F analogous suggest that, while antibodies can mediate
to sites described on RSV F (Fig. 2) (24, 25). protection, titers wane over time. This phe-
Sera from hamsters infected with recombinant nomenon likely contributes to the capacity of
parainfluenza 1 (rPIV1) expressing HMPV F HMPV to reinfect humans throughout life.
neutralized both homologous and heterolo-
gous HMPV viruses in vitro. Thus, the F
protein is a mediator of antibody cross-protec- AGE-RELATED ANTIBODY
tion, and it likely mediates some, although not DEVELOPMENT
full, protection against reinfection (19).
Unlike F, the glycoprotein (G) is highly Older adults are an at-risk population for
variable and contains only 31 to 35% similarity severe respiratory viral infections, including
between the A and B subgroups (12, 26). HMPV. One contributing factor is thought to
Convalescent sera from HMPV-infected chil- be waning humoral immunity. In a mouse
dren reacted with only one G protein, pre- model, both virus-specific and neutralizing
sumably the infecting subgroup. This antibody responses were higher in younger
specificity would suggest that the different mice infected with HMPV than in older mice
HMPV subgroups could be considered sero- (32). However, in a cohort of nonhospitalized
types if only categorized by the G protein (27); adults infected with HMPV, older adults had
however, F is thought to be the primary target a significantly higher acute HMPV titer com-
of neutralizing and protective antibodies. Sev- pared with young adults. Older adults also had
eral different approaches have shown that a significantly higher convalescent titer con-
G antibodies are not neutralizing in vitro or tributing to a significantly higher overall rise
protective in vivo (28, 29, 30). in titer compared with young adults. Older
adults had a trend toward a higher rise in
neutralizing antibody, although this was not
ANTIBODY RESPONSE TO INFECTION significant (33).

Twenty-one days postinfection, HMPV-


infected cotton rats developed a neutralizing IMMUNOGLOBULIN CLASSES
antibody response. The average serum-
neutralizing titer was 1:180, which correlated IgG and IgA are produced after HMPV
with lung protection (31). In macaques infection. In a BALB/c mouse model, IgG1
infected with wild-type (WT) HMPV, anti- and IgG2a were detectable 5 days after HMPV
body titer and efficacy waned over time, infection, and antibody titer peaked at day 8.
leading to symptomatic reinfection. Repeat No IgA or IgE was detected (34). IgG2a
inoculations of HMPV boosted neutralizing antibodies were the dominant immune re-
antibodies, but, despite boosting, antibody sponse in mice immunized with viruslike
titer waned 18 weeks after initial infection. particles (VLPs) (35).
240 SCHUSTER AND WILLIAMS
CHAPTER 14 Human Metapneumovirus 241

In an adult cohort, HMPV titers in patients rHMPV; however, the deletion mutant was
with acute infection were compared with not attenuated. Hamsters infected with the
HMPV titers in uninfected patients. Serum attenuated viruses HMPVDG and HMPVDSH/
IgG titer, but not nasal IgA, was associated G mounted a neutralizing antibody response,
with the likelihood of developing HMPV but at 6-fold lower titer than rHMPV, and the
reinfection (11.93 1.25 log2 in infected animals were not fully protected from chal-
individuals vs 12.86 1.23 in uninfected lenge (36). Therefore, the SH protein is not
individuals, P = 0.001). Although infected immunogenic and the G protein is weakly
adults had lower HMPV IgG titers, the immunogenic. Other immunogenic, attenuated
protective level of IgG remains unclear. viruses include temperature-sensitive HMPV,
HMPV-infected adults had significantly which was immunogenic and protective in
lower neutralizing titers compared with unin- hamsters (37); a virus lacking an N-linked
fected adults. In a longitudinal study, 71% of carbohydrate in the F protein, which was
adults with a baseline microneutralization immunogenic and protective in mice (38);
assay (MNA) titer of 10.5 log2 were infected and viruses with the F protein RGD-binding
compared with 36% infected among adults sequence mutated, which was immunogenic
with a baseline MNA titer >10.5 log2 (33). These and protective in cotton rats (39). The con-
data suggest that there may be a minimum served RGD motif of the F protein serves to
protective threshold of serum-neutralizing bind integrins as receptors for HMPV (40, 41).
antibody titer. In a phase 1 clinical trial, humans with
negative HMPV serology were infected
with rHMPV-SH (containing a stabilization
INDUCTION OF ANTIBODIES mutation in the SH protein). Only 15% had a
BY IMMUNIZATION 4-fold increase in IgG to HMPV F, 20%
had a 4-fold increase in serum HMPV F
An effective HMPV vaccine will need to induce IgA, and 30% had a 4-fold rise in nasal
an antibody response, and the goal is a high wash IgA (42). Thus, the development of a
neutralizing antibody titer. Potential types of live attenuated vaccine that will produce an
immunizations include live attenuated HMPV effective neutralizing antibody response in
strains, inactivated virus, DNA and protein previously infected adults may be difficult.
vaccines, and VLPs. DNA and protein vaccines Formalin-inactivated (FI) HMPV is immu-
and VLPs will be discussed separately under nogenic in cotton rats. The vaccine induced
antigenic proteins. neutralizing antibodies, and animals were
Live attenuated HMPV vaccines include protected against HMPV challenge (43). How-
strains containing gene deletions or other ever, the vaccine was less effective in ma-
attenuating mutations. Golden Syrian ham- caques. One dose of FI-HMPV induced binding
sters infected with HMPVDSH (deleted antibodies, but 2 doses were needed to yield
short hydrophobic protein) mounted a sim- neutralizing antibodies. Antibody titer rapidly
ilar neutralizing antibody response to declined 4 weeks later, and animals were not

FIGURE 1 Maximum clade credibility tree of HMPV and avian metapneumovirus (AMPV) F nucleotide
diversity. Phylogenetic analysis of 85 full-length HMPV F nucleotide sequences from Canada (CAN),
Japan (JPS or JPY), Tennessee (TN), or the Netherlands (NL) and 16 AMPV F sequences. The rst two digits
of the HMPV sequence names indicate the year of the isolate. The names of the AMPV sequences indicate
geographic origin (US, United States; UK, United Kingdom; MN, Minnesota) and year. The posterior
probability of divergence is indicated at each node. Scale bar represents time in years. Reprinted from
reference 13 with permission of the publisher http://creativecommons.org/licenses/by/2.0/legalcode.
doi:10.1128/microbiolspec.AID-0020-2014.f1
242 SCHUSTER AND WILLIAMS

FIGURE 2 Schematic of HMPV and RSV F protein domain structures and relative location of MARM
mutation sites. Indicated are the N terminus (N), signal peptide (SP), fusion peptide (FP), heptad repeat 1
(HR1), heptad repeat 2 (HR2), transmembrane domain (TM), and C terminus (C), as well as the F1 and F2
segments of the F protein. The amino acid positions that border domains (predicted) or cleavage sites
and C termini (known) are indicated. Also depicted are the relative positions of the HMPV epitope group
MARM mutations and corresponding MARM mutation sites on the RSV F protein. Reprinted with
permission from Ulbrandt ND, Ji H, Patel NK, Barnes AS, Wilson S, Kiener PA, Suzich J, McCarthy MP.
2008. J Gen Virol 89:31133118. Copyright 2008 Society for General Microbiology. Permission conveyed
through Copyright Clearance Center, Inc. doi:10.1128/microbiolspec.AID-0020-2014.f2

protected against challenge 15 weeks after a antibodies, which conferred lower airway
third immunization (44). Furthermore, FI- protection. The G protein, expressed by
HMPV was associated with enhanced dis- rPIV-1, was less immunogenic than WT
ease upon HMPV challenge in cotton rats HMPV and did not induce neutralizing anti-
and macaques, similar to that observed with bodies. Last, the SH protein did not induce
FI-RSV vaccine in the 1960s (43, 44). any binding or neutralizing antibodies. Nei-
ther rPIV-1/G nor rPIV-1/SH were asso-
ciated with significant protection against viral
ANTIGENIC PROTEINS replication upon HMPV challenge. Therefore,
the F protein is the main antigenic determi-
The HMPV genome contains 8 genes that nant (28).
encode 9 proteins (1). The F, G, and SH protein In Syrian golden hamsters, chimeric bovine/
are outer membrane proteins and likely human PIV3 containing HMPV F induced a
targets for antibodies. To identify antigenic neutralizing antibody response equivalent
determinants, each of these 3 proteins was to WT HMPV infection (45). This chimeric
inserted into rPIV-1, a model vaccine system. virus also was immunogenic in African green
In hamsters, immunization with PIV-1/ monkeys, but neutralizing antibody titers were
HMPV F induced an antibody titer similar to lower compared with animals infected with
WT HMPV and a high level of neutralizing WT HMPV (46).
CHAPTER 14 Human Metapneumovirus 243

Similarly, soluble F-protein vaccines, gen- MONOCLONAL ANTIBODIES


erated from both A and B subgroup lineage,
are protective in hamsters, and F-specific Since the HMPV F protein is highly immu-
antibodies were present in all animals. Ani- nogenic and protective, F-specific mAbs could
mals immunized with the A-lineage F protein have clinical use in high-risk hosts, similar to
had higher titers compared with B-lineage F, the respiratory syncytial virus anti-F protein
and an adjuvanted F protein was more immu- mAb, palivizumab (53). Murine mAbs gener-
nogenic than an unadjuvanted F protein. ated against the F protein of HMPV neutral-
Adjuvanted F induced a high neutralizing ized both A and B subgroups of the virus. Two
antibody titer (47). F-protein vaccines were of these mAbs were effective prophylactically
more immunogenic than DNA vaccines in in a hamster model. Although viral replication
a cotton rat model, and the protein vaccine was minimally decreased in the nasal tur-
yielded high neutralizing antibody titers binates, hamsters did not have replicating
(mean 1:570) (23). VLPs containing the F virus in the lungs at doses of 3 mg/kg. These
and matrix (M) proteins are immunogenic two mAbs, 234 and 338, bound to the F protein
and protective in a mouse model. After two of both the A and B subgroups with nanomolar
immunizations, animals had an F-specific affinity (54). Mice prophylaxed or treated
antibody response similar to WT HMPV and postinfection with mAb 338 had decreased
were protected against challenge (48). lung histopathology and airway obstruction
Although the F protein appears to be the compared with mock treated mice (55). DS7,
primary driver of humoral immunity, the G a fully human mAb, neutralized all four
protein does induce an antibody response. subgroups of HMPV in vitro, and intranasal
In a cotton rat model, soluble G protein was DS7 had therapeutic efficacy against HMPV
immunogenic, but it was not protective (30). infection in a cotton rat model. The DS7 mAb
Mice immunized with the G protein in a also had subnanomolar affinity for the F
vaccinia virus system developed a G-specific protein (24).
antibody response that neutralized homolo- Monoclonal antibody resistant mutants
gous but not heterologous virus (49). However, (MARMs) have identified 5 epitopes on the F
VLPs expressing the G protein were not immu- protein. Two of these epitopes have analogous
nogenic, unlike VLPs containing both the F epitopes on the RSV F protein (Fig. 2) (25).
and G proteins (35). Although SH is generally Crystallization of DS7 with the F protein
not immunogenic, a truncated version of the identified a novel antigenic site on the protein,
protein expressed by vaccinia virus induced a which is a highly conserved area among
modest neutralizing antibody response (49). HMPV subgroups (56).
In humans, the antibody response is The F protein is only 33% conserved
much more diverse. In an Irish cohort, anti- between RSV and HMPV, and polyclonal
bodies to HMPV M, nucleoprotein (N), and sera against one does not cross-react with
phosphoprotein (P) were identified (50). the other (1, 16). However, one human mAb
Antibody responses to N and M typically isolated from screening 114,000 B cells neu-
occurred only in sera with high neutralizing tralized both viruses. This mAb, MPE8, is an
titers (generally >1:160) (51). Other groups anti-RSV F mAb in its germ line configuration,
have postulated that many F and G antibodies but it contains light chain mutations in the
may be conformation dependent, whereas variable region, affording it activity against
antibodies to N and P may recognize polypep- HMPV. MPE8 had prophylactic efficacy
tides (52). Thus, an effective vaccine strategy against HMPV and RSV and prevented death
might include whole outer membrane proteins in a mouse model of pneumonia virus of
in conjunction with immunogenic areas of mice, another paramyxovirus. The epitope is
internal proteins. distinct from, but in close proximity to, the
244 SCHUSTER AND WILLIAMS

epitope recognized by palivizumab (57). An- Osterhaus AD. 2001. A newly discovered
other group reported a human mAb isolated by human pneumovirus isolated from young chil-
dren with respiratory tract disease. Nat Med
screening B cells against HMPV F protein 7:719724.
by ELISA (58). This mAb, 54G10, exhibited 2. Wolf DG, Zakay-Rones Z, Fadeela A, Greenberg
broadly neutralizing activity in vitro against all D, Dagan R. 2003. High seroprevalence of human
4 subgroups of HMPV and bound to recom- metapneumovirus among young children in
Israel. J Infect Dis 188:18651867.
binant HMPV F protein with subnanomolar
3. Ebihara T, Endo R, Kikuta H, Ishiguro N,
affinity. 54G10 provided potent prophylactic Yoshioka M, Ma X, Kobayashi K. 2003. Sero-
efficacy against all 4 HMPV subgroups in a prevalence of human metapneumovirus in Japan.
mouse model, as well as therapeutic efficacy J Med Virol 70:281283.
(tested only against A2). The generation 4. Ebihara T, Endo R, Kikuta H, Ishiguro N,
of MARMs identified the 54G10 epitope as Ishiko H, Hara M, Takahashi Y, Kobayashi K.
2004. Human metapneumovirus infection in
a region relatively conserved in RSV F and Japanese children. J Clin Microbiol 42:126132.
other paramyxoviruses (59). Consistent with 5. Pavlin JA, Hickey AC, Ulbrandt N, Chan YP,
a shared epitope, 54G10 neutralized RSV Endy TP, Boukhvalova MS, Chunsuttiwat S,
in vitro and exhibited both prophylactic and Nisalak A, Libraty DH, Green S, Rothman
therapeutic efficacy in a mouse model (58). AL, Ennis FA, Jarman R, Gibbons RV, Broder
CC. 2008. Human metapneumovirus reinfec-
The discovery of mAbs with activity against tion among children in Thailand determined
both HMPV and RSV raises the possibility of by ELISA using purified soluble fusion protein.
not only clinical antibodies that target both J Infect Dis 198:836842.
viruses, but also epitope-based vaccines that 6. Ebihara T, Endo R, Kikuta H, Ishiguro N,
Ishiko H, Kobayashi K. 2004. Comparison of the
elicit neutralizing antibodies against two dis-
seroprevalence of human metapneumovirus and
tinct viruses. human respiratory syncytial virus. J Med Virol
72:304306.
7. Dunn SR, Ryder AB, Tollefson SJ, Xu M,
CONCLUSIONS Saville BR, Williams JV. 2013. Seroepidemi-
ologies of human metapneumovirus and respira-
tory syncytial virus in young children, determined
Humoral immunity is important in HMPV with a new recombinant fusion protein enzyme-
infection. Further studies are needed to eluci- linked immunosorbent assay. Clin Vaccine
date the level of protective antibody, both after Immunol 20:16541656.
infection and after immunization, as well as to 8. Falsey AR, Erdman D, Anderson LJ, Walsh
explore the utility of monoclonal antibodies EE. 2003. Human metapneumovirus infections in
young and elderly adults. J Infect Dis 187:785790.
for prophylaxis and therapy against HMPV. 9. Lusebrink J, Wiese C, Thiel A, Tillmann RL,
Ditt V, Muller A, Schildgen O, Schildgen V.
2010. High seroprevalence of neutralizing
ACKNOWLEDGMENT capacity against human metapneumovirus in
all age groups studied in Bonn, Germany. Clin
Conflicts of interest: We disclose no conflicts.
Vaccine Immunol 17:481484.
10. Okamoto M, Sugawara K, Takashita E, Muraki
Y, Hongo S, Nishimura H, Matsuzaki Y. 2010.
CITATION
Longitudinal course of human metapneumovirus
Schuster JE, Williams JV. 2014. Human meta- antibody titers and reinfection in healthy adults.
pneumovirus. Microbiol Spectrum 2(5):AID- J Med Virol 82:20922096.
11. de Graaf M, Osterhaus AD, Fouchier RA,
0020-2014. Holmes EC. 2008. Evolutionary dynamics of
human and avian metapneumoviruses. J Gen
Virol 89:29332942.
REFERENCES
12. Yang CF, Wang CK, Tollefson SJ, Lintao LD,
1. van den Hoogen BG, de Jong JC, Groen J, Liem A, Chu M, Williams JV. 2013. Human
Kuiken T, de Groot R, Fouchier RA, metapneumovirus G protein is highly conserved
CHAPTER 14 Human Metapneumovirus 245

within but not between genetic lineages. Arch human metapneumovirus genotypes and geno-
Virol 158:12451252. type-specific seroprevalence in Yamagata,
13. Yang CF, Wang CK, Tollefson SJ, Piyaratna Japan. J Med Virol 80:10841089.
R, Lintao LD, Chu M, Liem A, Mark M, 23. Cseke G, Wright DW, Tollefson SJ, Johnson
Spaete RR, Crowe JE, Jr, Williams JV. 2009. JE, Crowe JE, Jr, Williams JV. 2007. Human
Genetic diversity and evolution of human metapneumovirus fusion protein vaccines that
metapneumovirus fusion protein over twenty are immunogenic and protective in cotton rats.
years. Virol J 6:138. doi:10.1186/1743-422X-6-138. J Virol 81:698707.
14. Alvarez R, Jones LP, Seal BS, Kapczynski DR, 24. Williams JV, Chen Z, Cseke G, Wright DW,
Tripp RA. 2004. Serological cross-reactivity of Keefer CJ, Tollefson SJ, Hessell A, Podsiad
members of the Metapneumovirus genus. Virus A, Shepherd BE, Sanna PP, Burton DR,
Res 105:6773. Crowe JE, Jr, Williamson RA. 2007. A recom-
15. Luo L, Sabara MI, Li Y. 2009. Analysis of binant human monoclonal antibody to human
antigenic cross-reactivity between subgroup C metapneumovirus fusion protein that neutralizes
avian pneumovirus and human metapneu- virus in vitro and is effective therapeutically in
movirus by using recombinant fusion proteins. vivo. J Virol 81:83158324.
Transbound Emerg Dis 56:303310. 25. Ulbrandt ND, Ji H, Patel NK, Barnes AS,
16. Wyde PR, Chetty SN, Jewell AM, Boivin G, Wilson S, Kiener PA, Suzich J, McCarthy MP.
Piedra PA. 2003. Comparison of the inhibition 2008. Identification of antibody neutralization
of human metapneumovirus and respiratory epitopes on the fusion protein of human meta-
syncytial virus by ribavirin and immune serum pneumovirus. J Gen Virol 89:31133118.
globulin in vitro. Antiviral Res 60:5159. 26. Ishiguro N, Ebihara T, Endo R, Ma X, Kikuta
17. Williams JV, Wang CK, Yang CF, Tollefson H, Ishiko H, Kobayashi K. 2004. High genetic
SJ, House FS, Heck JM, Chu M, Brown JB, diversity of the attachment (G) protein of human
Lintao LD, Quinto JD, Chu D, Spaete RR, metapneumovirus. J Clin Microbiol 42:34063414.
Edwards KM, Wright PF, Crowe JE, Jr. 2006. 27. Endo R, Ebihara T, Ishiguro N, Teramoto S,
The role of human metapneumovirus in upper Ariga T, Sakata C, Hayashi A, Ishiko H, Kikuta
respiratory tract infections in children: a 20-year H. 2008. Detection of four genetic subgroup-
experience. J Infect Dis 193:387395. specific antibodies to human metapneumovirus
18. Peret TC, Boivin G, Li Y, Couillard M, attachment (G) protein in human serum. J Gen
Humphrey C, Osterhaus AD, Erdman DD, Virol 89:19701977.
Anderson LJ. 2002. Characterization of 28. Skiadopoulos MH, Biacchesi S, Buchholz UJ,
human metapneumoviruses isolated from patients Amaro-Carambot E, Surman SR, Collins PL,
in North America. J Infect Dis 185:16601663. Murphy BR. 2006. Individual contributions
19. Skiadopoulos MH, Biacchesi S, Buchholz UJ, of the human metapneumovirus F, G, and SH
Riggs JM, Surman SR, Amaro-Carambot E, surface glycoproteins to the induction of neutral-
McAuliffe JM, Elkins WR, St Claire M, izing antibodies and protective immunity. Virol-
Collins PL, Murphy BR. 2004. The two ogy 345:492501.
major human metapneumovirus genetic lineages 29. Mok H, Tollefson SJ, Podsiad AB, Shepherd
are highly related antigenically, and the fusion (F) BE, Polosukhin VV, Johnston RE, Williams
protein is a major contributor to this antigenic JV, Crowe JE, Jr. 2008. An alphavirus repli-
relatedness. J Virol 78:69276937. con-based human metapneumovirus vaccine is
20. MacPhail M, Schickli JH, Tang RS, Kaur J, immunogenic and protective in mice and cotton
Robinson C, Fouchier RA, Osterhaus AD, rats. J Virol 82:1141011418.
Spaete RR, Haller AA. 2004. Identification of 30. Ryder AB, Tollefson SJ, Podsiad AB, Johnson
small-animal and primate models for evaluation JE, Williams JV. 2010. Soluble recombinant
of vaccine candidates for human metapneu- human metapneumovirus G protein is immuno-
movirus (hMPV) and implications for hMPV genic but not protective. Vaccine 28:41454152.
vaccine design. J Gen Virol 85:16551663. 31. Williams JV, Tollefson SJ, Johnson JE,
21. van den Hoogen BG, Herfst S, de Graaf M, Crowe JE, Jr. 2005. The cotton rat (Sigmodon
Sprong L, van Lavieren R, van Amerongen G, hispidus) is a permissive small animal model of
Yuksel S, Fouchier RA, Osterhaus AD, de Swart human metapneumovirus infection, pathogenesis,
RL. 2007. Experimental infection of macaques and protective immunity. J Virol 79:1094410951.
with human metapneumovirus induces transient 32. Darniot M, Pitoiset C, Petrella T, Aho S, Pothier
protective immunity. J Gen Virol 88:12511259. P, Manoha C. 2009. Age-associated aggravation
22. Matsuzaki Y, Itagaki T, Abiko C, Aoki Y, of clinical disease after primary metapneumovirus
Suto A, Mizuta K. 2008. Clinical impact of infection of BALB/c mice. J Virol 83:33233332.
246 SCHUSTER AND WILLIAMS

33. Falsey AR, Hennessey PA, Formica MA, formalin-inactivated virus vaccine and chal-
Criddle MM, Biear JM, Walsh EE. 2010. lenged. Vaccine 25:50345040.
Humoral immunity to human metapneumovirus 44. de Swart RL, van den Hoogen BG, Kuiken T,
infection in adults. Vaccine 28:14771480. Herfst S, van Amerongen G, Yuksel S, Sprong
34. Darniot M, Petrella T, Aho S, Pothier P, L, Osterhaus AD. 2007. Immunization of
Manoha C. 2005. Immune response and alter- macaques with formalin-inactivated human
ation of pulmonary function after primary human metapneumovirus induces hypersensitivity to
metapneumovirus (hMPV) infection of BALB/c hMPV infection. Vaccine 25:85188528.
mice. Vaccine 23:44734480. 45. Tang RS, Schickli JH, MacPhail M, Fernandes
35. Levy C, Aerts L, Hamelin ME, Granier C, F, Bicha L, Spaete J, Fouchier RA, Osterhaus
Szecsi J, Lavillette D, Boivin G, Cosset FL. AD, Spaete R, Haller AA. 2003. Effects of human
2013. Virus-like particle vaccine induces cross- metapneumovirus and respiratory syncytial virus
protection against human metapneumovirus antigen insertion in two 3 proximal genome
infections in mice. Vaccine 31:27782785. positions of bovine/human parainfluenza virus
36. Biacchesi S, Skiadopoulos MH, Yang L, type 3 on virus replication and immunogenicity.
Lamirande EW, Tran KC, Murphy BR, J Virol 77:1081910828.
Collins PL, Buchholz UJ. 2004. Recombinant 46. Tang RS, Mahmood K, Macphail M, Guzzetta
human Metapneumovirus lacking the small JM, Haller AA, Liu H, Kaur J, Lawlor HA,
hydrophobic SH and/or attachment G glyco- Stillman EA, Schickli JH, Fouchier RA,
protein: deletion of G yields a promising Osterhaus AD, Spaete RR. 2005. A host-range
vaccine candidate. J Virol 78:1287712887. restricted parainfluenza virus type 3 (PIV3) ex-
37. Herfst S, de Graaf M, Schrauwen EJ, Sprong pressing the human metapneumovirus (hMPV)
L, Hussain K, van den Hoogen BG, Osterhaus fusion protein elicits protective immunity in
AD, Fouchier RA. 2008. Generation of tem- African green monkeys. Vaccine 23:16571667.
perature-sensitive human metapneumovirus 47. Herfst S, de Graaf M, Schrauwen EJ, Ulbrandt
strains that provide protective immunity in ND, Barnes AS, Senthil K, Osterhaus AD,
hamsters. J Gen Virol 89:15531562. Fouchier RA, van den Hoogen BG. 2007.
38. Liu P, Shu Z, Qin X, Dou Y, Zhao Y, Zhao X. Immunization of Syrian golden hamsters with F
2013. A live attenuated human metapneumovirus subunit vaccine of human metapneumovirus
vaccine strain provides complete protection induces protection against challenge with homol-
against homologous viral infection and cross- ogous or heterologous strains. J Gen Virol
protection against heterologous viral infection in 88:27022709.
BALB/c mice. Clin Vaccine Immunol 20:12461254. 48. Cox RG, Erickson JJ, Hastings AK, Becker JC,
39. Wei Y, Zhang Y, Cai H, Mirza AM, Iorio RM, Johnson M, Craven RE, Tollefson SJ, Boyd KL,
Peeples ME, Niewiesk S, Li J. 2014. Roles of Williams JV. 2014. Human metapneumovirus
the putative integrin-binding motif of the virus-like particles induce protective B and T cell
human metapneumovirus fusion (f) protein in responses in a mouse model. J Virol 88:63686379.
cell-cell fusion, viral infectivity, and pathogen- 49. Tedcastle AB, Fenwick F, Robinson MJ,
esis. J Virol 88:43384352. Toms GL. 2014. Immunogenicity in mice of
40. Cox RG, Livesay SB, Johnson M, Ohi MD, human metapneumovirus with a truncated SH
Williams JV. 2012. The human metapneumovirus glycoprotein. J Med Virol 86:547557.
fusion protein mediates entry via an interaction 50. OShaughnessy L, Carr M, Crowley B,
with RGD-binding integrins. J Virol 86:1214812160. Carberry S, Doyle S. 2011. Recombinant expres-
41. Cseke G, Maginnis MS, Cox RG, Tollefson SJ, sion and immunological characterisation of pro-
Podsiad AB, Wright DW, Dermody TS, teins derived from human metapneumovirus.
Williams JV. 2009. Integrin alphavbeta1 J Clin Virol 52:236243.
promotes infection by human metapneumovirus. 51. Ishiguro N, Ebihara T, Endo R, Ma X, Kawai
Proc Natl Acad Sci USA 106:15661571. E, Ishiko H, Kikuta H. 2006. Detection of
42. Talaat KR, Karron RA, Thumar B, McMahon antibodies against human metapneumovirus by
BA, Schmidt AC, Collins PL, Buchholz UJ. Western blot using recombinant nucleocapsid
2013. Experimental infection of adults with and matrix proteins. J Med Virol 78:10911095.
recombinant wild-type human metapneumovirus. 52. Tedcastle AB, Fenwick F, Ingram RE, King
J Infect Dis 208:16691678. BJ, Robinson MJ, Toms GL. 2012. The char-
43. Yim KC, Cragin RP, Boukhvalova MS, Blanco acterization of monoclonal antibodies to human
JC, Hamlin ME, Boivin G, Porter DD, Prince metapneumovirus and the detection of multiple
GA. 2007. Human metapneumovirus: enhanced forms of the virus nucleoprotein and phospho-
pulmonary disease in cotton rats immunized with protein. J Med Virol 84:10611070.
CHAPTER 14 Human Metapneumovirus 247

53. Pediatrics. 1998. Palivizumab, a humanized Structure of the human metapneumovirus fusion
respiratory syncytial virus monoclonal antibody, protein with neutralizing antibody identifies a
reduces hospitalization from respiratory syncytial pneumovirus antigenic site. Nat Struct Mol Biol
virus infection in high-risk infants. The IMpact- 19:461463.
RSV Study Group. Pediatrics 102:531537. 57. Corti D, Bianchi S, Vanzetta F, Minola A, Perez
54. Ulbrandt ND, Ji H, Patel NK, Riggs JM, L, Agatic G, Guarino B, Silacci C, Marcandalli J,
Brewah YA, Ready S, Donacki NE, Folliot K, Marsland BJ, Piralla A, Percivalle E, Sallusto
Barnes AS, Senthil K, Wilson S, Chen M, Clarke F, Baldanti F, Lanzavecchia A. 2013. Cross-
L, MacPhail M, Li J, Woods RM, Coelingh K, neutralization of four paramyxoviruses by a
Reed JL, McCarthy MP, Pfarr DS, Osterhaus human monoclonal antibody. Nature 501:439443.
AD, Fouchier RA, Kiener PA, Suzich JA. 2006. 58. Schuster JE, Cox RG, Hastings AK, Boyd KL,
Isolation and characterization of monoclonal Wadia J, Chen Z, Burton DR, Williamson RA,
antibodies which neutralize human metapneu- Williams JV. 2014. A broadly neutralizing human
movirus in vitro and in vivo. J Virol 80:77997806. monoclonal antibody exhibits in vivo efficacy
55. Hamelin ME, Gagnon C, Prince GA, Kiener against both human metapneumovirus and respi-
P, Suzich J, Ulbrandt N, Boivin G. 2010. ratory syncytial virus. J Infect Dis doi:10.1093/
Prophylactic and therapeutic benefits of a infdis/jiu307.
monoclonal antibody against the fusion protein 59. McLellan JS, Chen M, Chang JS, Yang Y, Kim A,
of human metapneumovirus in a mouse model. Graham BS, Kwong PD. 2010. Structure of a
Antiviral Res 88:3137. major antigenic site on the respiratory syncytial
56. Wen X, Krause JC, Leser GP, Cox RG, Lamb RA, virus fusion glycoprotein in complex with neu-
Williams JV, Crowe JE, Jr, Jardetzky TS. 2012. tralizing antibody 101F. J Virol 84:1223612244.
Dengue Antibody-Dependent
Enhancement: Knowns and
Unknowns

SCOTT B. HALSTEAD1
15
Antibody-dependent enhancement (ADE) is a phenomenon involving infectious
IgG antibody immune complexes that mediate the worsening of diseases involving
a wide spectrum of microbes and vertebrates. ADE is a new type of Gell-Coombs
immunopathology: type I, IgE-mediated immediate hypersensitivity; type II,
antibody-mediated acute immune complex disease; type III, IgG-mediated
complement-dependent foreign antigen immune complex disease; type IV, cell-
mediated immune and autoimmune diseases; and type V, IgG immune complex
enhancement of microbial infection in Fc-receptor (FcR)-bearing cells. Three
of these immunopathologies are mediated by IgG antibodies. Type V immuno-
pathology differs in function from type II and III immunopathologies in that im-
mune complexes are not directly cytotoxic but serve to increase disease severity
by regulating the productivity of intracellular microbial infection. In type II
immunopathologies, IgG antibodies are often directed at autoantigens and include
acute rheumatic fever where microbial antigens mimic antigens in various human
tissues, generating an immune response that breaks down immune tolerance. In
type III immunopathologies foreign antigen-antibody complexes are often trapped
in the basement membranes of endothelial linings. Examples include acute serum
sickness, glomerular nephritis, and postimmunization diseases such as break-
through measles and respiratory syncytial virus infections in vaccine recipients

1
Department of Preventive Medicine and Biometrics, Uniformed Services University of the Health Sciences,
Bethesda, MD 20814.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0022-2014

249
250 HALSTEAD

that result in destructive complement-fixing controls (6, 7, 17). These mechanisms were
virus-IgG immune complexes predominantly in studied in mouse macrophage-like cells when
the lung (1). West Nile immune complexes attached to FcR-
An early report of the ADE phenomenon, in bearing cell surfaces more rapidly compared
vitro, was by Hawkes, who observed a greater with naked virus particles (18, 19). With feline
number of plaques in chick embryo fibroblast infectious peritonitis virus an increased number
monolayers infected with Murray Valley en- of peritoneal macrophages were infected in
cephalitis virus (MVEV) preincubated with vitro in the presence compared with the
high dilutions of chicken antisera than in absence of antibodies (20). It is also possible
virus-only controls (2). In further studies the that immune complexes were internalized more
authors concluded that this phenomenon was rapidly than was naked virus as observed in an
the result of antibodies stabilizing the sponta- HIV-1 model (21).
neous degradation of MVEV (3). A different These mechanical explanations of ADE
explanation emerged when enhanced infection changed radically when human macrophages
of dengue virus (DENV) was observed in were infected in vitro using Ross River virus
cultures of peripheral blood mononuclear (RRV) immune complexes, or in mouse models
cells from dengue-immune compared with in vivo. In humans, acute infections with RRV
nonimmune subhuman primates (4). This often evolve to a postinfection arthritis of many
phenomenon was subsequently attributed to months duration. It was observed that arthri-
enhanced growth of DENV infection in primary tis patients synovial cells stained for RRV
monocytes and macrophages in the presence of antigens and synovial fluids contained inter-
nonneutralizing enhancing dengue antibodies feron- (IFN). In an attempt to model this
(57). It was shown subsequently that MVEV phenomenon, chronic RRV infections were
infection enhancement occurred in functional established in mouse macrophage cell lines
chicken macrophages that comprise 2% of and in primary human monocytes/macro-
chick embryo fibroblast monolayers (8). Be- phages (22). Unexpectedly, the incubation of
cause of the conformational requirement that RRV with diluted RRV antiserum resulted in
Fc receptors and IgG Fc termini be of the enhanced infection in these cells through a
same vertebrate phylogenetic class, ADE in complex intracellular process involving the
chick embryo fibroblasts was observed only suppression of innate cellular immunity by
when MVEV antibodies were raised in immune complexes. This involved a reduction
chickens, not in mammals (9). Because mono- of the production of reactive nitrogen radicals
cytes and macrophages were identified as the via NOS2 and the downregulation of INF- and
principal hosts of in vivo DENV infection, the IFN- production by abolishing IRF-1 and
phenomenon of ADE was suggested as an nuclear factor-KB gene expression. Also,
immunopathologic mechanism (10 16). there was a marked increase in IL-10 gene
Different lines of scientific inquiry over the transcription and protein production (23, 24).
past four decades have sharpened our under- This immune complex suppressive phenome-
standing of microbial antibody-mediated path- non required an infectious agent since the
ogenesis mechanisms in vertebrate hosts. ligation of FcR by zymosan-antibody com-
During initial studies of ADE it was assumed plexes in the presence of RRV did not ablate
that the observed increased growth of virus, antiviral transcription (24). Thus, rather than
which in some cases was 100- to 1,000-fold, was simply involving an increase in the number of
the result of phenomena extrinsic to mononu- infected cells, ADE in RRV is a complex
clear phagocytes such as an increase in rates of intracellular phenomenon involving increased
attachment or internalization of immune com- intracellular production of virus as a result of
plexes to target cells resulting in an increased immune complex suppression of innate cellu-
number of infected cells compared with lar immunity.
CHAPTER 15 Dengue Antibody-Dependent Enhancement 251

These observations were quickly expanded be on iADE in dengue infections as the


to dengue. The DENVs are a group of four mechanism that controls the conversion of a
closely related members of the Flavivirus mild self-limited acute illness to the dengue
genus. DENV-1 through -4 share 60 to 70% vascular permeability syndrome (DVPS). DVPS
genetic homology and are inoculated by the is the underlying pathophysiological mecha-
bite of infected Aedes aegypti. Initial infections nism of dengue hemorrhagic fever/dengue
with any of the four DENVs raise protective shock syndrome (DHF/DSS) as defined by
type-specific antibodies, but the dominant the 1997 WHO Technical Guidelines (Dengue
population of antibodies are cross-reactive Haemorrhagic Fever: Diagnosis, Prevention,
and nonneutralizing. These efficiently en- Treatment and Control. 1997. World Health
hance infection by a different DENV type (5, Organization, Geneva, Switzerland, p. 184).
2529). In vitro studies of FcR-bearing cells ADE as a pathogenesis mechanism should
show that monoclonal dengue antibodies of lead ultimately to a coherent explanation for all
many specificities may form infectious im- the phenomena that comprise DVPS. The basic
mune complexes, the major requirement being physiological disturbances and the critical
attachment to a virion surface antigen at a timing of DVPS during the course of a dengue
subneutralizing antibody concentration (30, illness are described in Table 1. DVPS usually
31). In practice, antibodies directed at surface presents in individuals immune to a single
epitopes that are not involved in virus entry DENV who experience a heterotypic dengue
efficiently produce ADE (32). Indeed, the high infection (46). A small fraction of cases occurs
rate at which infants acquire severe dengue during a second heterotypic (third) DENV
disease during their first dengue infection infection (47). DVPS also occurs during primary
when maternal polyclonal dengue antibodies dengue infections in individuals circulating
have degraded below protective levels is a passively acquired dengue antibodies. In animal
unique illustration of the ADE phenomenon in models the simple passive transfer of dengue
human medicine (10, 3339). Studies of the antibodies sensitizes hosts to DVPS during a
pathogenesis of innate and acquired host subsequent DENV infection. Based on these
immune responses to many acute and chronic observations, it is conceivable that DVPS might
human and animal infectious diseases show occur during dengue infections in individuals
evidence that cross-linking of IgG immune who had previously received blood transfusions
complexes with Fc receptors increases intra- from dengue-immune donors. This phenome-
cellular infection, thus contributing to disease non has not yet been reported. But whatever the
severity by a mechanism labeled intrinsic case, DVPS regularly occurs during primary
ADE-(iADE) (40, 41). iADE distinguishes
intracellular mechanisms from the extrinsic TABLE 1 Dengue vascular permeability syndrome
mechanisms of ADEan increase in infectiv- A dengue syndrome that occurs late in the course (on
ity, infection rate, or the number of infected or near defervescence) of an acute dengue illness
consisting of:
cells by immune complexes compared with
(a) Thrombocytopenia
infection with the microorganism only. Ex- (b) Altered hemostasis: most commonly
trinsic and intrinsic ADE have been measured prolonged bleeding time and/or elevated
as contributing a 3-fold or a 100-fold increase aPTT (activated partial thromboplastin
in virus production, respectively (42). time) and/or elevated prothrombin time
(c) Activated complement, by classical and
The ADE phenomenon has attracted wide
alternative pathways
interest in viral pathogenesis research because (d) Elevated liver transaminase enzymes
many viruses replicate in macrophages in vivo, (e) Vascular permeability. Clinically signicant
and this phenomenon is correlated with en- loss of uid and small macromolecules
hanced disease in many partially immune (e.g., albumin) into interstitial spaces, most
commonly to serosal cavities
vertebrate hosts (4345). Here, the focus will
252 HALSTEAD

dengue infections in infants born to women with increased or decreased incidence of


who previously had acquired multiple dengue DVPS during secondary dengue infections.
infections (33, 34, 48). While in dengue- These have been described in recent re-
endemic countries this phenomenon contrib- views (49). These factors will not be further
utes 5% of all hospitalized cases, in general, it is considered or discussed here.
poorly studied and vastly under-reported.
As in all infectious diseases, dengue infec-
Enhancing Antibodies
tions progress through phases during which
afferent and efferent phenomena predominate. The sensitizing infection
Afferent phenomena are those that improve or Human experimentation has established that
potentiate the infection process and pathoge- there is a DENV-2 infection refractory period
nicity of the infecting microorganism. Efferent of three months following an initial DENV-1
phenomena are those that counter the patho- infection (50). Possibly related to this protec-
genic potential of the infecting microorganism tive phenomenon, sera obtained shortly after a
and that lead to the elimination of the infection. primary DENV infection were found to have
For most infections, researchers studying affer- abundant heterotypic antibodies that formed
ent phenomena focus on microbial offensive and large immune aggregates on the surface of
defensive weapons, but in dengue, enhancing primary human macrophages that were neu-
antibodies and the specific immune complexes tralized in solution but not eliminated via
that are made contribute powerfully to poten- phagocytic clearance (51). After this refractory
tiate infection and disease outcome in dengue. It period, second dengue infections occur, and a
is my contention that DVPS is the result either of portion are expressed as clinical illness. Vary-
pathogenic factors released directly by infected ing amounts of heterotypic neutralizing anti-
tissues and/or as an outcome of normal host bodies are raised following a primary dengue
efferent efforts to control and end infection. In infection. The natural histories of antibody
either of these cases, disease severity is directly responses following infection by DENV-1, -2,
related to the mass of dengue-infected tissues. -3, or -4 including heterotypic antibodies are
Because DVPS in individuals experiencing often unknown.
second dengue infections is essentially identical Infection-enhancing antibodies are an ob-
to that occurring in infants, it is crucial that served risk factor for enhanced dengue disease
pathogenesis hypotheses and research efforts (52), and an enhanced peak viremia (measured
focus on unitary mechanisms of DVPS that before antibody response starts) has been
explain passively as well as actively acquired shown to be an anticipatory correlate of severe
dengue immunity. The known and unknown disease (53, 54). Recent pathology studies of
afferent and efferent factors contributing to human autopsies and tissues from mouse
ADE regulation of dengue disease severity will models have firmly established the central
be identified and discussed below according to role of monocytes, macrophages, and imma-
the outline in Table 2. ture and mature dendritic cells as infected
target cells (1316, 55).
In dengue-endemic countries monotypic
infections with DENV-1, -2, -3, or -4 sensitize
KNOWNS
individuals to disease accompanying a first
heterotypic (second) dengue infection. This
Afferent
phenomenon accounts for around 95% of
Host Genetic Factors Contributing to hospitalized and carefully defined DVPS (47).
Susceptibility to DVPS The clinical outcomes of first heterotypic
Many human genetic factors have been dengue infection are time dependent. First
identified as being significantly associated heterotypic infections occurring at less than a
CHAPTER 15 Dengue Antibody-Dependent Enhancement 253

TABLE 2 Context for understanding known and studying unknown factors that contribute to the
pathogenesis of the dengue vascular permeability syndrome via ADE
Knowns
1. Afferent:
a. Host genetic factors enhancing susceptibility to DVPS
b Enhancing antibodies
i. Sensitizing infections
ii. Attributes of iADE
iii. Role of infection sequence
iv. Role of different myeloid cells
v. Role of different FcRs in iADE
c. Role of DENV in ADE
i. Enhanced growth of DENV-2 in primary human monocytes
ii. Differences in ability of DENV-2 strains to be neutralized by DENV-1 antibodies
iii. DENV-2 genetic differences associated with rapid enhancement of the severity of DENV-2
infection 20 years after infection with DENV-1
iv. Genetic differences in DENV-2 isolates recovered from disease of enhanced severity 4 years after
DENV-1 and 8 years after DENV-3
2. Efferent:
Overview

Unknowns
1. Afferent:
a. Host genetic factors enhancing susceptibility to DVPS
b. Enhancing antibodies
i. Sensitizing infections
ii. Attributes of iADE
iii. Role of infection sequence
iv. Role of different myeloid cells
v. Role of different FcRs in iADE
c. Role of DENV in ADE
i. Enhanced growth of DENV-2 in primary human monocytes
ii. Differences in ability of DENV-2 strains to be neutralized by DENV-1 antibodies
iii. DENV-2 genetic differences associated with rapid enhancement of the severity of DENV-2
infection 20 years after infection with DENV-1
iv. Genetic differences in DENV-2 isolates recovered from disease of enhanced severity 4 years after
DENV-1 and 8 years after DENV-3
2. Efferent:
Overview

two-year interval are partially protected, first heterotypic DENV-2 infections either 4
resulting in inapparent infections or mild or 20 years after a DENV-1 infection.
disease (5659). After that time, DVPS has In 1977 to 1979, around 45% of the popula-
been observed in 2 to 4% of all second dengue tion of Cuba (at that time, age groups <1 to 40
infections combined (for a discussion of the were dengue nave) was infected with DENV-1,
pathogenicity of different sequences of hetero- a virus introduced into Cuba only once. DENV-
typic dengue infection, see below) (30). The 2 circulated on the island in 1981, 4 years after
frequency and severity of DVPS increases as DENV-1, and DENV-2, at an interval of 20
the interval between the first and second years. Hospitalization rates per 10,000 second-
infections lengthens. This phenomenon was ary DENV-2 infections at the 20-year interval
observed when hospitalization and case fatality were 8.2 times higher than at those at a 4-year
rates were compared among individuals who interval. The case fatality rate was 4.7 times
were 15 to 39 years old when they experienced higher when second heterotypic DENV-2
254 HALSTEAD

infections occurred at a 20-year compared nized with killed JE vaccine developed mild
with a 4-year interval (60). This observation overt dengue illness at a higher frequency than
was subsequently correlated with a sustained did flavivirus-susceptible patients infected
decline in heterotypic DENV-2 neutralizing with the same DENV (64). Dengue infections
antibody titers over a period of 20 years in JE-immune patients not only increased the
following DENV-1 infections in 1977 to 1979 rate of mild overt dengue disease, but the
(61). Heterotypic antibodies have been ob- ensuing illness lasted 2 times longer than
served to be important in the modulation of dengue illnesses in nonimmune patients. This
second DENV infections in cultures of primary establishes the ability of JE antibodies to
human monocytes (52). Low dilutions of mildly enhance human dengue infections.
preinfection sera from children who had The in vitro correlates of this observation
inapparent secondary DENV-2 infections al- have not been described. Prior to this obser-
most invariably reduced or neutralized DENV- vation, based upon data trends in a large
2 infections in primary human monocytes, vaccine study, it was thought that JE vaccina-
while preinfection sera from children who tion reduced the severity of DHF accompa-
developed an illness requiring hospitalization nying a second dengue infection (65).
had little or no detectable heterotypic neutral- A large amount of literature has emerged
izing antibodies (52). This observation suggests on the role of nonneutralizing antibodies
that low levels of heterotypic neutralizing in enhancing DENV infections. It has been
antibodies (most were anti-DENV-1) did not observed that a considerable fraction of all
prevent DENV-2 infection but downregulated antibodies circulating after a first heterotypic
DENV-2 clinical responses. In this study, dengue infection are directed at prM (32, 66).
around one-fifth of monotypically dengue- prM antibodies target immature or partially
immune school children lacked heterotypic immature DENV via the pr peptide. A signif-
DENV-2 antibodies measured in primary icant fraction of all DENVs produced in vitro
human monocyte cultures and developed are immature or partially immature, as are an
DHF when infected by DENV-2. This is unknown fraction of DENVs circulating dur-
virtually the same ratio as for the occurrence ing natural human infections (67). Immature
of shock during sequential DENV-1 then DENVs cannot be processed in the endosome
DENV-2 in Rayong, Thailand (62). Prior to result in infection. prM antibodies were
infection with two different DENVs (second found to be highly cross-reactive with all
heterotypic dengue infection) also sensitizes to DENV serotypes and generally to have poor
DVPS, but at a frequency about 50-fold lower neutralizing capacity (32, 68). Recent studies
than a single DENV infection (47). showed that prM antibodies enhance the
In the in vitro ADE literature antibodies to infectivity of noninfectious immature DENV
many flaviviruses enhance DENV infection in particles (32, 69, 70). Monoclonal DENV
FcR-bearing cells (63). Should this be true for antibodies directed at several E domains
in vivo infections, opportunities abound for have been found to interact with immature
flavivirus group virus infections to sensitize to particles, resulting in infection enhancement
enhanced dengue infections. In Southeast Asia in FcR-bearing cells (71, 72).
the DENVs cocirculate with Japanese enceph-
alitis (JE), in India with JE and West Nile, in The attributes of iADE
Pakistan with West Nile, in Australia with the This is a remarkably complex phenomenon,
Kunjin strain of West Nile, and in most of the triggered by attachment of IgG-microbial
American region with yellow fever or yellow pathogen complexes to FcRs of many classes,
fever 17D vaccine. Importantly, in Thailand it resulting in messages being passaged and
was observed that children with previous expressed via the cytoplasmic tail. Much of
wild-type JE infections or who were immu- what we know about FcR signaling comes from
CHAPTER 15 Dengue Antibody-Dependent Enhancement 255

other pathogensLeishmania amastigotes, for same antigen was complexed with IgG anti-
instance. The importance of macrophage re- ovalbumin, T cell responses were driven to
ceptors in the generation of cytokines in Th2 and produced IL-4. This Th2-like pheno-
Leishmania-infected macrophages was recog- type was stable and was retained when the
nized when interleukin (IL)-12 production in T cells were subsequently restimulated under
BALB/c mouse bone marrow macrophages in nonbiasing conditions. Mice vaccinated with
response to lipopolysaccharide was sup- IgG-opsonized ovalbumin made high levels of
pressed after ligation of FcR, complement, or IgG Ab of the IgG1 isotype. The T cell biasing
scavenger receptors (73). Both mRNA synthe- and its reversal via FcR ligation was also
sis and protein secretion were diminished to observed in vivo. Using macrophages from
near undetectable levels following receptor gene knockout mice, the production of IFN
ligation. Suppression was specific to IL-12 and IL-4 by T cells was shown to be controlled
since TNF production was not inhibited. by the macrophage cytokines IL-12 and IL-10,
Also, the ligation of mouse FcR with immune respectively. These and other studies demon-
complexes was shown to enhance the produc- strate that the ligation of FcR on activated
tion of IL-10 (74). Stimulation of mouse bone macrophages reverses the Th1 biasing that
marrow macrophages by lipopolysaccharide accompanies innate immune responses to
resulted in some IL-10 production, but the microbial products.
addition of red blood cell (RBC) opsonized How do antibody-coated amastigotes result
with IgG antibodies dramatically enhanced IL- in the production of IL-10 by macrophages?
10 production. Immune complexes not only Ligation of macrophage FcR produces a rapid
induce activated macrophages to produce IL- and enhanced activation of two mitogen-
10, but they also induce both macrophages and activated protein kinases: ERK and p38 (82).
dendritic cells to switch off their production of The activation of ERK leads to the phospho-
IL-12 (73, 75). The IL-10 induction by IgG- rylation of serine 10 on histone H3 at the il-10
amastigotes did not occur in macrophages gene, making the promoter more accessible
from mice lacking the common gamma chain to transcription factors generated in response
that signals through FcR 1, 3, and 4, indicating to p38 activation (83). Activation of both
that one or all of these three receptors were mitogen-activated protein kinases was re-
involved. Subsequent studies using defined quired for IL-10 synthesis. In addition to
immune complexes demonstrated that all ERK activation, an inflammatory stimulus,
three of the FcRs that signal through gamma such as low-molecular-weight hyaluronic
were capable of signaling for IL-10 production acid from the extracellular matrix, must also
in macrophages (76). The implication from be present. The combination of these two
these studies is that in some settings, IgG itself signals resulted in the superinduction of IL-
biases the immune response toward a Th2- 10 (84). Macrophages lacking FcR, or macro-
type response. Indeed, for some species of phages treated with an inhibitor of spleen
Leishmania, chronicity of infection requires tyrosine kinase that is activated following
that amastigotes be coated with IgG (77). This FcR ligation, failed to activate ERK and
phenomenon is now very well established consequently failed to produce IL-10 following
(7880). infection with Leishmania amastigotes.
That IL-10 induction by ligation of FcR During in vitro ADE DENV-2 infection in
was a generic process was demonstrated with the THP-1 cell model (human monocytic Fc
a nonmicrobial antigen (81). Lipopolysaccha- receptor-bearing continuous cell line) intra-
ride-treated BALB/c mouse macrophages cellular DENV production was increased as a
when exposed to ovalbumin alone developed result of idiosyncratic Fc-receptor signaling
T cell responses driven to Th-1 and character- (85). When immune complexes ligate FcRI
ized by the production of IFN. When the and FcRIIA, at least two of the following
256 HALSTEAD

types of suppression pathways are expressed: peripheral blood mononuclear cells collected
DAK, Atg5-Atg12, SARM, and TANK plus the during the acute phase from children with
positive Th2 cytokine regulator, IL10. Collec- dengue fever (DF) or DHF were compared
tively, these phenomena downregulate using microarray analysis (88). Patients with
antiviral responses in ADE-infected target DHF had decreased levels of NO, reduced IFN
cells. DAK and Atg5-Atg12 of RIG-I/MDA5 transcript in peripheral blood mononuclear
abolish expression of RIG-I/MDA5 and weak- cells, and increased IL-10 blood levels com-
en the RIG-I/MDA5 signaling pathway as pared with patients with milder illness. IFN
monitored through levels of downstream gene upregulation and IFN- production were
signaling molecules: IPS-1, IKKi, TRAF-3, significantly elevated in patients with mild
TBK-I, etc. One outcome is decreased produc- compared with severe dengue illness. In other
tion of type-I IFN as well as IFN-activated studies, during the acute stage of severe
antiviral molecules (86). Activation of SARM disease increased production of IL-10 and
and TANK results in expression blockage of downregulation of multiple IFN regulatory
Toll-like receptors (TLR) 3, 4, and 7 (87). This genes were noted (8991). The protective role
inhibits MyD88-dependent and MyD88-inde- of IFN in moderating dengue infection has
pendent signaling pathways, resulting in an- been demonstrated in a mouse model and
other route for type I IFN suppression. As a suggested for humans with DF (9294). The
result of at least these two suppression precise role of immune-complex-elicited IL-
pathways, ADE-infected THP-1 cells secreted 10 production on the clinical evolution of
reduced levels of type I IFN and at the same severe dengue infections is not well under-
time suppressed the transcription and trans- stood but may be responsible for the observed
lation of IL-12, IFN-, and TNF-, facilitating Th1 to Th2 shift in DHF (95).
the expression and synthesis of the anti- Pathologic studies of human tissues have
inflammatory cytokines. ADE infection also established monocytes, macrophages, and im-
suppressed the innate anti-DENV mediator, mature and mature dendritic cells as signifi-
nitric oxide radicals, by disrupting the tran- cant targets for DENV infection (1316, 96, 97).
scription of the inducible nitric oxide synthase In humans, secondary dengue infections fol-
(iNOS) gene transcription factor, IRF-1(85). low a stereotypical course with severe out-
This suppressive mode is believed to be comes, shock or gastrointestinal hemorrhage,
mediated by IL-10 activity. IL-10 is synthe- accompanying vascular collapse that results
sized at an early phase of ADE infection in from capillary permeability occurring around
THP-1 cells. In this experimental setting, IL-10 the time of defervescence (98). Indirect evi-
not only induces Th2 biasing but operates via dence suggests that cytokines mediate dengue
the suppressors of cytokine signalling (SOCS) vascular permeability. Much work has been
system to suppress the JAK/STAT signaling directed at the measurement of cytokine blood
pathway, resulting in suppression of iNOS levels in patients late in the acute phase, just
gene expression and reduction of nitric oxide prior to onset of shock (99, 100). High levels of
radical production. The viral enhancement viremia early in the disease and high levels of
effect of IL-10 is abolished with small inter- pro-inflammatory and immunomodulatory cy-
fering RNA specific to the IL-10 gene (86). It tokines including IL-10 late in the disease are
can be concluded that in vitro, iADE infection associated with severe outcomes (101).
not only facilitates viral entry but also modifies However, during ADE-infection of primary
innate and adaptive intracellular antiviral monocytes, IL-10 synthesis peaked at the same
mechanisms, resulting in enhanced DENV serum dilution that produced peak virus yield
replication. (42). In addition, point mutations at the IL-10
Critically, the same responses are observed promoter, positions -1082 A/G, -819 C/T, and
in vivo. Genome-wide transcriptomes from -592 C/A, result in polymorphism that
CHAPTER 15 Dengue Antibody-Dependent Enhancement 257

differentiates monocytes into high, intermedi- Human monocytes, activated macrophages,


ate, and low IL-10 producers. How these and mature dendritic cells supported ADE,
phenotypes correlate with disease severity while immature dendritic cells did not. Infec-
requires more investigation. tion of macrophages by DENV-2 alone or as
fully neutralized immune complexes stimulat-
Role of infection sequence ed high levels of and IFN, and these were
Second DENV infections can occur in 12 downmodulated under ADE conditions and
combinations, at least 10 of which have been replaced by secretion of IL-6 and TNF. Type I
documented to result in hospitalized disease IFNs were not produced or suppressed by
(47). The sequence of infection may be highly iADE infection of monocytes with DENV-2
determinative of disease severity. Secondary (42, 107). However, during ADE infection of
DENV-2 infections resulted in shock syn- primary monocytes, IL-10 synthesis peaked at
drome patients, while secondary DENV-1, -3, the same serum dilution that produced peak
and -4 did not in the 1980 cohort study in virus yield (42). In addition, point mutations at
Rayong, Thailand (62). The specific infection the IL-10 promoter, positions -1082 A/G, -819
sequences associated with DSS cases were C/T, and -592 C/A, result in polymorphisms
known from virus isolations in acute phase that differentiate monocytes into high, inter-
sera and antibodies in pre-illness sera or by mediate, and low IL-10 producers.
applying the original antigenic sin phenome- It should be noted that FcR-bearing con-
non to paired sera (102). Although secondary tinuous cell lines that are incapable of produc-
DENV-1 infections were most common that ing interferon have been used to detect
year, DSS occurred only during secondary extrinsic ADE but do not detect or produce
DENV-2 infections. Burmese workers came iADE. A prime example is K562 cells, widely
to a similar conclusion in their 1984 to 1988 used to measure enhancing properties of
longitudinal seroepidemiological study in dengue antibodies (108). Published research
Yangon, Myanmar (103). By contrast, in an results using K-562 cells are often at odds with
Indonesian study DSS was associated with results obtained using primary human myeloid
sequences ending in DENV-1, -3, and -4, but cellsthe one group measuring extrinsic ADE
not DENV-2, even though secondary DENV-2 and the other iADE (52, 109).
infections occurred (104). DENV-3 was asso-
ciated with an outbreak of DHF/DSS in Tahiti Role of different FcgRs in iADE
in a population that had prior infection When the ability of DENV immune complexes
experience with DENV-1 and DENV-2 (105). to be internalized following interactions with
Also in Tahiti, DENV-1 circulating in 2001 was human FcRIA and FcRIIA was studied in a
enhanced to produce severe clinical disease by model system, the FcRIIA mediated both
antibodies to DENV-2 that had circulated 4 to iADE and immune phagocytosis, while FcRIA
5 years earlier (106). mediated uptake of immune complexes via
phagocytosis (110). Genes for human FcRIIA
Role of different myeloid cells have been transfected into continuous monkey
Much early published work on dengue and kidney cells, transforming them into cells
Ross River virus (RRV) used primary human capable of detecting and expressing the capac-
mononuclear phagocytes to study iADE. To ity of DENV immune complexes to be neu-
date, virtually all research has been carried out tralized or to enhance infections. These
using DENV-2. When dengue iADE was tested systems, employing Fc RIIA have been used
in four different primary human myeloid cells to assay in vivo viremia and to measure the
derived from the same peripheral blood leu- protective versus enhancing properties of sera
kocyte (PBL) donors, viral infection and containing mixtures of DENV and antibodies
cytokine responses differed significantly (42). (111115).
258 HALSTEAD

In the Leishmania mouse model, it was tion properties in human mononuclear


observed that mouse IgG1and IgG2a/c induce phagocytes, might contribute to enhanced
IL-10 from mouse macrophages in vitro equal- DENV infection and to the severity of the
ly well but through different FcR subtypes: disease.
IgG1 through FcRIII, and IgG2a/c primarily
through FcRI but also through FcRIII. In Differences in ability of DENV-2 strains
sharp contrast, mice lacking IgG1 develop to be neutralized by DENV-1 antibodies
earlier and stronger IgG2a/c, IgG3, and IgM Differences in genetic and viral structure
responses to Leishmania mexicana infection among DENV-2 viruses that are associated
and yet are more resistant to the infection with disease outcome during a first heterotyp-
(116). Thus, IgG1, but not IgG2a/c or IgG3, is ic dengue infection are well established, and
pathogenic in vivo, in agreement with prior the impact of these differences has been well
studies indicating that FcRIII is required for studied (118). The actual sites on the virion that
chronic disease. This calls into question the mediate these differences in human disease
assumption that mouse macrophages, which expression are still unknown. The first DENV
should secrete IL-10 in response to both IgG1 to be recovered in the northern hemisphere
and IgG2a/c immune complexes, are the most was DENV-2 TR 1751 (119). Dengue outbreaks
important source of IL-10 generated by IgG- prior to World War II have been attributed to
FcR engagement in L. mexicana infection. DENV-2 by serological studies in Panama and
Cuba (120, 121). In 1963, DENV-3 was intro-
duced into the northern hemisphere, first
Role of DENVs
recognized in Puerto Rico (122). The geo-
Enhanced growth of DENV-2 in primary graphic extent and intensity of transmission of
human monocytes these viruses was never measured, but condi-
A single study suggests that DENVs isolated tions for sequential infection existed and no
from patients with differing degrees of disease DHF/DSS outbreaks were reported. In 1977,
severity may themselves be biologically differ- DENV-1 was introduced into the Caribbean
ent. The biological behavior of wild-type and quickly spread throughout the region
DENV-2 isolated from children with mild (153). Again, sequential infectionsDENV-2
secondary dengue infections was compared then DENV-1 or DENV-3, then DENV-1
with DENV-2s from children with grade I-III were possible, but there are no reports of
dengue hemorrhagic fever. All isolates were DHF/DSS.
made in C6-36 cells from children presenting However, when an Asian DENV-2 was
to Childrens Hospital during the 1980 DENV- introduced into Cuba in 1981 following the
2 outbreak in Bangkok, Thailand (117). DENV- 1977 to 1979 virgin soil introduction of DENV-1,
2 that had been passaged in C6-36 cells only a major DHF/DSS epidemic ensued (123, 124).
once or twice from mild illnesses replicated to But similar sequential introductions of the
lower titers in cultures of human primary American genotype DENV-2 in 1995, five
monocytes either with or without enhancing years after introduction of DENV-1 in 1990,
concentrations of polyclonal dengue anti- failed to produce any DHF/DSS at all (125).
bodies than did DENV-2 strains isolated Fortunately, this event occurred in the Ama-
from DHF patients. The small number of zonian city of Iquitos, population 344,686,
subjects with mild illness jeopardizes the where an ongoing longitudinal serological
statistical significance of these observations. cohort permitted reconstruction of past events.
Nonetheless, these preliminary observations It was estimated that 49,000 secondary
suggest the possibility that viral factors, DENV-2 infections occurred in 1995 and that
whether surface antigens, attachment sites these should have produced about 10,000 cases
for entry into leukocytes, or intrinsic replica- of DHF/DSS. Careful study of hospital records
CHAPTER 15 Dengue Antibody-Dependent Enhancement 259

found no DHF/DSS-like disease. In fact, (129). During a 2001 to 2002 Havana outbreak
secondary infections were accompanied by a similar rapid increase in disease pathogenic-
mild disease at attack rates far below observed ity was observed in individuals who experi-
dengue infections. enced a DENV-3 infection 24 years after
Full-length sequences of the Asian and infection with DENV-1 (130132). Initially, it
American DENV-2 genomes from viremic was hypothesized that this phenomenon
sera revealed a total of six encoded amino might be caused by the emergence of neutral-
acid charge differences in the prM, E, NS4b, ization escape mutants, as DENV-2 was seri-
and NS5 genes along with structural changes ally passaged in individuals circulating DENV-
in the 5 and 3 nontranslated regions (126). 1 antibodies (129). As discussed above, DENV-1
Stored Peruvian anti-DENV-1 human sera infections produce heterotypic antibodies that
from 1990 were found to highly neutralize partially neutralize DENV-2. These might be
American genotype DENV-2 strains but not to expected to generate neutralization escape
neutralize Asian genotype DENV-2 strains mutants that are no longer neutralized and
(127). This suggests the existence of American that could produce enhanced infections and
genotype DENV-2 strains of envelope struc- disease. It was suggested that the proportion of
ture(s) analogous to structure(s) on DENV-1 individuals infected with escape mutants
strains. The loss or modification of this might increase with time, producing the
structure on Asian genotype DENV-2 strains observed month-to-month increases in path-
suggests that American DENV-2 strains are ogenicity.
more closely related to an ancestral DENV-2, In January 1997, the sensitive Cuban sur-
while the Asian genotype DENV-2 strains have veillance system detected dengue cases in
emerged more recently, possibly due to posi- Santiago de Cuba just two months after illness
tive selective pressure exerted by ADE (128). in the index case (identified retrospectively).
Most people who read the dengue literature Acute phase sera were sent to the Havana
must assume, based upon citations, that Amer- laboratory for serological testing and virus
ican genotype DENV-2 strains are intrinsically isolation. The DENV-2 strains that circulated
incapable of producing severe dengue disease. in 1981 and 1997 in Cuba both belonged to the
That any DENV possesses the intrinsic prop- American/Asian genotype circulating in the
erty of producing an enhanced dengue disease Americas since 1981. Twenty-nine DENV-2
is simply wrong. It is when viral attributes isolates were obtained during the early (low
come into play at any stage of a heterotypic pathogenicity) and the late (high pathogenic-
infection that virulence is an outcome. In ity) stages of the 1997 epidemic. The 1997
Iquitos, it appears that American genotype Cuban DENV-2 strain amino acid alignment
DENV-2 infections were down-modulated showed a substitution methionine/threonine
rather than enhanced in those individ- at position E340 specific for viruses isolated
uals circulating highly cross-reactive DENV-1 from Cuba, Venezuela, and Martinique (133).
antibodies. This nonconserved substitution is located in
an antigenic region containing multiple T- and
DENV-2 genetic differences associated B-cell epitopes. Another nonconservative
with rapid enhancement of the severity of change leucine/glutamine at E 131 was ob-
DENV-2 infection 20 years after infection served in Jamaica/83 and Cuba/97 isolates.
with DENV-1 Remarkably, only two nonconserved substitu-
A dramatic increase, month to month, in case tions in the E gene were found between the
fatality rates and the severity of dengue disease Jamaica 1983 and the 1997 Cuban strains,
was observed in humans of all ages who, in indicating that there has been very little in
1981 and 1997, experienced a DENV-2 infec- situ evolution of DENV-2 following its intro-
tion 4 and 20 years after a DENV-1 infection duction from Asia. Of interest, the Cuban
260 HALSTEAD

DENV-2 isolates maintain the presence of N at samples (130). Of 23 DENV-2 strains obtained
position 390 of Asian DENV-2 strains, a site during the course of the epidemic, five viruses
predicted to be a determinant of the American from the first part of the epidemic had thr at
genotype DENV-2 that causes only mild position 164, while the isolates on and after
disease (126). As the E gene sequences from 25 February had ser at this position (130, 136).
these isolates were found to be conserved over By July, mosquito control efforts were well
the period of the outbreak, the hypothesis that established and cases began to decline, with
envelope gene escape mutants contributed to a marked reduction in cases in August. The
the observed increased disease severity is switch from threonine to serine at NS1
negated (133). position 164 appears to have been fixed within
The complete genes of six of the 1997 a few months of the virus having been intro-
DENV-2 isolates were sequenced. A strong duced into Cuba.
conservation of structural genes and proteins
and of the noncoding regions was again Genetic differences in DENV-2 isolates
noted. However, nucleotide substitutions recovered from disease of enhanced
were observed in NS genes, notably in NS1 severity 4 years after DENV-1 and 8 years
and NS5 (134). Most synonymous mutations after DENV-3
generally correlated with the time of sam- Similar dengue disease phenomena were ob-
pling. It was possible to divide the isolates served among pediatric patients in Managua,
into two groups based upon five substitutions Nicaragua, and adults in Taiwan (137, 138).
those from the beginning of the 1997 Further information has been published (136).
epidemic (January to February) and from In Nicaragua, a prospective seroepidemi-
the latter parts of the epidemic (June to July). ological cohort study made possible a complex
One of these five nucleotide substitutions dissection of the contributing host and viral
produced a functionally significant amino factors involved (138). In the dengue season of
acid replacement, threonine (thr) to serine 20052006, 34 patients with DENV-2 infec-
(ser), at residue 164 in the NS1 protein. An tions were admitted to Hospital Infantil
alignment of this NS1 protein sequence in Manuel de Jesus Rivera in Managua, 10 with
GenBank revealed that amino acids in this the diagnosis of DHF/DSS (30%). During
region are highly conserved. DENV-2 strains 20062007 and 20082009, the severity of
usually have Thr at this position, with the DENV-2 disease increased dramatically (85%
exception of the Thailand strain PUO-280 of of cases were secondary infections). Of 102
the Asian I genotype, which has Ser, like the children with DENV-2 infections admitted to
latest Cuban isolates (135). Strains of DENV-2 the hospital during these nonconsecutive trans-
may differ significantly in the structure of mission seasons 64% had DHF/DSS, an unpar-
genomic viral RNA 3 nontranslated region alleled increase in this hospitals 20-year
(NTR), and these structural differences have experience with dengue admissions. Differing
also been correlated with mild disease (126). from the situation in Cuba, where DENV-2
However, the DENV strains from the 1997 introductions in 1981 and 1997 were rapidly
epidemic had no difference in their 3 NTR or controlled by effective vector control, dengue
5 NTR compared with other Asian/American transmission in Nicaragua was continuous.
strains. In Cuba, introductions of DENV-2 in 1981
In further work, nucleotides 379 to 601 and 1997 and DENV-3 in 2001 were preceded
were sequenced from 15 viruses from different by DENV-1 infections in 1977-1978, whereas in
periods of the outbreak. Data from six DENV- Nicaragua DENV-3 had circulated in Managua
2 strains sequenced previously were included in 1994 to 1998, DENV-2 had circulated in 1999
(133). Two additional complete DENV-2 to 2002, and DENV-1 in 2002 to 2005, with
strains were sequenced directly from serum DENV-2 again in 2005 to 2009. During this
CHAPTER 15 Dengue Antibody-Dependent Enhancement 261

latter period a large number of DENV-2 and cough may be followed after 2 to 5 days by
strains were collected from children with rapid deterioration and physical collapse (139).
dengue diseases of all degrees of severity. In Thailand, the median day of admission to the
Over 200 of these viruses were subjected to hospital after the onset of fever is day 4. In this
full-length or partial sequencing, and as had second phase, the patient usually has cold and
been observed in Cuba, a stable clade shift was clammy extremities, a warm trunk, a flushed
noted: NI-1 to NI-2B in 20082009. This face, and diaphoresis. Patients are restless and
replacement event was associated with nine irritable and complain of midepigastric pain.
nonsynonymous amino acid mutations (R97K Frequently, scattered petechiae appear on the
in capsid [C], K94R in nonstructural protein 1 forehead and extremities, spontaneous ecchy-
[NS1], and P245T in nonstructural protein 3 moses may develop, and easy bruisability and
[NS3], N245S in NS4B, M492V in envelope bleeding at sites of venipuncture are common
[E], L279F in NS1, and K200Q, T290I, and findings. Circumoral and peripheral cyanosis
R401K in nonstructural protein 5 [NS5]) and may occur. Respirations are rapid and often
four mutations in the viral NTR. Clade NI-2B labored. The pulse is weak, rapid, and thready,
peak viremia titers were higher than viremias and the heart sounds are faint. The pulse
with clade NI-1 viruses in children with the pressure frequently is narrow (<20 mm Hg);
diagnosis of dengue fever. By analyzing the age systolic and diastolic pressure may be low or
distribution of cases, it was concluded that NI- unobtainable. The liver may become palpable
1 DENV-2 produced disease with an apparent two or three finger breadths below the costal
increase in pathogenicity in children who margin and usually is firm and nontender.
were immune to DENV-1 because there was Early sonograms show thickening of the
cross-protection when infections occurred at gallbladder duct and after initiation of intrave-
short intervals, while more severe disease was nous resuscitation may show thickening of the
observed at longer intervals. By contrast, clade gall bladder duct, perivesicular edema, ascites,
NI-2B DENV-2 infections produced disease of and pleural effusions. At this same time chest
increased pathogenicity in children of all ages radiographs may show unilateral (right) or
who had been infected by DENV-3 during bilateral pleural effusions. Approximately 2 to
1994 to 1998. The authors noted that clade 5% of patients have gross ecchymosis or
NI-2B DENV-2 strains grew to higher titers gastrointestinal bleeding. After a 24- or 36-
than clade NI-1 viruses in C6/36, primary hour period of crisis, convalescence is fairly
human dendritic cells, U937 DC-SIGN, and rapid in children who recover. The tempera-
K-562 cells, but ADE was not studied using ture may return to normal before or during the
DENV-3 antibodies. This group made no stage of shock.
attempt to grow clade NI-1 or NI-2B DENV- There is evidence of an important human
2 strains in the presence of human DENV-1 or dengue resistance gene. Epidemiologic studies
DENV-3 antibodies in vitro in primary human of the 1981 Cuban outbreak demonstrated a
monocytes or macrophages to model ADE higher risk for DHF/DSS in white than in black
infections. individuals (121). A search for DHF/DSS in
black children in Haiti revealed no cases,
despite the presence of high dengue type 1, 2,
Efferent
and 4 infection rates and circulation of the
The incubation period of disease ending in Southeast Asian genotype dengue 2 viruses
DVPS is unknown but is presumed to be the (140). Several HLA antigens have shown dif-
same as that of dengue fever. In children, fering frequencies in DHF/DSS cases and
progression of the illness is characteristic. A controls (141). Early in the acute vascular
relatively mild first phase with an abrupt onset permeability stage of secondary DENV infec-
of fever, malaise, vomiting, headache, anorexia, tion, rapid activation of the complement system
262 HALSTEAD

occurs (142, 143). During shock, blood levels of hypothesis that a competent immune elimina-
C1q, C3, C4, C5, C6, C7, C8, and C3 proactivator tion system is available to generate the
are depressed, and C3 catabolic rates are cytokines that produce DHF/DSS (147149).
elevated. The blood clotting and fibrinolytic It is widely held that the process of
systems are activated (139, 144). As yet, neither eliminating DENV-infected cells generates a
the mediator of vascular permeability nor the cascade of chemokines and cytokines that
complete mechanism of altered hemostasis has contribute to the pathophysiology of dengue
been identified unequivocally. The kinin system disease syndromes. This has been termed a
apparently is not involved. Studies show that perfect cytokine storm (150). Given the
levels of tumor necrosis factor, interleukin-2, evidence of increased infected cell mass in
and interferon- are elevated at the time of severe dengue infections, the cytokines gener-
vascular permeability (144). The strongest ated by interactions between virus-infected
correlates to vascular permeability are inverse cells and the host immune response would
levels of platelet counts and reductions in appear to be quantitatively proportional to
accelerated partial thromboplastin time (145). viral load and not exaggerated or abnormal.
Capillary damage allows fluid, electrolytes, Since dengue is not a cytophilic virus, cellular
protein, and, in some instances, red blood cells infection proceeds until the infected cell is
to leak into intravascular spaces. This internal eliminated. A wide misconception is that peak
redistribution of fluid, together with deficits viremia that occurs early in infection repre-
caused by fasting, thirsting, and vomiting, sents peak viral load. In fact, the quantity of
results in hemoconcentration, hypovolemia, virus in the blood during the course of
increased cardiac work, tissue hypoxia, meta- infection only describes the kinetics of extra-
bolic acidosis, and hyponatremia. A mild degree cellular virus clearance. As discussed above, it
of disseminated intravascular coagulation, plus is likely that elimination of DENV-infected
liver damage and thrombocytopenia, could cells continues well after onset of antibody
contribute additively to produce hemorrhage. production, resulting in peak cellular infection
In serial blood samples taken early in the (viral load) at around the time of deferves-
illness of individuals experiencing secondary cence, and cellular infection is not eliminated
dengue infections it is possible to identify peak until well after the end of viremia. Of interest,
viremia or dengue NS1 concentrations that can a major effort failed to detect circulating CD8+
be measured. These were used successfully to T cells during the late acute illness phase
observe enhanced dengue infections and to of DHF patients (151). This suggested to the
predict subsequent disease severity (53, 54, authors that CD8+ T cells might be located
146). It is critical that viremia be measured in only in the tissues where DENV replication
the absence of antibodiesa very difficult had occurred. Alternatively, as in a Japanese
task. encephalitis model, the antibody component of
Because of the delay of onset of serious the adaptive immune response may play a much
vascular permeability to around deferves- more important role in terminating dengue
cence, there is a widespread hypothesis that infections than previously thought (152).
vascular permeability is somehow related to T
cell activityspecifically, the result of den-
gue-infected cells being attacked by activated UNKNOWNS
T lymphocytes. Cytokine production should
be quantitatively related to the number of The elements of the preceding section are
infected target cells. The reduced risk for briefly reprised here. Ideally, these are ac-
DHF/DSS in protein-calorie malnourished companied by the identification of research
children and the increased risk for DHF/DSS questions and provision of detailed research
in girls versus boys are consistent with the protocols. The state of dengue research on the
CHAPTER 15 Dengue Antibody-Dependent Enhancement 263

intracellular ADE is very primitive. The best I fections. The failure of anyone in the past
can do is to suggest a research approach. In four decades to seriously study this very
almost all instances the approach involves the important and biologically unique phe-
direct investigation of primary human myeloid nomenon is simply mind-boggling. I am
cells with infectious DENV immune com- sorry to say I do not regard the published
plexes. It is crucial that researchers use cells studies of Libraty and Simmons as having
from flavivirus-nave donors. Long ago, it was asked serious research questions (35, 36).
shown that dengue-immune individuals cir- Kliks led the way (34). Why not use
culate mononuclear phagocytes that act as if a maternal serum as a surrogate of cord
dengue antibody was strongly attached (6). blood for each individual infant and test it
Thus, when DENV is added, the iADE and along with the virus isolated? This unique
extrinsic ADE phenomena ensue. Many of the clinical phenomenon not only illustrates
most complex problems in iADE can only be the role played by dengue antibodies in
approached by investigating primary human modulating disease expression but, also,
myeloid cells. the bifurcated role of dengue antibodies
protection for several months after birth at
high concentrations and enhancing infec-
Afferent
tions some months later at subneutralizing
Host Genetic Factors Enhancing concentrations.
Susceptibility to DVPS The contribution of immature DENV par-
Human genetic factors promoting dengue ticles to iADE is of interest. Immature DEN
disease severity are not discussed in this virions are not infectious for human mye-
article. The contribution of human genetic loid cells, but in the presence of enhancing
variables will be better understood when dengue antibodies, ADE infection occurs
they can be placed into a rational under- readily (32, 69). It has been surmised that
standing of how extrinsic phenomena and immature DENVs are released into circu-
normal human biology control ADE. lation during human infections, as anti-
bodies to prM (immature DENV antigen)
Enhancing Antibodies frequently are observed (32).
Sensitizing infections
As described above, the initial single dengue Attributes of iADE
infection is known as the "sensitizing infec- The precise mechanisms fueling enhanced
tion." The immune response to this infection production of DENV in monocytes and
participates in ADE accompanying infection macrophages require further study, as neither
with a heterotypic dengue virus. the production of IL-10 nor suppression of
Studies of iADE have almost exclusively type I interferon are critical to the iADE
focused on DENV-2. A high priority is to phenomenon.
extend studies as quickly as possible to the
complete range of DENV. ADE should be Role of infection sequence
tested using type-specific polyclonal There are at least 12 sequences of infection
human antibodies and not mouse or even that make 12 unique immune complexes. We
human monoclonal antibodies. ADE is already have preliminary data that suggest that
complex. The test systems should retain the outcome of infection differs according to
essential complexity. the specific immune complexes (e.g., JE
DVPS in infants occurs under completely antibody-DENV immune complexes). One of
different immunological circumstances the most critical absent studies is the charac-
than during first heterotypic dengue in- terization of infection, cytokine, and innate
264 HALSTEAD

immune responses of different immune com- butions to these differences have not been
plexes in primary human myeloid cells (which studied in the appropriate in vitro system, e.g.,
cells to use is an important question and is primary human myeloid cells from flavirus-
addressed below). Antibodies to JE complexed nave individuals.
with DENV result in mild ADE. Thus, this
immune complex sends messages down the DENV-2 genetic differences associated
tail of the FcR that are very different from a with a major increase in disease severity
complex formed by a DENV and antibodies of DENV-2 infection 20 years after infec-
from a primary dengue infection. What is the tion with DENV-1
exact effect on iADE, and what is the molec- What are the kinetics of growth of DENV-2 in
ular mechanism that controls response differ- the primary human myeloid cell assay system
ences, if any? with and without the NS1 amino acid changes?

Role of different myeloid cells Genetic differences in DENV-2 isolates


How myeloid cell phenotypes correlate with recovered from disease of enhanced
disease severity and iADE requires more severity 4 years after DENV-1 and 8 years
investigation. We must start by going back to after DENV-3
human autopsies and identifying macrophage The authors have a surfeit of research mate-
targets for DENV infection using comprehen- rials (138). The only sensible approach to
sive cell markers. These investigations are studying these phenomena is to test the
currently in their infancy (16). All human reagents in the ADE primary human myeloid
myeloid cells should be studied separately. cell system. This will have to be done on a large
scale. Differences in the viral productivity of
Role of different FcgRs in iADE cells infected using infectious immune com-
This is another huge field for research. It plexes should provide outcomes that can be
should be remembered the DVPS occurs in related to any of a number of viral, antibody, or
individuals who circulate myeloid cells that cellular differences.
express various FcRs, often on the same cell.
Ultimately, iADE needs to be studied using this Efferent
inherent complexity.
The final scenario that results in DVPS has been
Role of DENV in ADE amazingly difficult to learn about. Complement
split products and key cytokines have been
Enhanced growth of DENV-2 recovered thought to mediate vascular permeability at the
from severe compared with mild disease same time these factors were thought to
in primary human monocytes produce thrombocytopenia and altered hemo-
The simple experiment as designed in the stasis. The failure of cortisone administered
published paper should be repeated (117). early to suppress cytokine production or reduce
the incidence of vascular permeability was a
Differences in ability of DENV-2 strains wake-up call. An alternative mechanism is
to be neutralized by DENV-1 antibodies needed. I suggest that DVPS is a dengue
The actual sites on the virion that mediate toxemia. Dengue NS1 directly causes all of the
these differences in human disease expression elements of DVPSactivated complement,
are still unknown. Differences in genetic and thrombocytopenia, altered hemostasis, and vas-
viral structure between DENV-2 viruses that cular damage. As a direct toxin, NS1 should
are and are not associated with epidemic produce its damage in proportion to the con-
DHF/DSS during a first heterotypic dengue centration of NS1 presented to key systems and
infection are well established. Viral contri- tissues. ADE functions directly to increase the
CHAPTER 15 Dengue Antibody-Dependent Enhancement 265

concentration of NS1 proportionate to infected Identity of blood and tissue leukocytes supporting
cell mass. But why doesnt DVPS occur earlier in vitro infection. J Exp Med 146:218228.
8. Kliks S, Halstead SB. 1980. An explanation for
in the disease when peak NS1 blood levels are enhanced virus plaque formation in chick embryo
achieved? Why does DVPS suddenly worsen at cells. Nature 285:504505.
defervescence? I posit that this is related to the 9. Kliks S, Halstead SB. 1983. Role of antibodies
onset of killer T cell activity. T cells damage and host cells in plaque enhancement of Murray
dengue-infected cells, resulting in a final bolus Valley encephalitis virus. J Virol 46:394404.
10. Halstead SB. 1980. Immunological parameters of
release of NS1. When this exceeds a threshold,
Togavirus disease syndromes, p 107173. In
DVPS reaches its critical level of damage. This Schlesinger RW (ed), The Togaviruses, Biology,
hypothesis also includes the possibility that Structure, Replication. Academic Press, New York,
NS1-antibody complexes may retain significant NY.
toxicity. I challenge the dengue research com- 11. Boonpucknavig S, Boonpucknavig V,
Bhamarapravati N, Nimmannitya S. 1979. Im-
munity to prove me wrong.
munofluorescence study of skin rash in patients
with dengue hemorrhagic fever. Arch Pathol Lab
Med 103:463466.
ACKNOWLEDGMENT 12. Hall WC, Crowell TP, Watts DM, Barros VL,
Kruger H, Pinheiro F, Peters CJ. 1991. Demon-
Conflict of interest: I disclose no conflicts.
stration of yellow fever and dengue antigens in
formalin-fixed paraffin-embedded human liver by
immunohistochemical analysis. Am J Trop Med
CITATION Hyg 45:408417.
Halstead SB. 2014. Dengue antibody-depen- 13. Durbin AP, Vargas MJ, Wanionek K, Hammond
SN, Gordon A, Rocha C, Balmaseda A, Harris E.
dent enhancement: knowns and unknowns.
2008. Phenotyping of peripheral blood mononu-
Microbiol Spectrum 2(6):AID-0022-2014. clear cells during acute dengue illness
demonstrates infection and increased activation
of monocytes in severe cases compared to classic
REFERENCES dengue fever. Virology 376:429435.
1. Delgado MF, Coviello S, Monsalvo AC, Melendi 14. Balsitis SJ, Coloma J, Castro G, Alava A, Flores
GA, Hernandez JZ, Batalle JP, Diaz L, Trento A, D, McKerrow JH, Beatty PR, Harris E. 2009.
Chang HY, Mitzner W, Ravetch J, Melero JA, Tropism of dengue virus in mice and humans
Irusta PM, Polack FP. 2009. Lack of antibody defined by viral nonstructural protein 3-specific
affinity maturation due to poor Toll-like receptor immunostaining. Am J Trop Med Hyg 80:416424.
stimulation leads to enhanced respiratory syncy- 15. Jessie K, Fong MY, Devi S, Lam SK, Wong KT.
tial virus disease. Nat Med 15:3441. 2004. Localization of dengue virus in naturally
2. Hawkes RA. 1964. Enhancement of the infectivity infected human tissues, by immunohistochemis-
of arboviruses by specific antisera produced in try and in situ hybridization. J Infect Dis 189:1411
domestic fowls. Aust J Exp Biol Med Sci 42:465 1418.
482. 16. Aye KS, Charngkaew K, Win N, Wai KZ, Moe K,
3. Hawkes RA, Lafferty KJ. 1967. The enhancement Punyadee N, Thiemmeca S, Suttitheptumrong
of virus infectivity by antibody. Virology 33:250 A, Sukpanichnant S, Malasit P, Halstead SB.
261. 2014. Pathologic highlights of dengue hemorrhag-
4. Halstead SB, Chow J, Marchette NJ. 1973. ic fever in 13 autopsy cases from Myanmar. Hum
Immunologic enhancement of dengue virus rep- Pathol 45:12211233.
lication. Nat New Biol 243:2426. 17. Halstead SB. 1982. Immune enhancement of
5. Halstead SB, ORourke EJ. 1977. Antibody- viral infection. Prog Allergy 31:301364.
enhanced dengue virus infection in primate 18. Gollins SW, Porterfield JS. 1984. Flavivirus
leukocytes. Nature 265:739741. infection enhancement in macrophages: radioac-
6. Halstead SB, ORourke EJ. 1977. Dengue viruses tive and biological studies on the effect of antibody
and mononuclear phagocytes. I. Infection en- on virus fate. J Gen Virol 65:12611272.
hancement by non-neutralizing antibody. J Exp 19. Gollins SW, Porterfield JS. 1985. Flavivirus
Med 146:201217. infection enhancement in macrophages: an elec-
7. Halstead SB, ORourke EJ, Allison AC. 1977. tron microscopic study of viral cellular entry.
Dengue viruses and mononuclear phagocytes. II. J Gen Virol 66:19691982.
266 HALSTEAD

20. Olsen C, Scott F. 1993. Evaluation of antibody- 31. Halstead SB, Venkateshan CN, Gentry MK,
dependent enhancement of feline infectious peri- Larsen LK. 1984. Heterogeneity of infection
tonitis virus infectivity using in situ hybridization. enhancement of dengue 2 strains by monoclonal
Microb Pathog 14:275285. antibodies. J Immunol 132:15291532.
21. Robinson WE Jr, Montefiori DC, Gillespie DH, 32. Dejnirattisai W, Jumnainsong A, Onsirisakul N,
Mitchell WM. 1989. Complement-mediated, an- Fitton P, Vasanawathana S, Limpitikul W,
tibody-dependent enhancement of HIV-1 infec- Puttikhunt C, Edwards C, Duangchinda T,
tion in vitro is characterized by increased protein Supasa S, Chawansuntati K, Malasit P,
and RNA syntheses and infectious virus release. J Mongkolsapaya J, Screaton G. 2010. Cross-
Acquir Immune Defic Syndr 2:3342. reacting antibodies enhance dengue virus infection
22. Linn ML, Aaskov JG, Suhrbier A. 1996. Anti- in humans. Science 328:745748.
body-dependent enhancement and persistence in 33. Halstead SB, Nimmannitya S, Cohen SN. 1970.
macrophages of an arbovirus associated with Observations related to pathogenesis of dengue
arthritis. J Gen Virol 77:407411. hemorrhagic fever. IV. Relation of disease severity
23. Lidbury BA, Mahalingam S. 2000. Specific to antibody response and virus recovered. Yale J
ablation of antiviral gene expression in macro- Biol Med 42:311328.
phages by antibody-dependent enhancement of 34. Kliks SC, Nimmannitya S, Nisalak A, Burke DS.
Ross River virus infection. J Virol 74:83768381. 1988. Evidence that maternal dengue antibodies
24. Suhrbier A, La Linn M. 2003. Suppression of are important in the development of dengue
antiviral responses by antibody-dependent en- hemorrhagic fever in infants. Am J Trop Med
hancement of macrophage infection. Trends Hyg 38:411419.
Immunol 24:165168. 35. Libraty DH, Acosta LP, Tallo V, Segubre-
25. Halstead SB. 2009. Antibodies determine viru- Mercado E, Bautista A, Potts JA, Jarman RG,
lence in dengue. Ann NY Acad Sci 1171(Suppl 1): Yoon IK, Gibbons RV, Brion JD, Capeding RZ.
E48E56. 2009. A prospective nested case-control study of
26. Smith SA, Zhou Y, Olivarez NP, Broadwater dengue in infants: rethinking and refining the
AH, de Silva AM, Crowe JE Jr. 2012. Persistence antibody-dependent enhancement dengue hem-
of circulating B memory cell clones with potential orrhagic fever model. PLoS Med 6:e1000171.
for dengue virus disease enhancement for decades doi:10.1371/journal.pmed.1000171.
following infection. J Virol 86:26652675. 36. Simmons CP, Chau TN, Thuy TT, Tuan NM,
27. de Alwis R, Beltramello M, Messer WB, Hoang DM, Thien NT, Lien le B, Quy NT, Hieu
Sukupolvi-Petty S, Wahala WM, Kraus A, NT, Hien TT, McElnea C, Young P, Whitehead
Olivarez NP, Pham Q, Brian J, Tsai WY, Wang S, Hung NT, Farrar J. 2007. Maternal antibody
WK, Halstead S, Kliks S, Diamond MS, Baric R, and viral factors in the pathogenesis of dengue
Lanzavecchia A, Sallusto F, de Silva AM. 2011. In- virus in infants. J Infect Dis 196:416424.
depth analysis of the antibody response of indi- 37. Chau TN, Anders KL, Lien le B, Hung NT, Hieu
viduals exposed to primary dengue virus infection. LT, Tuan NM, Thuy TT, Phuong le T, Tham NT,
PLoS Negl Trop Dis 5:e1188. doi:10.1371/journal. Lanh MN, Farrar JJ, Whitehead SS, Simmons
pntd.0001188. CP. 2010. Clinical and virological features of
28. Beltramello M, Williams KL, Simmons CP, dengue in Vietnamese infants. PLoS Negl Trop
M a c a g n o A, Si mo n e l l i L , Q uy e n NT , Dis 4:e657. doi:10.1371/journal.pntd.0000657.
Sukupolvi-Petty S, Navarro-Sanchez E, Young 38. Chau TN, Hieu NT, Anders KL, Wolbers M,
PR, de Silva AM, Rey FA, Varani L, Whitehead Lien le B, Hieu LT, Hien TT, Hung NT, Farrar J,
SS, Diamond MS, Harris E, Lanzavecchia A, Whitehead S, Simmons CP. 2009. Dengue virus
Sallusto F. 2010. The human immune response to infections and maternal antibody decay in a
dengue virus is dominated by highly cross- prospective birth cohort study of Vietnamese
reactive antibodies endowed with neutralizing infants. J Infect Dis 200:18931900.
and enhancing activity. Cell Host Microbe 8:271 39. Chau TN, Quyen NT, Thuy TT, Tuan NM,
283. Hoang DM, Dung NT, Lien le B, Quy NT, Hieu
29. Wahala WM, Kraus AA, Haymore LB, NT, Hieu LT, Hien TT, Hung NT, Farrar J,
Accavitti-Loper MA, de Silva AM. 2009. Dengue Simmons CP. 2008. Dengue in Vietnamese
virus neutralization by human immune sera: role infants: results of infection-enhancement assays
of envelope protein domain III-reactive antibody. correlate with age-related disease epidemiology,
Virology 392:103113. and cellular immune responses correlate with
30. Halstead SB. 2003. Neutralization and antibody disease severity. J Infect Dis 198:516524.
dependent enhancement of dengue viruses. Adv 40. Halstead SB, Mahalingam S, Marovich MA,
Virus Res 60:421467. Ubol S, Mosser DM. 2010. Intrinsic antibody-
CHAPTER 15 Dengue Antibody-Dependent Enhancement 267

dependent enhancement of microbial infection in Nisalak A. 2000. Dengue viremia titer, antibody
macrophages: disease regulation by immune com- response pattern, and virus serotype correlate
plexes. Lancet Infect Dis 10:712722. with disease severity. J Infect Dis 181:29.
41. Ubol S, Halstead SB. 2010. How innate immune 54. Libraty DH, Endy TP, Houng HS, Green S,
mechanisms contribute to antibody-enhanced viral Kalayanarooj S, Suntayakorn S, Chansiriwongs
infections. Clin Vaccine Immunol 17:18291835. W, Vaughn DW, Nisalak A, Ennis FA, Rothman
42. Boonnak K, Dambach KM, Donofrio GC, AL. 2002. Differing influences of virus burden and
Marovich MA. 2011. Cell type specificity and immune activation on disease severity in second-
host genetic polymorphisms influence antibody ary dengue-3 virus infections. J Infect Dis 185:
dependent enhancement of dengue virus infec- 12131221.
tion. J Virol 85:16711683. 55. Wu SJ, Grouard-Vogel G, Sun W, Mascola JR,
43. Sullivan NJ. 2001. Antibody-mediated enhance- Brachtel E, Putvatana R, Louder MK, Filgueira
ment of viral disease. Curr Top Microbiol Immunol L, Marovich MA, Wong HK, Blauvelt A, Murphy
260:145169. GS, Robb ML, Innes BL, Birx DL, Hayes CG,
44. Tirado SM, Yoon KJ. 2003. Antibody-dependent Frankel SS. 2000. Human skin Langerhans cells
enhancement of virus infection and disease. Viral are targets of dengue virus infection [see
Immunol 16:6986. comments]. Nat Med 6:816820.
45. Huisman W, Martina BE, Rimmelzwaan GF, 56. Montoya M, Gresh L, Mercado JC, Williams KL,
Gruters RA, Osterhaus AD. 2009. Vaccine- Vargas MJ, Gutierrez G, Kuan G, Gordon A,
induced enhancement of viral infections. Vaccine Balmaseda A, Harris E. 2013. Symptomatic versus
27:505512. inapparent outcome in repeat dengue virus infec-
46. Halstead SB. 1970. Observations related to patho- tions is influenced by the time interval between
genesis of dengue hemorrhagic fever. VI. Hypoth- infections and study year. PLoS Negl Trop Dis 7:
eses and discussion. Yale J Biol Med 42:350362. e2357. doi:10.1371/journal.pntd.0002357.
47. Gibbons RV, Kalanarooj S, Jarman RG, Nisalak 57. Gordon A, Kuan G, Mercado JC, Gresh L, Aviles
A, Vaughn DW, Endy TP, Mammen MP Jr, W, Balmaseda A, Harris E. 2013. The Nicaraguan
Srikiatkhachorn A. 2007. Analysis of repeat pediatric dengue cohort study: incidence of
hospital admissions for dengue to estimate the inapparent and symptomatic dengue virus infec-
frequency of third or fourth dengue infections tions, 20042010. PLoS Negl Trop Dis 7:e2462.
resulting in admissions and dengue hemorrhagic doi:10.1093/aje/kwp092.
fever, and serotype sequences. Am J Trop Med 58. Anderson KB, Gibbons RV, Cummings DA,
Hyg 77:910913. Nisalak A, Green S, Libraty DH, Jarman RG,
48. Halstead SB, Lan NT, Myint TT, Shwe TN, Srikiatkhachorn A, Mammen MP, Darunee B,
Nisalak A, Soegijanto S, Vaughn DW, Endy T. Yoon IK, Endy TP. 2013. A shorter time interval
2002. Infant dengue hemorrhagic fever: research between first and second dengue infections is
opportunities ignored. Emerg Infect Dis 12:1474 associated with protection from clinical illness in
1479. a school-based cohort in Thailand. J Infect Dis
49. Coffey LL, Mertens E, Brehin AC, Fernandez- 209:360368.
Garcia MD, Amara A, Despres P, Sakuntabhai A. 59. Grange L, Simon-Loriere E, Sakuntabhai A,
2009. Human genetic determinants of dengue Gresh L, Paul R, Harris E. 2014. Epidemiological
virus susceptibility. Microbes Infect 11:143156. risk factors associated with high global frequency
50. Sabin AB. 1952. Research on dengue during of inapparent dengue virus infections. Front
World War II. Am J Trop Med Hyg 1:3050. Immunol 5:280.
51. Chan KR, Zhang SL, Tan HC, Chan YK, Chow 60. Guzman MG, Kouri G, Valdes L, Bravo J,
A, Lim AP, Vasudevan SG, Hanson BJ, Ooi Vazquez S, Halstead SB. 2002. Enhanced sever-
EE. 2011. Ligation of Fc gamma receptor IIB ity of secondary dengue-2 infections: death rates
inhibits antibody-dependent enhancement of in 1981 and 1997 Cuban outbreaks. Rev Panam
dengue virus infection. Proc Natl Acad Sci USA Salud Publica 11:223227.
108:1247912484. 61. Guzman MG, Alvarez M, Rodriguez-Roche R,
52. Kliks SC, Nisalak A, Brandt WE, Wahl L, Burke Bernardo L, Montes T, Vazquez S, Morier L,
DS. 1989. Antibody-dependent enhancement of Alvarez A, Gould EA, Kouri G, Halstead SB.
dengue virus growth in human monocytes as a risk 2007. Neutralizing antibodies after infection with
factor for dengue hemorrhagic fever. Am J Trop dengue 1 virus. Emerg Infect Dis 13:282286.
Med Hyg 40:444451. 62. Sangkawibha N, Rojanasuphot S, Ahandrik S,
53. Vaughn DW, Green S, Kalayanarooj S, Innis BL, Viriyapongse S, Jatanasen S, Salitul V,
Nimmannitya S, Suntayakorn S, Endy TP, Phanthumachinda B, Halstead SB. 1984. Risk
Raengsakulrach B, Rothman AL, Ennis FA, factors in dengue shock syndrome: a prospective
268 HALSTEAD

epidemiologic study in Rayong, Thailand. I. The preferential recognition of immature flaviviruses


1980 outbreak. Am J Epidemiol 120:653669. by a fusion-loop antibody. EMBO J 28:32693276.
63. Halstead SB, Porterfield JS, ORourke EJ. 1980. 73. Sutterwala FS, Noel GJ, Clynes R, Mosser DM.
Enhancement of dengue virus infection in 1997. Selective suppression of interleukin-12 in-
monocytes by flavivirus antisera. Am J Trop Med duction after macrophage receptor ligation. J Exp
Hyg 29:638642. Med 185:19771985.
64. Anderson KB, Gibbons RV, Thomas SJ, 74. Sutterwala FS, Noel GJ, Salgame P, Mosser DM.
Rothman AL, Nisalak A, Berkelman RL, Libraty 1998. Reversal of proinflammatory responses by
DH, Endy TP. 2011. Preexisting Japanese en- ligating the macrophage Fcgamma receptor type I.
cephalitis virus neutralizing antibodies and in- J Exp Med 188:217222.
creased symptomatic dengue illness in a school- 75. Anderson CF, Lucas M, Gutierrez-Kobeh L,
based cohort in Thailand. PLoS Negl Trop Dis 5: Field AE, Mosser DM. 2004. T cell biasing by
e1311. doi:10.1371/journal.pntd.0001311. activated dendritic cells. J Immunol 173:955961.
65. Hoke CH Jr,Nisalak A, Sangawhipa N, 76. Ioan-Facsinay A, de Kimpe SJ, Hellwig SM, van
Jatanasen S, Laorakpongse T, Innis BL, Lent PL, Hofhuis FM, van Ojik HH, Sedlik C, da
Kotchasenee S, Gingrich JB, Latendresse J, Silveira SA, Gerber J, de Jong YF, Roozendaal
Fukai K, et al. 1988. Protection against Japanese R, Aarden LA, van den Berg WB, Saito T,
encephalitis by inactivated vaccines. N Engl J Med Mosser D, Amigorena S, Izui S, van Ommen
319:608614. GJ, van Vugt M, van de Winkel JG, Verbeek JS.
66. Lai CY, Tsai WY, Lin SR, Kao CL, Hu HP, King 2002. FcgammaRI (CD64) contributes substan-
CC, Wu HC, Chang GJ, Wang WK. 2008. tially to severity of arthritis, hypersensitivity
Antibodies to envelope glycoprotein of dengue responses, and protection from bacterial infec-
virus during the natural course of infection are tion. Immunity 16:391402.
predominantly cross-reactive and recognize 77. Kima PE, Constant SL, Hannum L, Colmenares
epitopes containing highly conserved residues at M, Lee KS, Haberman AM, Shlomchik MJ,
the fusion loop of domain II. J Virol 82:66316643. McMahon-Pratt D. 2000. Internalization of Leish-
67. Yu IM, Zhang W, Holdaway HA, Li L, mania mexicana complex amastigotes via the Fc
Kostyuchenko VA, Chipman PR, Kuhn RJ, receptor is required to sustain infection in murine
Rossmann MG, Chen J. 2008. Structure of the cutaneous leishmaniasis. J Exp Med 191:10631068.
immature dengue virus at low pH primes proteo- 78. Padigel UM, Farrell JP. 2005. Control of infec-
lytic maturation. Science 319:18341837. tion with Leishmania major in susceptible BALB/c
68. Rodenhuis-Zybert IA, Wilschut J, Smit JM. 2011. mice lacking the common gamma-chain for FcR is
Partial maturation: an immune-evasion strategy of associated with reduced production of IL-10 and
dengue virus? Trends Microbiol 19:248254. TGF-beta by parasitized cells. J Immunol 174:
69. Rodenhuis-Zybert IA, van der Schaar HM, 63406345.
Voorham JMD, van der Ende-Metselaar H, 79. Mosser DM. 2003. The many faces of macro-
Lei HY, Wilschut J, Smit JM. 2010. Immature phage activation. J Leukoc Biol 73:209212.
dengue virus: a veiled pathogen? PLoS Pathog 6: 80. Miles SA, Conrad SM, Alves RG, Jeronimo SM,
e1000718. doi:10.1371/journal.ppat.1000718. Mosser DM. 2005. A role for IgG immune
70. Rodenhuis-Zybert IA, Moesker B, da Silva complexes during infection with the intracellular
Voorham JM, van der Ende-Metselaar H, Dia- pathogen Leishmania. J Exp Med 201:747754.
mond MS, Wilschut J, Smit JM. 2011. A fusion- 81. Anderson CF, Gerber JS, Mosser DM. 2002.
loop antibody enhances the infectious properties Modulating macrophage function with IgG im-
of immature flavivirus particles. J Virol 85:11800 mune complexes. J Endotoxin Res 8:477481.
11808. 82. Lucas M, Zhang X, Prasanna V, Mosser DM.
71. da Silva Voorham JM, Rodenhuis-Zybert IA, 2005. ERK activation following macrophage
Ayala Nunez NV, Colpitts TM, van der Ende- FcgammaR ligation leads to chromatin modifica-
Metselaar H, Fikrig E, Diamond MS, Wilschut J, tions at the IL-10 locus. J Immunol 175:469477.
Smit JM. 2012. Antibodies against the envelope 83. Murray HW, Masur H, Keithly JS. 1982. Cell-
glycoprotein promote infectivity of immature mediated immune response in experimental vis-
dengue virus serotype 2. PLoS One 7:e29957. ceral leishmaniasis. I. Correlation between resis-
doi:10.1371/journal.pone.0029957. tance to Leishmania donovani and lymphokine-
72. Cherrier MV, Kaufmann B, Nybakken GE, Lok generating capacity. J Immunol 129:344350.
SM, Warren JT, Chen BR, Nelson CA, 84. Yang Z, Mosser DM, Zhang X. 2007. Activation of
Kostyuchenko VA, Holdaway HA, Chipman the MAPK, ERK, following Leishmania ama-
PR, Kuhn RJ, Diamond MS, Rossmann MG, zonensis infection of macrophages. J Immunol
Fremont DH. 2009. Structural basis for the 178:10771085.
CHAPTER 15 Dengue Antibody-Dependent Enhancement 269

85. Chareonsirisuthigul T, Kalayanarooj S, Ubol S. 95. Chaturvedi UC, Raghupathy R, Pasca AS,
2007. Dengue virus (DENV) antibody-dependent Elbishbishi EA, Agarwal R, Nagar R, Misra A,
enhancement of infection upregulates the produc- Kapoor S, Mathur A, Khan M, Azizieh F. 1999.
tion of anti-inflammatory cytokines, but suppresses Shift from a Th1-type response to Th1-type in
anti-DENV free radical and pro-inflammatory dengue haemorrhagic fever. Curr Sci 76:6369.
cytokine production, in THP-1 cells. J Gen Virol 96. Kou Z, Quinn M, Chen H, Rodrigo WW, Rose
88:365375. RC, Schlesinger JJ, Jin X. 2008. Monocytes, but
86. Ubol S, Phuklia W, Kalayanarooj S, Modhiran not T or B cells, are the principal target cells for
N. 2010. Mechanisms of immune evasion in- dengue virus (DV) infection among human pe-
duced by a complex of dengue virus and pre- ripheral blood mononuclear cells. J Med Virol
existing enhancing antibodies. J Infect Dis 201: 80:134146.
923935. 97. Blackley S, Kou Z, Chen H, Quinn M, Rose RC,
87. Modhiran N, Kalayanarooj S, Ubol S. 2010. Schlesinger JJ, Coppage M, Jin X. 2007. Primary
Subversion of innate defenses by the interplay human splenic macrophages, but not T or B cells,
between DENV and pre-existing enhancing anti- are the principal target cells for dengue virus
bodies: TLRs signaling collapse. PLoS Negl Trop infection in vitro. J Virol 81:1332513334.
Dis 4:e924. doi:10.1371/journal.pntd.0000924. 98. Cohen SN, Halstead SB. 1966. Shock associated
88. Ubol S, Masrinoul P, Chaijaruwanich J, with dengue infection. I. Clinical and physiologic
Kalayanarooj S, Charoensirisuthikul T, Kasisith manifestations of dengue hemorrhagic fever in
J. 2008. Differences in global gene expression in Thailand, 1964. J Pediatrics 68:448456.
peripheral blood mononuclear cells indicate a 99. Rothman AL. 2010. Cellular immunology of
significant role of the innate responses in pro- sequential dengue virus infection and its role in
gression of dengue fever but not dengue hemor- disease pathogenesis. Curr Top Microbiol Immu-
rhagic fever. J Infect Dis 197:14591467. nol 338:8398.
89. Chen LC, Lei HY, Liu CC, Shiesh SC, Chen SH, 100. Bethell DB, Flobbe K, Cao XT, Day NP, Pham
Liu HS, Lin YS, Wang ST, Shyu HW, Yeh TM. TP, Buurman WA, Cardosa MJ, White NJ,
2006. Correlation of serum levels of macrophage Kwiatkowski D. 1998. Pathophysiologic and
migration inhibitory factor with disease severity prognostic role of cytokines in dengue hemor-
and clinical outcome in dengue patients. Am J rhagic fever. J Infect Dis 177:778782.
Trop Med Hyg 74:142147. 101. Green S, Vaughn DW, Kalayanarooj S,
90. Simmons CP, Popper S, Dolocek C, Chau TN, Suntayakorn S, Nisalak A, Nimmannitya S,
Griffiths M, Dung NT, Long TH, Hoang DM, Innis BL, Kurane I, Rothman AL, Ennis FA.
Chau NV, Thao le TT, Hien TT, Relman DA, 1997. 46th Meeting of the American Society of
Farrar J. 2007. Patterns of host genome-wide Tropical Medicine and Hygiene, Orlando, FL.
gene transcript abundance in the peripheral blood AJTMH 57:113.
of patients with acute dengue hemorrhagic fever. 102. Halstead SB, Rojanasuphot S, Sangkawibha N.
J Infect Dis 195:10971107. 1983. Original antigenic sin in dengue. Am J Trop
91. Nguyen TH, Lei HY, Nguyen TL, Lin YS, Huang Med Hyg 32:154156.
KJ, Le BL, Lin CF, Yeh TM, Do QH, Vu TQ, 103. Thein S, Aung MM, Shwe TN, Aye M, Zaw A,
Chen LC, Huang JH, Lam TM, Liu CC, Halstead Aye K, Aye KM, Aaskov J. 1997. Risk factors in
SB. 2004. Dengue hemorrhagic fever in infants: a dengue shock syndrome. Am J Trop Med Hyg
study of clinical and cytokine profiles. J Infect Dis 56:566572.
189:221232. 104. Graham RR, Juffrie M, Tan R, Hayes CG,
92. Shresta S, Kyle JL, Snider HM, Basavapatna M, Laksono I, Maroef C, Erlin, Sutaryo,Porter KR,
Beatty PR, Harris E. 2004. Interferon-dependent Halstead SB. 1999. A prospective sero-
immunity is essential for resistance to primary epidemiologic study on dengue in children four to
dengue virus infection in mice, whereas T- and B- nine years of age in Yogyakarta, Indonesia I. Studies
cell-dependent immunity are less critical. J Virol in 19951996. Am J Trop Med Hyg 61:412419.
78:27012710. 105. Chungue E, Spiegel A, Roux J, Laudon F,
93. Shresta S, Sharar KL, Prigozhin DM, Snider Cardines R. 1990. Dengue-3 in French Polynesia:
HM, Beatty PR, Harris E. 2005. Critical roles for preliminary data. Med J Aust 152:557558.
both STAT1-dependent and STAT1-independent 106. Hubert B, Halstead SB. 2009. Dengue 1 virus and
pathways in the control of primary dengue virus dengue hemorrhagic fever, French Polynesia,
infection in mice. J Immunol 175:39463954. 2001. Emerg Infect Dis 15:12651270.
94. Navarro-Sanchez E, Despres P, Cedillo-Barron 107. Kou Z, Lim JY, Beltramello M, Quinn M, Chen
L. 2005. Innate immune responses to dengue H, Liu SN, Martinez-Sobrido L, Diamond MS,
virus. Arch Med Res 36:425435. Schlesinger JJ, de Silva A, Sallusto F, Jin X. 2011.
270 HALSTEAD

Human antibodies against dengue enhance den- with severe and mild dengue disease. Am J Trop
gue viral infectivity without suppressing type I Med Hyg 45:644651.
interferon secretion in primary human mono- 118. Rico-Hesse R. 2003. Microevolution and viru-
cytes. Virology 410:240247. lence of dengue viruses. Adv Virus Res 59:315341.
108. Chotiwan N, Roehrig JT, Schlesinger JJ, Blair 119. Anderson CR, Downs WG. 1956. Isolation of
CD, Huang CY. 2014. Molecular determinants of dengue virus from a human being in Trinidad.
dengue virus 2 envelope protein important for Science 124:224225.
virus entry in FcgammaRIIA-mediated antibody- 120. Rosen L. 1958. Observations on the epidemiology
dependent enhancement of infection. Virology of dengue in Panama. Am J Hyg 68:4558.
456457:238246. 121. Guzman MG, Kouri GP, Bravo J, Soler M,
109. Laoprasopwattana K, Libraty D, Endy T, Nisalak Vazquez S, Morier L. 1990. Dengue hemorrhagic
A, Chunsittiwat S, Vaughn DW, Reed G, Ennis fever in Cuba, 1981: a retrospective seroepide-
FA, Rothman A, Green S. 2005. Dengue virus miologic study. Am J Trop Med Hyg 42:179184.
(DV) enhancing antibody activity in preillness 122. Neff JM, Morris L, Gonzalez-Alcover R,
plasma does not predict subsequent disease sever- Coleman PH, Lyss SB, Negron H. 1967. Dengue
ity or viremia in secondary DV infection. J Infect fever in a Puerto Rican community. Am J Epide-
Dis 192:510519. miol 86:162184.
110. Rodrigo WW, Jin X, Blackley SD, Rose RC, 123. Kouri GP, Guzman MG, Bravo JR, Triana C.
Schlesinger JJ. 2006. Differential enhancement 1989. Dengue haemorrhagic fever/dengue shock
of dengue virus immune complex infectivity syndrome: lessons from the Cuban epidemic, 1981.
mediated by signaling-competent and signaling- Bull World Health Organ 67:375380.
incompetent human Fcgamma RIA (CD64) or 124. Guzman MG, Deubel V, Pelegrino JL, Rosario
FcgammaRIIA (CD32). J Virol 80:1012810138. D, Marrero M, Sariol C, Kouri G. 1995. Partial
111. Moi ML, Lim CK, Takasaki T, Kurane I. 2010. nucleotide and amino acid sequences of the
Involvement of the Fc{gamma} receptor IIA envelope and the envelope/nonstructural pro-
cytoplasmic domain in antibody-dependent en- tein-1 gene junction of four dengue-2 virus strains
hancement of dengue virus infection. J Gen Virol isolated during the 1981 Cuban epidemic. Am J
91:103111. Trop Med Hyg 52:241246.
112. Moi ML, Lim CK, Kotaki A, Takasaki T, Kurane 125. Watts DM, Porter KR, Putvatana P, Vasquez B,
I. 2010. Development of an antibody-dependent Calampa C, Hayes CG, Halstead SB. 1999. Failure
enhancement assay for dengue virus using stable of secondary infection with American genotype
BHK-21 cell lines expressing FcgammaRIIA. dengue 2 to cause dengue haemorrhagic fever [see
J Virol Methods 163:205209. comments]. Lancet 354:14311434.
113. Moi ML, Lim CK, Kotaki A, Takasaki T, Kurane 126. Leitmeyer KC, Vaughn DW, Watts DM, Salas R,
I. 2010. Discrepancy in dengue virus neutralizing Villalobos I, de Chacon, Ramos C, Rico-Hesse R.
antibody titers between plaque reduction neutral- 1999. Dengue virus structural differences that
izing tests with Fcgamma receptor (FcgammaR)- correlate with pathogenesis. J Virol 73:47384747.
negative and FcgammaR-expressing BHK-21 127. Kochel TJ, Watts DM, Halstead SB, Hayes CG,
cells. Clin Vaccine Immunol 17:402407. Espinosa A, Felices V, Caceda R, Bautista T,
114. Moi ML, Lim CK, Kotaki A, Takasaki T, Kurane Montoya Y, Douglas S, Russell KL. 2002. Effect
I. 2011. Detection of higher levels of dengue of dengue-1 antibodies on American dengue-2
viremia using FcgammaR-expressing BHK-21 viral infection and dengue haemorrhagic fever.
cells than FcgammaR-negative cells in secondary Lancet 360:310312.
infection but not in primary infection. J Infect Dis 128. Vasilakis N, Weaver SC. 2008. The history and
203:14051414. evolution of human dengue emergence. Adv Virus
115. Moi ML, Takasaki T, Saijo M, Kurane I. 2014. Res 72:176.
Determination of antibody concentration as the 129. Guzman MG, Kouri G, Halstead SB. 2000. Do
main parameter in a dengue virus antibody- escape mutants explain rapid increases in dengue
dependent enhancement assay using FcgammaR- case-fatality rates within epidemics? Lancet
expressing BHK cells. Arch Virol 159:103116. 355:19021903.
116. Buxbaum LU. 2011. Type I IFNs promote the early 130. Rodriguez-Roche R, Sanchez L, Burgher Y,
IFN-gamma response and the IL-10 response in Rosario D, Alvarez M, Kouri G, Halstead SB,
Leishmania mexicana infection. Parasite Immunol Gould EA, Guzman MG. 2011. Virus role during
32:153160. intraepidemic increase in dengue disease severity.
117. Morens DM, Marchette NJ, Chu MC, Halstead Vector Borne Zoonotic Dis 11:675681.
SB. 1991. Growth of dengue type 2 virus isolates in 131. Alvarez M, Rodriguez R, Bernardo L, Vasquez
human peripheral blood leukocytes correlates S, Morier L, Gonzalez D, Castro O, Kouri G,
CHAPTER 15 Dengue Antibody-Dependent Enhancement 271

Halstead SB, Guzman MG. 2006. Dengue hem- genic role of complement in dengue hemorrhagic
orrhagic fever caused by sequential dengue 1 - 3 shock syndrome. N Engl J Med 289:9961000.
infections at a long interval: Havana epidemic, 143. Memoranda. 1973. Pathogenic mechanisms in
20012002. Am J Trop Med Hyg 75:11131117. dengue hemorrhagic fever. Report of an interna-
132. Gonzalez D, Castro OE, Kouri G, Perez JM, tional collaborative study. Bull. WHO 48:117132.
Martinez E, Vazquez S, Rosario D, Cancio R, 144. Halstead SB. 2008. Pathophysiology, p 285326.
Guzman MG. 2005. Classical dengue hemorrhag- In Halstead SB (ed), Dengue, vol. 5. Imperial
ic fever resulting from two dengue infections College Press, London, United Kingdom.
spaced 20 years or more apart: Havana, dengue 3 145. Wills B, Tran VN, Nguyen TH, Truong TT, Tran
epidemic, 20012. Int J Infect Dis 9:280285. TN, Nguyen MD, Tran VD, Nguyen VV, Dinh
133. Rodriguez-Roche R, Alvarez M, Gritsun T, TT, Farrar J. 2009. Hemostatic changes in
Rosario D, Halstead SB, Kouri G, Guzman MG. Vietnamese children with mild dengue correlate
2005. Dengue virus type 2 in Cuba, 1997: conser- with the severity of vascular leakage rather than
vation of E gene sequence in isolates obtained at bleeding. Am J Trop Med Hyg 81:638644.
different times during the epidemic. Arch Virol 146. Libraty DH, Young PR, Pickering D, Endy TP,
150:415425. Kalayanarooj S, Green S, Vaughn DW, Nisalak A,
134. Rodriguez-Roche R, Alvarez M, Gritsun T, Ennis FA, Rothman AL. 2002. High circulating
Halstead SB, Kouri G, Gould EA, Guzman MG. levels of the dengue virus nonstructural protein
2005. Virus evolution during a severe dengue NS1 early in dengue illness correlate with the
epidemic in Cuba, 1997. Virology 334:154159. development of dengue hemorrhagic fever. J Infect
135. Blok J, Gibbs AJ, McWilliam SM, Vitarana UT. Dis 186:11651168.
1991. NS 1 gene sequences from eight dengue-2 147. Kalayanarooj S, Nimmannitya S. 2005. Is dengue
viruses and their evolutionary relationships with severity related to nutritional status? Southeast
other dengue-2 viruses. Arch Virol 118:209223. Asian J Trop Med Public Health 36:378384.
136. Halstead SB. 2014. Intraepidemic increases in 148. Nimmannitya S, Halstead SB, Cohen S,
dengue disease severity: applying lessons on Margiotta MR. 1969. Dengue and chikungunya
surveillance and transmission, p 84101. In virus infection in man in Thailand, 19621964.
Whitehorn J, Farrar J (ed), Clinical Insights: I. Observations on hospitalized patients with
Dengue Fever: Transmission, Diagnosis and Sur- hemorrhagic fever. Am J Trop Med Hyg 18:
veillance. Future Medicine, London, United King- 954971.
dom. 149. Anders KL, Nguyet NM, Chau NV, Hung NT,
137. Chen HL, Lin SR, Liu HF, King CC, Hsieh SC, Thuy TT, Lien le B, Farrar J, Wills B, Hien TT,
Wang WK. 2008. Evolution of dengue virus type 2 Simmons CP. 2011. Epidemiological factors asso-
during two consecutive outbreaks with an in- ciated with dengue shock syndrome and mortality
crease in severity in southern Taiwan in 2001 in hospitalized dengue patients in Ho Chi Minh
2002. Am J Trop Med Hyg 79:495505. City, Vietnam. Am J Trop Med Hyg 84:127134.
138. OhAinle M, Balmaseda A, Macalalad AR, Tellez 150. Pang T, Cardosa MJ, Guzman MG. 2007. Of
Y, Zody MC, Saborio S, Nunez A, Lennon NJ, cascades and perfect storms: the immuno-
Birren BW, Gordon A, Henn MR, Harris E. 2011. pathogenesis of dengue haemorrhagic fever-den-
Dynamics of dengue disease severity determined by gue shock syndrome (DHF/DSS). Immunol Cell Biol
the interplay between viral genetics and serotype- 85:4345.
specific immunity. Sci Transl Med 3:114ra128. 151. Dung NT, Duyen HT, Thuy NT, Ngoc TV, Chau
139. Farrar J. 2008. Clinical features, p 121191. In NV, Hien TT, Rowland-Jones SL, Dong T,
Halstead SB (ed), Dengue, vol. 1. Imperial College Farrar J, Wills B, Simmons CP. 2010. Timing of
Press, London, United Kingdom. CD8+ T cell responses in relation to commence-
140. Halstead SB, Streit TG, Lafontant JG, ment of capillary leakage in children with dengue.
Putvatana R, Russell K, Sun W, Kanesa-Thasan J Immunol 184:72817287.
N, Hayes CG, Watts DM. 2001. Haiti: absence of 152. Larena M, Regner M, Lee E, Lobigs M. 2011.
dengue hemorrhagic fever despite hyperendemic Pivotal role of antibody and subsidiary contribu-
dengue virus transmission. Am J Trop Med Hyg tion of CD8+ T cells to recovery from infection in a
65:180183. murine model of Japanese encephalitis. J Virol
141. Halstead SB. 2008. Pathogenesis: risk factors prior 85:54465455.
to infection, p 219256. In Halstead SB (ed), 153. Pan American Health Organization. 1978. Den-
Dengue, vol. 5. Imperial College Press, London, gue in the Caribbean, 1977. Proceedings of a
United Kingdom. workshop held in Montego Bay, Jamaica, 8 to 11
142. Bokisch VA, Top FH Jr, Russell PK, Dixon FJ, May 1978. Scientific Publication 375. Pan Amer-
Muller-Eberhard HJ. 1973. The potential patho- ican Health Organization, Washington, DC.
Immunotherapeutic Approaches
To Prevent Cytomegalovirus-
Mediated Disease

EDITH ACQUAYE-SEEDAH,1 ZACHARY P. FRYE,2 and JENNIFER A. MAYNARD2


16
INTRODUCTION

First visualized in 1904 as large inclusions in tissue sections from luetic infants
and isolated in 1957 (1), the human cytomegalovirus (CMV) is a remarkably
successful pathogen. Worldwide, there is a 50 to 90% probability of infection by
age 50 without any clear markers of genetic susceptibility. Primary infection
results in life-long latency, requiring continuous vigilance by the host immune
system and characterized by serum antibody titers and a strong cytotoxic T-cell
response. While most individuals will be infected with at least one strain of CMV,
infection rarely leads to disease in immunocompetent individuals. However,
CMV is a primary cause of congenital neurological defects and causes disease in
those with compromised immune systems, such as transplant patients, with only
limited therapies available.
Symptomatic CMV disease is often observed in infants who received the virus
from an infected mother in utero, patients receiving hematopoietic stem cell or
solid organ transplants, and patients with immunosuppressive diseases such as
human immunodeficiency virus (HIV) and AIDS (Table 1). Primary maternal
infection during gestation represents an 40% risk of intrauterine transmission to

1
Department of Biochemistry, University of Texas at Austin, Austin, TX 78712; 2Department of Chemical
Engineering, University of Texas at Austin, Austin, TX 78712.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0009-2013

273
274 ACQUAYE-SEEDAH ET AL.

TABLE 1 Treatment spectrum for high-risk CMV demographic groups


Demographic Infection risk Treatment

Healthy individual Primary infection, followed by latency; None; typically asymptomatic


occasionally mononucleosis syndrome
similar to that caused by Epstein-Barr virus
Fetus of CMV- mother, Primary infection with high risk of serious Antiviral therapy or IVIG provided to
newborn neurological effects (e.g., hearing loss, mother during gestation to prevent
retardation, microcephaly, seizures in primary infection
1017%); high mortality rate of those
with symptomatic disease (30% reported)
CMV- solid organ transplant Primary infection risk if the donor is CMV+; IVIG to prevent primary infection
patient historically, 1050% develop symptomatic Cellular immunotherapy with
disease including leukopenia, retinitis, genetically modied T cells expressing
tissue-invasive disease (e.g., nephritis, CMV-specic TCRs
pancreatitis); increased graft rejection;
early disease during the rst 3 months
has been reduced signicantly due to
prophylactic antiviral therapy
CMV+ solid organ transplant Lower risk of secondary infection due to Antiviral therapy
patient reactivation of latent infection while
immunosuppressed; risk of acquiring a
different CMV strain if the donor is CMV+;
increased graft rejection
CMV+ and CMV- stem cell Primary infection (30% CMV- recipients) Antiviral therapy for rst 100 days
transplant patient and secondary infection (70% CMV+ posttransplant; high mortality rate if
recipients) due to reactivation of latent CMV disease appears after the rst
infection while immunosuppressed; most 100 days (50%) Adoptive transfer of
common form of disease is CMV pneumonia CMV-specic T cells isolated from
with a 3060% mortality rate donor material to prevent infection
Some evidence to support IVIG
HIV/AIDS patient Primary or secondary infection when CD4+ Antiviral therapy
T cell count is <50/l; retinitis, pneumonitis

the fetus, with congenital viral infection affect- CMV infection and transplant failure, while
ing 0.5 to 3% of all births (40,000 per year in secondary infection may result due to reacti-
the United States)more children than are vation of latent CMV in a CMV+ recipient.
affected with Down syndrome, fetal alcohol Similar risks are observed for HIV and AIDS
syndrome, and spina bifida (2). The conse- patients with T-cell counts of <50/l and high-
quences of congenital disease are severe: vision risk elderly with reduced naive T-cell receptor
and hearing loss, mental retardation, and even (TCR) diversity (4) but are less common given
death, amounting to an estimated $4 billion per the prevalence of antiretroviral therapies.
year in the United States alone. The rate of For these reasons, when the Institute of
vertical transmission is reduced to 2% in Medicine was commissioned in 1999 to prior-
pregnant women who have previously been itize vaccine development based upon quality-
infected with CMV and have circulating anti- adjusted life years, a vaccine to prevent CMV
CMV antibodies (3), suggesting a protective was judged head and shoulders above all
role for antibodies against primary infection. other potential vaccines with regard to cost
Solid organ transplantation, particularly of a effectiveness (estimated at $300,000 per af-
CMV seropositive (CMV+) organ into a CMV fected child) (5). Remarkably, while a CMV
seronegative (CMV-) recipient, with the asso- vaccine has been a top priority for nearly 40
ciated immunosuppressive regimen, greatly years (5, 6), none of the candidates evaluated
increases patient susceptibility to primary has been licensed. One challenge has been the
CHAPTER 16 Immunotherapeutic Approaches to Prevent CMV Disease 275

lack of a clear serological correlate of protec-


tion, which may be different for the different
patient groups. In the meantime, antiviral
therapeutics are the standard of care, with
high-titer CMV immunoglobulin and adoptive
T-cell transfer emerging as alternatives. Con-
sidering the success of antibody therapeutics
to treat some viral diseases (7), the apparent
protective role of antibodies against CMV and
recent identification of neutralizing epitopes in FIGURE 1 Human CMV structure. The 235-kb double-
CMV, antibody therapeutics present an attrac- stranded linear DNA genome is surrounded by an
tive option until a vaccine becomes available. icosahedral nucleocapsid, enveloped by the viral
This review focuses on the current use and tegument proteins (including pp65, which harbors a
dominant cytotoxic T lymphocyte epitope) and lipid
potential of antibody therapies to treat CMV
bilayer, which is studded with at least 20 glycopro-
disease. teins. The fusogenic glycoprotein gB binds some cell
surface receptors and appears to be immunodomi-
nant, but neutralizing antibodies recognizing this
protein only block viral entry into broblasts. The
THE INFECTION gH/L dimer appears to bind specic receptors and
potentiate gB-membrane fusion. When occurring as a
Virus Structure and Life Cycle gH/L/O complex, it is also involved in entry into
broblasts, a process that appears to occur via direct
CMV is a ubiquitous beta herpesvirus type 5, membrane fusion. In contrast, the gH/L/UL128-131
related to the herpes simplex viruses, Epstein- pentameric complex is required for entry into epi-
thelial and endothelial cells, a process mediated by
Barr virus, and varicella zoster, the causative
endocytosis and low-pH fusion. The gM/N complex is
agent of chicken pox. Specialized strains the most abundant on the virion surface, initiating
appear to have coevolved with and become adsorption to cells by binding heparin sulfate
restricted to their respective mammalian hosts. proteoglycans. The gN may be heavily glycosylated
The human CMV genome is among the largest to shield the virion against antibody recognition.
doi:10.1128/microbiolspec.AID-0009-2013.f1
and most complex animal virus genomes, with
a large 235-kb double-stranded linear DNA
genome encoding 165 open reading frames molecules of glycoprotein B (gB), in which
(8). The genome and some viral mRNAs are each 160-kDa monomer is cleaved and then
surrounded by an icosahedral nucleocapsid, joined by a disulfide bond to include a 115-kDa
enveloped by the viral tegument proteins and surface unit and a 55-kDa transmembrane
a lipid bilayer, which is studded with at least component. The gB has been reported to
20 different glycoproteins forming a number of bind epidermal growth factor receptors and
complexes. The whole unit spans 200 to 300 nm integrins and is a type 3 fusogen, which
in diameter (Fig. 1). CMV can spread directly initiates membrane fusion. (ii) A disulfide-
from cell to cell but destroys infected cells by linked heterodimer formed by the gM and gN
lytic replication, resulting in a high serum viral proteins is the most abundant complex and
load which correlates with end-organ disease. may initiate adsorption to cells by binding
CMV disease is monitored by quantitative PCR heparin sulfate proteoglycans. (iii) The gH/gL/
to detect CMV DNA in leukocytes or serum gO disulfide-linked trimer is notable, as all
and detection of the tegument structural herpes viruses use a gH/gL complex to me-
protein pp65 in peripheral blood leukocytes diate fusion of the viral envelope with the
by immunostaining. cellular membrane, although the CMV gH/gL
The virion contains several major glycopro- cocrystal structure does not resemble a typical
tein complexes at the cell surface. (i) Two fusogen (9). The gH is an 86-kDa glycoprotein,
276 ACQUAYE-SEEDAH ET AL.

present in nuclear and cytoplasmic mem-


branes of infected cells and in the viral enve-
lope. While gH binds the v3 integrin, the
32-kDa gL forms extensive contacts with gH
and is required for gH to fold and localize
properly at the cell surface. The gO does not
appear to be crucial for membrane fusion but
appears to chaperone gH/gL incorporation
into new virions (10). (iv) A second gH/gL
complex combined with the trimeric products
of UL128, 130, and 131 (UL128-131) has recently
been characterized in greater detail, revealing FIGURE 2 Human CMV life cycle. (1) The virion binds
critical roles during invasion of endothelial to cells via the gB and gH/L/UL128-131 glycoproteins
and epithelial cells (11). and specic cellular receptors, followed by direct
In vivo, the virus infects a broad spectrum membrane fusion (broblasts) or endocytosis and
of cells, including macrophages, fibroblasts, low-pH-mediated membrane fusion (endothelial and
epithelial cells). (2) The virion contents are released
epithelial, neural, and muscle cells, allowing it into the cytoplasm, allowing the nucleocapsid to
to affect multiple organ systems and cause a translocate to the nucleus for DNA replication and
wide range of diseases, including pneumonia, transcription and packaged viral transcripts to be
gastrointestinal disease, retinitis, hepatitis, and directly translated. (3) Transcripts are translated in
encephalitis. CMV entry into cells is a complex the cytoplasm, followed by processing in the endo-
plasmic reticulum and Golgi body. (4) Viral DNA and
process initiated by adsorption onto the cell proteins are assembled and enveloped to create new
surface, followed by specialized invasion strat- virions, followed by (5) release into the extracellular
egies for different cell types (Fig. 2). In surroundings or directly into another cell. (6) During
particular, fibroblast entry is mediated by the this process, fragments of viral proteins are com-
fusogenic gB and potentiated by the gH/gL/gO bined with host MHC class I in the endoplasmic
reticulum for presentation on the cell surface.
complex, possibly involving 1 integrins and Antibodies can directly affect the cellular attachment
epidermal growth factor receptors, followed and internalization steps to prevent primary infec-
by direct fusion of the viral envelope with the tion, while T-cell recognition of viral pMHC com-
cell membrane (11, 12). In contrast, infection of plexes is crucial for identifying and lysing infected
endothelial cells, epithelial cells, and macro- cells. doi:10.1128/microbiolspec.AID-0009-2013.f2
phages requires the gH/gL/UL128-131 pen-
tamer. When associated with the cell surface,
Virus-Immune Dtente
this complex triggers endocytosis, followed by
a gB- and gH-dependent fusion of the viral Primary infection of immunocompetent CMV-
membrane with the endosomal membrane (13, individuals rarely leads to active disease but
14). In addition to the gH/gL/UL128-131 pen- instead leads to a lifelong latent infection,
tamer, viral entry into these cells appears to characterized by periodic reactivation and
require a different gB conformation to trigger secondary infection. During latency, multiple
fusion (12). The ability of CMV strains to copies of the CMV genome appear to reside in
invade fibroblasts has been used historically a small number of mononuclear cells. Second-
to characterize strains, vaccines, and anti- ary or superinfection with a new CMV strain
bodies. However, it has been recognized that can be monitored by serum viral titer, a
during laboratory culture, strains such as correlate for disease severity. Maintenance of
AD169 and the Towne vaccine strain readily latency requires continual monitoring by the
acquire mutations in the UL128-131 genes that host immune system, such that 5 to 10% of all
restrict their tropism to fibroblasts, raising CD8+ and CD4+ T cells recognize CMV
concerns over their clinical relevance. peptides, a number which increases to 30%
CHAPTER 16 Immunotherapeutic Approaches to Prevent CMV Disease 277

in the elderly. To prevent disease in suscepti- number of B cells and that the epitope may be
ble populations, current treatments include difficult to access and/or poorly immunogenic
(i) antiviral agents to prevent replication (19). More recently, sequencing of gB-specific
(primarily ganciclovir) in the first 3 months memory B cells isolated from seven seropos-
posttransplantation, which are associated itive individuals showed that >90% of the
with significant toxicity and not approved corresponding antibodies are nonneutralizing.
for use in pregnant women; (ii) polyvalent This work also identified two additional,
immunoglobulin preparations enriched in potently neutralizing gB epitopes: AD-4 and
antibodies against CMV (CMV-IVIG); and AD-5. Antibodies binding these epitopes do
(iii) adoptive immunotherapy by ex vivo not block viral attachment but neutralize a
expansion of CMV-specific T cells, which postadsorption fusion event (20). Collectively,
are then reinfused into the patient. Antivirals neutralizing anti-gB antibodies affect early
and CMV-IVIG target viral proteins involved events of infection and have been shown to
in replication to restrict viral dissemination prevent viral penetration of fibroblasts and
and limit disease severity in CMV- pregnant cell-to-cell spread. However, while gB is key
women and solid organ recipients, while T- for viral entry, it is not essential for viral
cell therapy has demonstrated efficacy in attachment, assembly, or egress, and thus, anti-
maintaining latency after stem cell transplan- gB antibodies may not represent the most
tation but has not been standardized for critical components of the protective humoral
routine use. immune response.
While the magnitude of their role has been More recently, analysis with cell surface-
debated, antibodies appear to limit viral loads expressed and purified CMV antigens sug-
by inhibiting aspects of cellular invasion. An gested that the major neutralizing antibody
early analysis purified antibodies from the response blocking entry into epithelial cells is
serum of seropositive individuals by immuno- directed against the gH/gL complex and not
adsorption onto recombinant vaccinia virus gB (21). Anti-gH/L neutralizing antibodies
expressing gB. The purified antibodies were isolated from patient memory B cells either
then used to neutralize CMV strain AD169 neutralized viral infection on all cell types
invasion into fibroblasts, concluding that 40 to tested with moderate 50% inhibitory concen-
70% of neutralizing antibodies target gB (15, tration (IC50) values or potently protected
16). Three antigenic domains were later de- epithelial, endothelial, and myeloid cells with
fined: AD-1, a conformational epitope located low IC50 values, between 20 and 200 pM, but
between residues 552 and 635; AD-2, a linear did not protect fibroblasts. Competition assays
epitope between residues 69 and 78; and AD-3, showed that antibodies in the first group
another linear epitope at the carboxy terminus, recognized at least three distinct neutralizing
between residues 783 and 906 (17). AD-1 is sites on gB and two on gH, while antibodies in
involved in gB oligomerization and is immuno- the second group recognized at least seven
dominant, with antibodies binding this epitope distinct sites on the gH/L/UL128-131 complex,
present in all tested individuals. Interestingly, some of which span the interfaces between
these responses are comprised of neutralizing adjacent proteins (21, 22). Even short peptides
and nonneutralizing antibodies competing for from UL128 and UL130 elicit high neutralizing
the same site, which may assist in viral immune titers in rabbits which are able to block viral
evasion (18). Antibodies recognizing AD-2 are entry into epithelial cells from the mucosa
only present in about half of individuals but are (23). This is consistent with other reports that
potently neutralizing. Sequencing has revealed anti-gH antibodies, which are present at high
similar variable region usage for antibodies levels during acute infection, block cell-to-cell
binding AD-2 within a single individual, spread (24) while those against UL128 and
suggesting that they are derived from a small UL130 specifically inhibit endothelial cell
278 ACQUAYE-SEEDAH ET AL.

invasion (14, 23, 25). Less potently neutralizing tion. Several immunodominant T-cell epitopes
human monoclonal antibodies recognizing have been identified, including the tegument
other viral glycoproteins (gB, gH, or gM/gN phosphoproteins 65 (pp65, the most abundant
complex) have been isolated which neutral- protein in the tegument) and 150 (pp150) and
ized infection of epithelial/endothelial cells as the immediate-early protein 1 (IE-1) which are
well as fibroblasts (22), although their epitopes conserved across strains (34, 35). Transcripts
are less well characterized. It has been of pp65 are packaged within virions and
suggested that the extensive glycosylation on translated directly after cellular invasion;
gN may serve to shield these glycoproteins thus their display provides an early marker
from neutralizing antibodies (26). for infection. The dominant effector cells in
Cellular immune responses to CMV are adoptive immunotherapy respond to these
mediated primarily by the cytolytic activity of three peptides (29, 30). One clinical study
CD8+ cells, but these require CD4+ help to adoptively transferred pp65 or IE-1-specific
persist (27). After a primary infection, latency CD8+ T cells, observing a dramatic reduction
is maintained by an array of immune evasion of viral load in all 9 patients (36). In contrast, in
tactics, including down-regulation and degra- vitro experiments using peptides derived from
dation of antigen-presenting major histocom- 213 open reading frames demonstrated T-cell
patibility complex (MHC) molecules to avoid responses to more than 70% of expressed
recognition by cytotoxic T cells (28). The role genes (37). This raises the question are the
of cellular immunity has been demonstrated dominant epitopes we observe in vivo actually
convincingly by adoptive transfer studies in critical for CMV suppression or an artifact of
which donor-derived, CMV-specific CD8+- viral immune evasion tactics?
T-cell lines or clones were first isolated by Looking at the other half of the interaction,
coculture with virus or antigen-pulsed den- analyses of the CD8+ TCR sequence diver-
dritic cells, expanded in vitro, and finally, sity has identified a large number of public
infused into HLA-matched hematopoietic TCRs, comprised of the same germ line
stem cell transfer patients. This procedure variable regions and used by different individ-
restored antigen-specific immunity and pre- uals to recognize these dominant CMV
vented CMV-associated clinical outcomes (29, peptides when presented by HLA (38). The
30); however, it is time-intensive and labori- cells displaying public TCRs recognize virally
ous, requiring a total of 4 to 6 weeks for cell infected cells with greater avidity and exhibit
line expansion (31). Recent efforts to stream- a more terminally differentiated phenotype
line the process have included direct isolation (39). Recently, tetramers comprised of the
of CMV-specific T cells via HLA tetramers pp65495503 peptide in complex with the
prior to in vitro expansion and T-cell trans- human HLA A*0201 were used to isolate
fection with genes encoding CMV-specific human CD8 + T cells, which were then
TCRs (32). Several T-cell lines recognizing subjected to single-cell sequencing of the
pp65 peptides in complex with multiple HLA TCR alpha and beta chains. Public and
restrictions have been shown to be effective in nonpublic TCR sequences were observed,
vitro, in terms of cytokine secretion and target and TCR sequence diversity was correlated
cell lysis (33), but it remains to be seen whether with serum antibody titers as an indirect
these can be transferred to the clinic. measure of viral titer and disease propensity.
As these procedures evolve, a key question The primary conclusion of this study was that
becomes what T-cell specificities are re- the diversity, but not the magnitude, of the
quired for protection? Careful analysis of cellular response inversely correlated with the
the molecular details of this recognition serum anti-CMV titer, which is itself a marker
event requires identification of the peptides for increased viral load (40). These data
and corresponding TCRs involved in protec- suggest that immunotherapeutic approaches
CHAPTER 16 Immunotherapeutic Approaches to Prevent CMV Disease 279

based on a few immunodominant peptides or blast but not epithelial cell invasion. More-
public TCRs will be less effective than one recent efforts to improve live attenuated
based on a larger set of interactions. vaccines include coadministration with inter-
leukin-12 as an adjuvant (46) and chime-
rization with the unattenuated Toledo strain.
Vaccine Development
The moderate protective effects observed in
A vaccine to prevent CMV disease has been a live attenuated CMV vaccine trials, coupled
top priority since 1999, and despite consider- with concerns that heavily attenuated strains
able effort, none are likely to receive regulato- are less relevant (47) and pose regulatory
ry approval soon (5, 6). Challenges have concerns, have slowed development of these
included the lack of a clear serological corre- vaccines.
late of protection, appropriate animal models, The second most developed vaccine strat-
and target population. To maximize the clin- egy is a subunit vaccine, based on the obser-
ical benefit, it has been suggested that all vation that gB dominates the humoral immune
seronegative young women, CMV- stem cell response. While prior trials focused on safety
donors, and solid organ transplant recipients and immunogenicity, in 2009, a subunit vac-
be immunized to protect future children and cine was shown for the first time to be effective
transplant recipients, respectively. Over the against primary maternal CMV infection and,
last 50 years, two vaccine approaches have later, in transplant recipients. This vaccine is
been extensively evaluated in clinical trials: comprised of a squalene-water emulsion of an
live attenuated vaccine and gB-based subunit engineered gB with the novel proprietary
vaccines, with numerous other DNA, vectored adjuvant MF59. The gB protein is modified
virus, and peptide formulations in develop- to remove the internal cleavage site and
ment (for a review, see reference 41). introduce a stop codon before the transmem-
Two live attenuated strains have been brane region, resulting in a soluble product
developed and evaluated in vaccine trials, the expressed in CHO cells. In a phase II placebo-
laboratory-adapted AD169 and Townes controlled efficacy trial, 18 of 225 women
strains. The first clinical trials with Townes, receiving the vaccine versus 31 of 216 receiving
originally a neonatal clinical isolate, demon- placebo acquired primary CMV infection
strated that it could elicit both neutralizing during a 1-year period, for overall vaccine
antibodies and T-cell responses lasting up to 10 efficacy of 50%. Peak levels of anti-gB were
years, with a good safety profile. It was about fivefold higher than in naturally sero-
subsequently shown to reduce viral load and positive individuals but did not induce anti-
mitigate CMV disease, but not to prevent bodies preventing endothelial/epithelial cell
primary or secondary infection, when admin- invasion (48). A subsequent phase II random-
istered to kidney transplant patients at least ized study with kidney and liver transplant
8 weeks prior to transplantation (42, 43, 44). patients showed that anti-gB titers correlated
When administered to seronegative mothers, inversely with the duration of viremia (49).
Townes showed no efficacy in reducing This adjuvanted gB subunit vaccine remains
transmission from children shedding CMV the most promising candidate, but additional
virions (45). In a challenge study with healthy trials are required for licensure. A variety of
volunteers, vaccinated individuals were additional vaccine designs have been evaluat-
protected more than seronegative individuals ed, including recombinant canary pox and
but were still 5 to 10 times more likely to peptide vaccines designed to elicit strong
become ill than naturally seropositive individ- cellular responses against known immuno-
uals. This may be a result of Townes deletions dominant peptides (50).
in the UL128-131 region, meaning that the While still in early stages, new approaches
vaccine can induce antibodies blocking fibro- based on the pentameric gH/L/128-131
280 ACQUAYE-SEEDAH ET AL.

complexnotably absent from the AD169 and to-lot variability is observed (56). While the
Townes vaccine strainsappear promising. enzyme-linked immunosorbent assay titer is a
This complex is essential for viral invasion of primary characteristic, it does not always track
endothelial/epithelial cells, and clinical stud- with the neutralization titer and in some cases,
ies have determined that antibodies recogniz- standard IVIG can have a higher neutralizing
ing the UL128-131 complex appear within 2 to titer than CMV-IVIG (57). This activity typi-
4 weeks after infection, last for at least a year, cally protects endothelial and epithelial cells
and are stronger than responses to gH (25). more potently than fibroblasts. A recent report
Moreover, the bulk of the neutralizing re- suggested that the bulk of the neutralizing
sponse in CMV-IVIG is now thought to be response in CMV-IVIG (Cytogam) is due to
mediated by antibodies binding gH/L/UL128- antibodies recognizing the gH/L/128-131
131 (21), and these have been proposed as a pentamer with little role for anti-gB antibodies
correlate of protection (51, 52). When the (21). Antibody isotype may also be a key
complex is restored to the AD169 vaccine criterion, as IgG3 antibodies have been found
strain, it induced 10-fold-higher neutralizing to be 10-fold more potently neutralizing than
antibody titers than AD169 or the gB subunit the IgG1 isotype (58). Collectively, the anti-
vaccine in rabbits and monkeys (53). This bodies in CMV-IVIG are thought to act by
effect has been shown consistently across neutralizing the virus ability to infect both
several animal models, as a modified vaccinia fibroblast and endothelial/epithelial cells (59),
Ankara virus expressing all five rhesus ho- reducing inflammation, and/or reducing cyto-
mologs developed neutralizing antibody titers kine-mediated immune responses, although
similar to natural infection and exhibited these latter mechanisms have not been well
reduced plasma loads after postvaccination characterized (60).
challenge in monkeys (54). Finally, an alpha- CMV-IVIG was first licensed in 1991 and is
virus replicon particle coexpressing only gH/gL well tolerated with no severe side effects. It has
produced broadly cross-reactive complement- been used for prophylaxis of CMV disease
independent neutralizing antibodies in mice at associated with at-risk patients receiving solid
higher titers than those with gB (55). organ and stem cell transplants, typically,
CMV- recipients with CMV+ donors. To im-
prove efficacy, it is often combined with
antiviral therapy. However, the results of
THERAPEUTIC ANTIBODIES
clinical trials evaluating its use have been
variable, most likely due to lot-to-lot variability,
Polyclonal CMV-IVIG
various doses and schedules, and mixed patient
The available antibody-based anti-CMV ther- populations with different risk profiles. While
apies are polyvalent immunoglobulin prepa- early studies suggested that prophylactic
rations enriched in antibodies against CMV CMV-IVIG could reduce the severity of CMV
(CMV-IVIG) (Table 2) and were of particular infection in liver transplant patients (61),
interest before antivirals became widely avail- others have suggested that it is less effective
able. The CMV-IG is made by pooling serum than combinations of antivirals (62) and that
from 1,000 donors screened for high anti- IVIG-antiviral combination therapy could ac-
CMV serum titers and purifying the antibodies tually increase both survival and rejection risk
by cold ethanol precipitation, followed by a (63). With high-risk kidney transplant re-
low-pH or solvent-detergent virus inactivation cipients, IVIG is associated with an excellent
step. This process results in a product with 50 1-year survival rate (64) and combination
mg of immunoglobulin G (IgG)/ml and anti-gB therapy with low-dose valaciclovir was effica-
titers of 1:400,000, two- to fourfold higher cious in preventing CMV disease (65). A meta-
than in seropositive individuals, although lot- analysis pooling 11 trials with almost 700 solid
CHAPTER 16 Immunotherapeutic Approaches to Prevent CMV Disease 281

TABLE 2 Antibody-based CMV therapeutics


Therapeutic Target Indication Development stage Source or reference

Human immune sera; CMV envelope Transplant Approved www.cytogam.com/


polyclonal (Cytogam; glycoproteins recipients,
CSL/Behring, MedImmune) (block virus entry) congenital CMV
Human immune sera; CMV envelope Transplant Approved http://paviour.org/
polyclonal (Cytotect; glycoproteins recipients, cytotect.htm
Biotests AG) (block virus entry) congenital CMV
Human monoclonal CMV envelope CMV retinitis; Phase I/II trials 85, 87
antibody (MSL109; PDL) glycoprotein H stem cell transplant completed;
(block virus entry) recipients no further
progress
Human monoclonal CMV envelope CMV retinitis Phase I/II 83, 92
antibody MSL109 in glycoprotein H
combination with (block virus entry)
ganciclovir
Human IgG1 monoclonal gB epitope CMV retinitis in HIV+ Phase II 81; US5043281
antibody C23/regavirumab individuals, bone (1990)
(Teijin Ltd.) marrow recipients
Human neutralizing CMV Preclinical 22, 88; US7947274
monoclonal antibodies gUL131A-128 (2011)
(Humabs, LLC) trimer
TCR-like antibody CMV peptide p64 Preclinical 90
or p65 displayed
by MHC
TCR anti CD3-bispecic CMV-infected Preclinical 91
antibody cells
Humanized antibodies gB, gH epitopes CMV infection in Preclinical 24, 80
(Scotgen Biopharm) immunocompromised
patients
Combination of Human CMV Preclinical US5126130 (1987)
monoclonal glycoprotein A
antibodies (Childrens
Hospital, Inc.)

organ transplant recipients found a positive IVIG therapy is approved, but is not standard
effect of prophylactic CMV-IVIG on recip- treatment, for transplant patients.
ients: improved total survival and moderately The presence of maternal antibodies to
reduced CMV disease and CMV-associated CMV reduces the risk of fetal infection (3),
deaths (66). CMV-IVIG use in stem cell indicating the potential for antibodies to
transplant patients is less promising. A multi- prevent and/or treat congenital CMV infec-
center, randomized, double-blind, dose-effect, tion. Passive immunization studies with preg-
placebo-controlled study of 200 stem cell nant guinea pigs have demonstrated that
transplants concluded that the use of immu- antibodies to the virus or gB reduce the fetal
noglobulin therapy did not significantly reduce infection rate and enhance fetal survival (69,
CMV infection and disease (67). A second 70). Furthermore, adoptive transfer of B
meta-analysis combining 30 trials with 4,223 memory cells was protective in T- and B-cell-
stem cell transplant patients observed no deficient mice (71). Subsequently, several
differences in mortality, infections of any nonrandomized human trials using CMV-
kind, CMV infections, or graft-versus-host IVIG to prevent CMV infection in utero have
disease (68). As a result of this variability, been performed. In one, 181 pregnant women
282 ACQUAYE-SEEDAH ET AL.

with confirmed primary CMV infection were their biochemical and biophysical properties
divided into a treatment group (women who engineered to meet specific design targets; and
had amniocentesis to confirm fetal infection) since they are purified from monoclonal
and a prevention group (those who did not cultures, minimal lot-to-lot variability (78).
undergo amniocentesis) (72). The treatment Most efforts in developing CMV-neutralizing
group included 55 women, 31 who received monoclonal antibodies have targeted the gH
Cytotect IVIG, resulting in one infected new- viral glycoprotein, as low anti-gH titer is a
born (3%), and 14 who received no treatment, risk factor for CMV disease (79) (Table 2).
resulting in seven infected newborns (50%). Phase I clinical trials of several anti-gB and
The prevention group included 37 women anti-gH human monoclonal antibodies have
receiving Cytotect IVIG, resulting in 6 infected demonstrated a pharmacokinetic profile simi-
newborns (16%), and 47 who did not receive lar to that of human IgG1 and lack of immuno-
treatment, resulting in 19 infected newborns genicity, supporting their safe use in passive
(40%). More recently, a case-controlled study immunotherapy applications (80, 81, 82). One
compared 32 children with hearing and/or anti-gH IgG1 human monoclonal antibody
psychomotor deficits with 32 healthy, age- and isolated from the spleen of a CMV-seropositive
gestation-matched control children. The individual was able to block in vitro infection
mothers of both groups had confirmed or of multiple cell types by laboratory and clinical
probable primary CMV infection at <20 strains of CMV, with IC50 values between 0.03
weeks of gestation. The only risk factor for and 1.02 g/ml (83, 84). As this antibody also
these children was whether the mother had effectively treated CMV retinitis when com-
received CMV-IVIG after infection: of the case bined with ganciclovir in rat models, it was
patients, 4/32 received IVIG, while 27/32 evaluated in a series of clinical trials as MSL-
control mothers received IVIG (73). While 109 (Sevirumab/ Protovir; Protein Design
promising, larger studies are needed to draw a Labs), but unfortunately, it has not shown
clear conclusion, especially for an expensive sufficient efficacy.
intervention that requires screening for sero- A randomized placebo-controlled human
logic confirmation of infection followed by trial to assess the efficacy of MSL-109 to delay
monthly CMV-IVIG administration (74). progression of CMV retinitis in AIDS patients
was halted after it failed to slow progression of
the blinding eye disease (85). Indeed, the MSL-
Monoclonal Antibodies
109 treatment group had a higher mortality
The apparent protective role of antibodies rate than the placebo group. Subsequently,
against congenital CMV suggests that these serum CMV titers were associated with a
polyclonal immune responses could be higher risk of mortality in AIDS patients with
deconvoluted to a discrete number of highly CMV retinitis, implying that MSL-109 was
protective epitopes. These could, in principle, ineffective in clearing plasma CMV (86).
be targeted with high efficacy by a monoclonal Similarly, stem cell transplant recipients with
or oligoclonal antibody preparation (75, 76). CMV+ donors or recipients received MSL-109
This perspective is supported by the early biweekly in a randomized, double-blind study.
characterization of human monoclonal anti- The MSL-109 treatment group showed no
body C23 which neutralized the ability of difference in survival or pp65 antigenemia
AD169 and clinical CMV isolates to infect versus the placebo groups (87). Recently, it has
lung fibroblasts 1,000 times more potently been suggested that the MSL-109 failure may
than human IVIG (77). Monoclonal antibodies have been due to the rapid generation of
have the additional benefits of being very well nongenetic escape variants. It appears that
characterized, with epitopes, binding affini- infected cells present gH on their surface
ties, and neutralizing mechanisms described; during viral replication. The gH is available
CHAPTER 16 Immunotherapeutic Approaches to Prevent CMV Disease 283

to bind and endocytose circulating MSL-109, been explored for HIV and cancer treatment
and the gH/MSL-109 complex is then incor- (89). An antibody that could be used for this
porated into the viral envelope with the MSL- approach has been isolated from a large human
109 Fc domain exposed. The gH/L complex is Fab phage display library that recognizes HLA
no longer available to bind epithelial/endothe- A2/pp65495503 with a 300-nM affinity and is
lial receptors, but the Fc domain of MSL-109 is able to specifically detect peptide on the
available to bind Fc receptors, altering the surface of fibroblasts in vitro (90). A similar
cellular trophism of the virus (84). approach formed a bispecific antibody by
Recently, a novel method was used to chemically linking OKT3 (an FDA-approved
characterize the repertoire of CMV-reactive anti-CD3 antibody which binds and activates T
antibodies in CMV-immune donors. Human B cells) to CMV-IVIG polyclonal anti-CMV
lymphocytes from CMV-seropositive donors antibodies. After incubation with CMV-in-
were immortalized with Epstein-Barr virus, fected fibroblasts, specific lysis of CMV-in-
followed by screening for CMV neutralization. fected cells was demonstrated in vitro (80% for
Of the 1,664 clones screened, 29 neutralized the test versus <20% for the control) (91).
CMV infectivity. Of the four further charac- Additional reports documenting CMV-neutral-
terized, two bound gB AD-2, two bound gH, izing human monoclonal antibodies (Table 2)
and all were able to neutralize lab strain AD169 reflect the breadth of efforts to development of
or a clinical isolate in both fibroblast and human/humanized monoclonal antibodies to
endothelial invasion assays. Importantly, these neutralization CMV.
monoclonal antibodies were 20-fold more
potent than the Cytotect IVIG, supporting
development of antibody cocktails over IVIG FUTURE CHALLENGES
(88). As described above, it appears that
additional potently neutralizing epitopes are Based on the high rates of CMV infection in
present on the pentameric gH/gL/UL128-131 adults and the large percentage of CMV-
complex and may present promising targets specific T cells in seropositive individuals,
for passive immunotherapy (21, 22). Other less preventing infection appears to be an unattain-
potently neutralizing human monoclonal an- able goal. In the absence of a vaccine, antibody
tibodies isolated from memory B cells recog- therapies to prevent or control disease in
nized viral glycoprotein gB, gH, or gM/gN and susceptible populations may be the best solu-
were shown to neutralize infection of both tion. Here, there is strong evidence that adop-
epithelial/endothelial cells and fibroblasts tive transfer of cytotoxic T cells with CMV
(22). specificity is able to control secondary disease
Apart from traditional passive immuno- in stem cell transplant patients, while admin-
therapies relying on a single antibody to istration of hyperimmune IVIG appears effec-
sequester a single epitope, new paradigms are tive in some solid organ transplant patients and
emerging which could be applied to prevent is promising in seronegative pregnant women.
CMV replication or lyse CMV-infected cells. In conjunction with recent insights into the
For instance, adoptive immunotherapy has function of CMV glycoprotein complexes,
also shown that T cells recognizing the pp65, further elucidation of protective B- and T-cell
pp150, and I-E1 peptides form the dominant epitopes present in the UL128-131 trimer and
population of effector cells and uniquely associated gH/gL pentamer is expected to
identify CMV+ cells early in infection (34, 35). contribute to design of immunotherapeutics
Antibodies mimicking the specificity of public with greater specificity and potency. Ulti-
TCRs recognizing these peptides could deliver mately, potent cocktails of high-affinity mono-
a cytotoxic payload or direct cytotoxic lym- clonal antibodies binding neutralizing epitopes
phocytes to these cells, approaches which have on the virus and TCR-like antibodies to target
284 ACQUAYE-SEEDAH ET AL.

infected cells may prove effective at sup- 9. Vanarsdall AL, Ryckman BJ, Chase MC,
pressing CMV replication. Johnson DC. 2008. Human cytomegalovirus
glycoproteins gB and gH/gL mediate epithelial
cell-cell fusion when expressed either in cis or in
trans. J Virol 82:1183711850.
ACKNOWLEDGMENTS 10. Ryckman BJ, Chase MC, Johnson DC. 2010.
This work was supported by generous grants Human cytomegalovirus TR strain glycoprotein O
acts as a chaperone promoting gH/gL incorpo-
from NSF (DBI-0964137) and NIH (NIGMS ration into virions but is not present in virions.
R01GM095638) to J.A.M. J Virol 84:25972609.
Conflicts of interest: We declare no conflict. 11. Ryckman BJ, Chase MC, Johnson DC. 2008.
HCMV gH/gL/UL128-131 interferes with virus
entry into epithelial cells: evidence for cell type-
CITATION specific receptors. Proc Natl Acad Sci USA 105:
1411814123.
Acquaye-Seedah E, Frye ZP, Maynard JA. 12. Vanarsdall AL, Chase MC, Johnson DC. 2011.
2013. Immunotherapeutic approaches to pre- Human cytomegalovirus glycoprotein gO com-
vent cytomegalovirus-mediated disease. plexes with gH/gL, promoting interference with
viral entry into human fibroblasts but not entry
Microbiol Spectrum 2(1):AID-0009-2013.
into epithelial cells. J Virol 85:1163811645.
13. Wang D, Shenk T. 2005. Human cytomegalovirus
virion protein complex required for epithelial and
REFERENCES endothelial cell tropism. Proc Natl Acad Sci USA
1. Craig JM, Macauley JC, Weller TH, Wirth P. 102:1815318158.
1957. Isolation of intranuclear inclusion producing 14. Wang D, Yu QC, Schroer J, Murphy E, Shenk T.
agents from infants with illnesses resembling 2007. Human cytomegalovirus uses two distinct
cytomegalic inclusion disease. Proc Soc Exp Biol pathways to enter retinal pigmented epithelial
Med 94:412. cells. Proc Natl Acad Sci USA 104:2003720042.
2. Johnson J, Anderson B, Pass RF. 2012. Preven- 15. Britt WJ, Vugler L, Butfiloski EJ, Stephens EB.
tion of maternal and congenital cytomegalovirus 1990. Cell surface expression of human cytomeg-
infection. Clin Obstet Gynecol 55:521530. alovirus (HCMV) gp55-116 (gB): use of HCMV-
3. Fowler KB, Stagno S, Pass RF. 2003. Maternal recombinant vaccinia virus-infected cells in anal-
immunity and prevention of congenital cytomeg- ysis of the human neutralizing antibody response.
alovirus infection. JAMA 289:10081011. J Virol 64:10791085.
4. Smithey MJ, Li G, Venturi V, Davenport MP, 16. Marshall GS, Rabalais GP, Stout GG, Waldeyer
Nikolich-Zugich J. 2012. Lifelong persistent viral SL. 1992. Antibodies to recombinant-derived gly-
infection alters the naive T cell pool, impairing coprotein B after natural human cytomegalovirus
CD8 T cell immunity in late life. J Immunol 189: infection correlate with neutralizing activity.
53565366. J Infect Dis 165:381384.
5. Arvin AM, Fast P, Myers M, Plotkin S, 17. Navarro D, Lennette E, Tugizov S, Pereira L.
Rabinovich R. 2004. Vaccine development to 1997. Humoral immune response to functional
prevent cytomegalovirus disease: report from the regions of human cytomegalovirus glycoprotein B.
National Vaccine Advisory Committee. Clin Infect J Med Virol 52:451459.
Dis 39:233239. 18. Speckner A, Glykofrydes D, Ohlin M, Mach
6. Khanna R, Diamond DJ. 2006. Human cyto- M. 1999. Antigenic domain 1 of human cytomeg-
megalovirus vaccine: time to look for alternative alovirus glycoprotein B induces a multitude of
options. Trends Mol Med 12:2633. different antibodies which, when combined,
7. Marasco WA, Sui J. 2007. The growth and results in incomplete virus neutralization. J Gen
potential of human antiviral monoclonal antibody Virol 80(Pt 8):21832191.
therapeutics. Nat Biotechnol 25:14211434. 19. Schrader JW, McLean GR. 2007. Location,
8. Dolan A, Cunningham C, Hector RD, Hassan- location, timing: analysis of cytomegalovirus
Walker AF, Lee L, Addison C, Dargan DJ, epitopes for neutralizing antibodies. Immunol
McGeoch DJ, Gatherer D, Emery VC, Griffiths Lett 112:5860.
PD, Sinzger C, McSharry BP, Wilkinson GW, 20. Potzsch S, Spindler N, Wiegers AK, Fisch T,
Davison AJ. 2004. Genetic content of wild-type Rucker P, Sticht H, Grieb N, Baroti T, Weisel
human cytomegalovirus. J Gen Virol 85:1301 F, Stamminger T, Martin-Parras L, Mach M,
1312. Winkler TH. 2011. B cell repertoire analysis
CHAPTER 16 Immunotherapeutic Approaches to Prevent CMV Disease 285

identifies new antigenic domains on glycopro- Reconstitution of cellular immunity against cyto-
tein B of human cytomegalovirus which are megalovirus in recipients of allogeneic bone
target of neutralizing antibodies. PLoS Pathog marrow by transfer of T-cell clones from the
7:e1002172. donor. N Engl J Med 333:10381044.
21. Fouts AE, Chan P, Stephan JP, Vandlen R, 31. Sellar RS, Peggs KS. 2012. Therapeutic strategies
Feierbach B. 2012. Antibodies against the gH/gL/ for the prevention and treatment of cytomegalo-
UL128/UL130/UL131 complex comprise the virus infection. Expert Opin Biol Ther 12:11611172.
majority of the anti-cytomegalovirus (anti-CMV) 32. Einsele H, Hebart H. 2004. CMV-specific
neutralizing antibody response in CMV hyperim- immunotherapy. Hum Immunol 65:558564.
mune globulin. J Virol 86:74447447. 33. Schub A, Schuster IG, Hammerschmidt W,
22. Macagno A, Bernasconi NL, Vanzetta F, Dander Moosmann A. 2009. CMV-specific TCR-
E, Sarasini A, Revello MG, Gerna G, Sallusto F, transgenic T cells for immunotherapy. J Immunol
Lanzavecchia A. 2010. Isolation of human mono- 183:68196830.
clonal antibodies that potently neutralize human 34. Greenberg PD, Reusser P, Goodrich JM, Riddell
cytomegalovirus infection by targeting different SR. 1991. Development of a treatment regimen for
epitopes on the gH/gL/UL128-131A complex. human cytomegalovirus (CMV) infection in bone
J Virol 84:10051013. marrow transplantation recipients by adoptive
23. Saccoccio FM, Sauer AL, Cui X, Armstrong AE, transfer of donor-derived CMV-specific T cell
el Habib SE, Johnson DC, Ryckman BJ, clones expanded in vitro. Ann N Y Acad Sci 636:
Klingelhutz AJ, Adler SP, McVoy MA. 2011. 184195.
Peptides from cytomegalovirus UL130 and UL131 35. Slezak SL, Bettinotti M, Selleri S, Adams S,
proteins induce high titer antibodies that block Marincola FM, Stroncek DF. 2007. CMV pp65
viral entry into mucosal epithelial cells. Vaccine and IE-1 T cell epitopes recognized by healthy
29:27052711. subjects. J Transl Med 5:17.
24. Hamilton AA, Manuel DM, Grundy JE, Turner 36. Cobbold M, Khan N, Pourgheysari B, Tauro S,
AJ, King SI, Adair JR, White P, Carr FJ, Harris McDonald D, Osman H, Assenmacher M,
WJ. 1997. A humanized antibody against human Billingham L, Steward C, Crawley C, Olavarria
cytomegalovirus (CMV) gpUL75 (gH) for prophy- E, Goldman J, Chakraverty R, Mahendra P,
laxis or treatment of CMV infections. J Infect Dis Craddock C, Moss PA. 2005. Adoptive transfer of
176:5968. cytomegalovirus-specific CTL to stem cell trans-
25. Genini E, Percivalle E, Sarasini A, Revello MG, plant patients after selection by HLA-peptide
Baldanti F, Gerna G. 2011. Serum antibody tetramers. J Exp Med 202:379386.
response to the gH/gL/pUL128-131 five-protein 37. Sylwester AW, Mitchell BL, Edgar JB, Taormina
complex of human cytomegalovirus (HCMV) in C, Pelte C, Ruchti F, Sleath PR, Grabstein KH,
primary and reactivated HCMV infections. J Clin Hosken NA, Kern F, Nelson JA, Picker LJ. 2005.
Virol 52:113118. Broadly targeted human cytomegalovirus-specific
26. Kropff B, Burkhardt C, Schott J, Nentwich J, CD4+ and CD8+ T cells dominate the memory
Fisch T, Britt W, Mach M. 2012. Glycoprotein N compartments of exposed subjects. J Exp Med 202:
of human cytomegalovirus protects the virus from 673685.
neutralizing antibodies. PLoS Pathog 8:e1002999. 38. Trautmann L, Rimbert M, Echasserieau K,
27. Schmueck M, Fischer AM, Hammoud B, Saulquin X, Neveu B, Dechanet J, Cerundolo
Brestrich G, Fuehrer H, Luu SH, Mueller K, V, Bonneville M. 2005. Selection of T cell clones
Babel N, Volk HD, Reinke P. 2012. Preferential expressing high-affinity public TCRs within
expansion of human virus-specific multifunction- human cytomegalovirus-specific CD8 T cell re-
al central memory T cells by partial targeting of sponses. J Immunol 175:61236132.
the IL-2 receptor signaling pathway: the key role 39. Wynn KK, Fulton Z, Cooper L, Silins SL,
of CD4+ T cells. J Immunol 188:51895198. Gras S, Archbold JK, Tynan FE, Miles JJ,
28. Reddehase MJ. 2000. The immunogenicity of McCluskey J, Burrows SR, Rossjohn J,
human and murine cytomegaloviruses. Curr Opin Khanna R. 2008. Impact of clonal competition
Immunol 12:390396. for peptide-MHC complexes on the CD8+ T-cell
29. Riddell SR, Watanabe KS, Goodrich JM, Li CR, repertoire selection in a persistent viral infection.
Agha ME, Greenberg PD. 1992. Restoration of Blood 111:42834292.
viral immunity in immunodeficient humans by the 40. Wang GC, Dash P, McCullers JA, Doherty PC,
adoptive transfer of T cell clones. Science 257:238 Thomas PG. 2012. T cell receptor alphabeta
241. diversity inversely correlates with pathogen-
30. Walter EA, Greenberg PD, Gilbert MJ, Finch specific antibody levels in human cytomegalovirus
RJ, Watanabe KS, Thomas ED, Riddell SR. 1995. infection. Sci Transl Med 4:128142.
286 ACQUAYE-SEEDAH ET AL.

41. Schleiss MR. 2008. Cytomegalovirus vaccine of human cytomegalovirus gH/gL/pUL128-130-131


development. Curr Top Microbiol Immunol 325: complex and virus spreading may correlate with
361382. virus control in vivo. J Clin Immunol 32:13241331.
42. Plotkin SA, Higgins R, Kurtz JB, Morris PJ, 52. Lilleri D, Kabanova A, Revello MG, Percivalle E,
Campbell DA, Jr, Shope TC, Spector SA, Sarasini A, Genini E, Sallusto F, Lanzavecchia A,
Dankner WM. 1994. Multicenter trial of Towne Corti D, Gerna G. 2013. Fetal human cytomega-
strain attenuated virus vaccine in seronegative lovirus transmission correlates with delayed
renal transplant recipients. Transplantation 58: maternal antibodies to gH/gL/pUL128-130-131
11761178. complex during primary infection. PLoS One 8:
43. Elek SD, Stern H. 1974. Development of a vaccine e59863.
against mental retardation caused by cytomegalo- 53. Fu TM, Wang D, Freed DC, Tang A, Li F, He X,
virus infection in utero. Lancet i:15. Cole S, Dubey S, Finnefrock AC, ter Meulen J,
44. Yow MD, Demmler GJ. 1992. Congenital cyto- Shiver JW, Casimiro DR. 2012. Restoration of
megalovirus disease20 years is long enough. viral epithelial tropism improves immunogenicity
N Engl J Med 326:702703. in rabbits and rhesus macaques for a whole virion
45. Adler SP, Starr SE, Plotkin SA, Hempfling SH, vaccine of human cytomegalovirus. Vaccine 30:
Buis J, Manning ML, Best AM. 1995. Immunity 74697474.
induced by primary human cytomegalovirus in- 54. Wussow F, Yue Y, Martinez J, Deere JD,
fection protects against secondary infection Longmate J, Herrmann A, Barry PA, Diamond
among women of childbearing age. J Infect Dis DJ. 2013. A vaccine based on the rhesus cytomeg-
171:2632. alovirus UL128 complex induces broadly neutral-
46. Jacobson MA, Sinclair E, Bredt B, Agrillo L, izing antibodies in rhesus macaques. J Virol
Black D, Epling CL, Carvidi A, Ho T, Bains R, 87:13221332.
Girling V, Adler SP. 2006. Safety and immuno- 55. Loomis RJ, Lilja AE, Monroe J, Balabanis
genicity of Towne cytomegalovirus vaccine with KA, Brito LA, Palladino G, Franti M, Mandl
or without adjuvant recombinant interleukin-12. CW, Barnett SW, Mason PW. 2013. Vectored co-
Vaccine 24:53115319. delivery of human cytomegalovirus gH and gL
47. Cui X, Meza BP, Adler SP, McVoy MA. 2008. proteins elicits potent complement-independent
Cytomegalovirus vaccines fail to induce epithelial neutralizing antibodies. Vaccine 31:919926.
entry neutralizing antibodies comparable to nat- 56. Roy DM, Grundy JE. 1992. Evaluation of neutral-
ural infection. Vaccine 26:57605766. izing antibody titers against human cytomegalo-
48. Pass RF, Zhang C, Evans A, Simpson T, Andrews virus in intravenous gamma globulin preparations.
W, Huang ML, Corey L, Hill J, Davis E, Flanigan Transplantation 54:11091110.
C, Cloud G. 2009. Vaccine prevention of maternal 57. Planitzer CB, Saemann MD, Gajek H, Farcet
cytomegalovirus infection. N Engl J Med 360: MR, Kreil TR. 2011. Cytomegalovirus neutraliza-
11911199. tion by hyperimmune and standard intravenous
49. Griffiths PD, Stanton A, McCarrell E, Smith C, immunoglobulin preparations. Transplantation
Osman M, Harber M, Davenport A, Jones G, 92:267270.
Wheeler DC, OBeirne J, Thorburn D, Patch D, 58. Gupta CK, Leszczynski J, Gupta RK, Siber GR.
Atkinson CE, Pichon S, Sweny P, Lanzman M, 1996. IgG subclass antibodies to human cytomeg-
Woodford E, Rothwell E, Old N, Kinyanjui R, alovirus (CMV) in normal human plasma samples
Haque T, Atabani S, Luck S, Prideaux S, Milne and immune globulins and their neutralizing
RS, Emery VC, Burroughs AK. 2011. Cytomega- activities. Biologicals 24:117124.
lovirus glycoprotein-B vaccine with MF59 adju- 59. Adler SP, Nigro G. 2009. Findings and conclusions
vant in transplant recipients: a phase 2 randomised from CMV hyperimmune globulin treatment trials.
placebo-controlled trial. Lancet 377:12561263. J Clin Virol 46(Suppl 4):S54S57.
50. La Rosa C, Longmate J, Lacey SF, Kaltcheva T, 60. Filipovich AH, Peltier MH, Bechtel MK,
Sharan R, Marsano D, Kwon P, Drake J, Dirksen CL, Strauss SA, Englund JA. 1992.
Williams B, Denison S, Broyer S, Couture L, Circulating cytomegalovirus (CMV) neutralizing
Nakamura R, Kelsey MI, Krieg AM, Diamond activity in bone marrow transplant recipients:
DJ, Zaia JA. 2012. Clinical evaluation of safety and comparison of passive immunity in a randomized
immunogenicity of PADRE-cytomegalovirus study of four intravenous IgG products adminis-
(CMV) and tetanus-CMV fusion peptide vaccines tered to CMV-seronegative patients. Blood 80:
with or without PF03512676 adjuvant. J Infect Dis 26562660.
205:12941304. 61. Snydman DR, Werner BG, Dougherty NN,
51. Lilleri D, Kabanova A, Lanzavecchia A, Gerna Griffith J, Rubin RH, Dienstag JL, Rohrer RH,
G. 2012. Antibodies against neutralization epitopes Freeman R, Jenkins R, Lewis WD, Hammer S,
CHAPTER 16 Immunotherapeutic Approaches to Prevent CMV Disease 287

ORourke E, Grady GF, Fawaz K, Kaplan MM, Winkler TH, Mach M. 2007. Protection from
Hoffman MA, Katz AT, Doran M. 1993. Cyto- CMV infection in immunodeficient hosts by
megalovirus immune globulin prophylaxis in liver adoptive transfer of memory B cells. Blood 110:
transplantation. A randomized, double-blind, 34723479.
placebo-controlled trial. Ann Intern Med 119: 72. Nigro G, Adler SP, La Torre R, Best AM. 2005.
984991. Passive immunization during pregnancy for con-
62. King SM, Superina R, Andrews W, Winston DJ, genital cytomegalovirus infection. N Engl J Med
Dunn S, Busuttil RW, Colombani P, Paradis K. 353:13501362.
1997. Randomized comparison of ganciclovir plus 73. Nigro G, Adler SP, Parruti G, Anceschi MM,
intravenous immune globulin (IVIG) with IVIG Coclite E, Pezone I, Di Renzo GC. 2012. Immu-
alone for prevention of primary cytomegalovirus noglobulin therapy of fetal cytomegalovirus infec-
disease in children receiving liver transplants. Clin tion occurring in the first half of pregnancya
Infect Dis 25:11731179. case-control study of the outcome in children.
63. Fisher RA, Kistler KD, Ulsh P, Bergman GE, J Infect Dis 205:215227.
Morris J. 2012. The association between cyto- 74. Cahill AG, Odibo AO, Stamilio DM, Macones
megalovirus immune globulin and long-term GA. 2009. Screening and treating for primary
recipient and graft survival following liver trans- cytomegalovirus infection in pregnancy: where
plantation. Transpl Infect Dis 14:121131. do we stand? A decision-analytic and economic
64. Leroy F, Sechet A, Abou Ayache R, Thierry A, analysis. Am J Obstet Gynecol 201:466, e17.
Belmouaz S, Desport E, Bauwens M, Bridoux F, 75. Nowakowski A, Wang C, Powers DB, Amersdorfer
Touchard G. 2006. Cytomegalovirus prophylaxis P, Smith TJ, Montgomery VA, Sheridan R, Blake
with intravenous polyvalent immunoglobulin in R, Smith LA, Marks JD. 2002. Potent neutraliza-
high-risk renal transplant recipients. Transplant tion of botulinum neurotoxin by recombinant oligo-
Proc 38:23242326. clonal antibody. Proc Natl Acad Sci USA 99:11346
65. Toussaint ND, Tan MB, Nicholls K, Walker RG, 11350.
Cohney SJ. 2011. Low-dose valaciclovir and 76. Maynard JA, Maassen CBM, Leppla SH, Brasky
cytomegalovirus immunoglobulin to prevent K, Patterson JL, Iverson BL, Georgiou G. 2002.
cytomegalovirus disease in high-risk renal trans- Protection against anthrax toxin by recombinant
plant recipients. Nephrology (Carlton) 16:113117. antibody fragments correlates with antigen affin-
66. Bonaros N, Mayer B, Schachner T, Laufer G, ity. Nat Biotechnol 20:597601.
Kocher A. 2008. CMV-hyperimmune globulin 77. Masuho Y, Matsumoto Y, Tomiyama T, Sugano
for preventing cytomegalovirus infection and dis- T, Ono S. 1990. Characterization of human anti-
ease in solid organ transplant recipients: a meta- cytomegalovirus monoclonal antibody as biologics.
analysis. Clin Transplant 22:8997. Dev Biol Stand 71:127136.
67. Cordonnier C, Chevret S, Legrand M, Rafi H, 78. Maynard J, Georgiou G. 2000. Antibody engi-
Dhedin N, Lehmann B, Bassompierre F, Gluckman neering. Annu Rev Biomed Eng 2:339376.
E. 2003. Should immunoglobulin therapy be 79. Rasmussen L, Morris S, Wolitz R, Dowling A,
used in allogeneic stem-cell transplantation? A Fessell J, Holodniy M, Merigan TC. 1994.
randomized, double-blind, dose effect, placebo- Deficiency in antibody response to human cyto-
controlled, multicenter trial. Ann Intern Med 139:8 megalovirus glycoprotein gH in human immuno-
18. deficiency virus-infected patients at risk for
68. Raanani P, Gafter-Gvili A, Paul M, Ben-Bassat I, cytomegalovirus retinitis. J Infect Dis 170:673677.
Leibovici L, Shpilberg O. 2009. Immunoglobulin 80. Tempest PR, White P, Buttle M, Carr FJ,
prophylaxis in hematopoietic stem cell transplan- Harris WJ. 1995. Identification of framework
tation: systematic review and meta-analysis. J Clin residues required to restore antigen binding during
Oncol 27:770781. reshaping of a monoclonal antibody against the
69. Chatterjee A, Harrison CJ, Britt WJ, Bewtra C. glycoprotein gB of human cytomegalovirus. Int J
2001. Modification of maternal and congenital Biol Macromol 17:3742.
cytomegalovirus infection by anti-glycoprotein b 81. Arizono H, Sugano T, Kaida S, Shibusawa K,
antibody transfer in guinea pigs. J Infect Dis Karasawa Y, Esumi Y, Kondo S, Kiyoki M. 1994.
183:15471553. Pharmacokinetics of a new human monoclonal
70. Bratcher DF, Bourne N, Bravo FJ, Schleiss MR, antibody against cytomegalovirus. Third commu-
Slaoui M, Myers MG, Bernstein DI. 1995. Effect nication: correspondence of the idiotype activity
of passive antibody on congenital cytomegalovirus and virus neutralization activity of the new
infection in guinea pigs. J Infect Dis 172:944950. monoclonal antibody, regavirumab in rat serum
71. Klenovsek K, Weisel F, Schneider A, Appelt U, and its pharmacokinetics in rats and monkeys.
Jonjic S, Messerle M, Bradel-Tretheway B, Arzneimittelforschung 44:909913.
288 ACQUAYE-SEEDAH ET AL.

82. Drobyski WR, Gottlieb M, Carrigan D, Ostberg Buick R, Falciola L, Murphy M, Garotta G,
L, Grebenau M, Schran H, Magid P, Ehrlich P, Malavasi F. 2008. Generation of potent neutral-
Nadler PI, Ash RC. 1991. Phase I study of safety izing human monoclonal antibodies against cyto-
and pharmacokinetics of a human anticyto- megalovirus infection from immune B cells. BMC
megalovirus monoclonal antibody in allogeneic Biotechnol 8:85.
bone marrow transplant recipients. Transplanta- 89. Liddy N, Bossi G, Adams KJ, Lissina A, Mahon
tion 51:11901196. TM, Hassan NJ, Gavarret J, Bianchi FC,
83. Nokta M, Tolpin MD, Nadler PI, Pollard RB. Pumphrey NJ, Ladell K, Gostick E, Sewell AK,
1994. Human monoclonal anti-cytomegalovirus Lissin NM, Harwood NE, Molloy PE, Li Y,
(CMV) antibody (MSL 109): enhancement of in Cameron BJ, Sami M, Baston EE, Todorov PT,
vitro foscarnet- and ganciclovir-induced inhibi- Paston SJ, Dennis RE, Harper JV, Dunn SM,
tion of CMV replication. Antivir Res 24:1726. Ashfield R, Johnson A, McGrath Y, Plesa G,
84. Manley K, Anderson J, Yang F, Szustakowski J, June CH, Kalos M, Price DA, Vuidepot A,
Oakeley EJ, Compton T, Feire AL. 2011. Human Williams DD, Sutton DH, Jakobsen BK. 2012.
cytomegalovirus escapes a naturally occurring Monoclonal TCR-redirected tumor cell killing.
neutralizing antibody by incorporating it into Nat Med 18:980987.
assembling virions. Cell Host Microbe 10:197209. 90. Makler O, Oved K, Netzer N, Wolf D, Reiter Y.
85. Gilpin AM, Holbrook JT, Jabs DA, Meinert 2010. Direct visualization of the dynamics of
CL. 2003. Data and safety monitoring board antigen presentation in human cells infected
deliberations resulting in the early termination with cytomegalovirus revealed by antibodies
of the Monoclonal Antibody Cytomegalovirus mimicking TCR specificity. Eur J Immunol 40:
Retinitis Trial. Control Clin Trials 24:9298. 15521565.
86. Jabs DA, Gilpin AM, Min YI, Erice A, Kempen 91. Lum LG, Ramesh M, Thakur A, Mitra S, Deol
JH, Quinn TC. 2002. HIV and cytomegalovirus A, Uberti JP, Pellett PE. 2012. Targeting
viral load and clinical outcomes in AIDS and cytomegalovirus-infected cells using T cells
cytomegalovirus retinitis patients: Monoclonal armed with anti-CD3 x anti-CMV bispecific
Antibody Cytomegalovirus Retinitis Trial. AIDS antibody. Biol Blood Marrow Transplant 18:
16:877887. 10121022.
87. Boeckh M, Bowden RA, Storer B, Chao NJ, 92. Borucki MJ, Spritzler J, Asmuth DM, Gnann J,
Spielberger R, Tierney DK, Gallez-Hawkins G, Hirsch MS, Nokta M, Aweeka F, Nadler PI,
Cunningham T, Blume KG, Levitt D, Zaia JA. Sattler F, Alston B, Nevin TT, Owens S,
2001. Randomized, placebo-controlled, double- Waterman K, Hubbard L, Caliendo A, Pollard
blind study of a cytomegalovirus-specific mono- RB. 2004. A phase II, double-masked, random-
clonal antibody (MSL-109) for prevention of ized, placebo-controlled evaluation of a human
cytomegalovirus infection after allogeneic hema- monoclonal anti-cytomegalovirus antibody (MSL-
topoietic stem cell transplantation. Biol Blood 109) in combination with standard therapy versus
Marrow Transplant 7:343351. standard therapy alone in the treatment of AIDS
88. Funaro A, Gribaudo G, Luganini A, Ortolan E, patients with cytomegalovirus retinitis. Antivir
Lo Buono N, Vicenzi E, Cassetta L, Landolfo S, Res 64:103111.
Rotavirus

MANUEL A. FRANCO1 and HARRY B. GREENBERG2,3


17
INTRODUCTION

Rotaviruses (RV) are ubiquitous highly infectious double-stranded RNA viruses


of importance in public health because of the severe acute gastroenteritis (GE)
they cause in young children and many other animal species. They are very well
adapted to their host, causing frequent symptomatic and asymptomatic
reinfections. Antibodies are the major component of the immune system that
protects infants against RV reinfection. The relationship between the virus and
the B cells (Bc) that produce these antibodies is complex and incompletely
understood (1). In this review, the following basic aspects of RV-specific Bc (RV-
Bc) will be addressed: (i) ontogeny; (ii) use of immunoglobulin (Ig) genes;
(iii) differential distribution (compartmentalization) in the intestinal and
systemic immune systems; (iv) specificity of RV-Ig produced and the mechanisms
by which it mediates protection; and finally, (v) practical applications for the use
of RV-Ig, including RV-Ig as a prophylactic or therapeutic agent and as a correlate
of protection. The immune response generated against RV vaccines has been
recently reviewed (2, 3) and will only be briefly discussed. The focus of this review
is antibodies induced by natural RV infection in humans, but reference to studies

1
Facultad de Ciencias y Medicina, Ponticia Universidad Javeriana, Bogot, Colombia; 2Departments of Medicine
and Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305; 3Veterans
Affairs Palo Alto Health Care System, Palo Alto, CA 94304.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0011-2013

289
290 FRANCO AND GREENBERG

of the murine and porcine animal models of RV at the membrane surface has been developed.
infection will be made when necessary. Bc that bind green fluorescent protein (GFP)-
containing RV virus-like particles (VLPs) made
from viral core protein VP2 and intermediate
capsid protein VP6 (an immunodominant
THE ANTIBODY RESPONSE AGAINST RV
antibody target) are considered RV-Bc. Figure
1 shows an example of a typical experiment
(i) Ontogeny of RV-Bc and RV-Ig
using RV GFP-containing VLPs, and Table 1
A newborn receives important quantities of RV- gives a summary of the subsets of RV-Bc that
IgG and RV-IgM transplacentally. High levels
of these transplacentally transmitted Igs corre-
late with lower numbers of RV infections in
young children and probably mediate protec-
tion against severe RV disease (4). In addition,
maternal antibodies are also transferred to the
infant through breast milk. It has been postu-
lated that colostrum secretory IgA (sIgA) can be
systemically absorbed to some degree (5), but
most antibodies in the serum of neonates are
transplacentally acquired and do not contain
RV-IgA. Although breast milk antibodies are
thought to have a local intestinal antiviral effect,
this effect is relatively modest when it comes to
preventing severe RV disease (1). In animal
models, high-titered serum maternal antibodies
(similar to what is observed in children from
low-income countries) have some protective
effect and can also inhibit the response to an RV
vaccine (6). In contrast, low-titered maternal
antibody in serum (as seen in children from
high-income countries) did not protect against
infection. The experimental conditions that FIGURE 1 Flow cytometry experiment to characterize
induced low-titered maternal antibody in and compare RV-Bc to total Bc subsets (from reference
serum had a complex effect on subsequent 11 with modications). Top row plots illustrate the Bc
subsets considered. Middle row dot plots are gated on
immune response, which included an increase total CD20+ Bc. Bottom row plots are gated on CD20+
in the numbers of RV antibody-secreting cells Bc that bind uorescent RV VLPs (RV-Bc). As a rst step
(ASC) in the intestine induced upon infection in in this analysis, Bc are evaluated for the expression of
a porcine model system (7). In children, the IgD and CD27 (left dot plots). IgD+ CD27- Bc are naive
presence of high-titered maternal antibodies in cells, and IgD- CD27- (IgM, IgG, or IgA+) are a low-
frequency subset of mBc. The CD27+ Bc of the left
both serum and milk is thought to partially panels are further analyzed for the expression of IgD
explain the lower immunogenicity of RV and IgM (right dot plots). IgM- IgD- cells are classical
vaccines in low-income countries (3). The switched mBc; IgM+ IgD- cells are called IgM-only mBc
effect of maternal antibody levels and quality and resemble in many ways the switched mBc. IgD+
on RV vaccine immunogenicity in children IgM- cells are a poorly characterized subset of mBc.
Double-positive IgM+ IgD+ mBc are a heterogeneous
needs to be further investigated (3). population of mBc; in the present experiment, they
Fortunately, RV is one of a few microbial are further subdivided into IgMhi IgDlow (a phenotype
antigens for which a flow cytometry assay to that resembles marginal zone Bc) and IgMlow IgDhi.
identify and characterize Bc expressing RV-Ig doi:10.1128/microbiolspec.AID-0011-2013.f1
CHAPTER 17 Rotavirus 291

TABLE 1 Blood-circulating Bc subsets in healthy adults and their enrichment or not in RV-Bc
Bc subset common name Function and/or relationship of the Bc subset
(phenotype) to disease Enrichment in RV-Bc Reference(s)
- + +
Naive (CD27 IgD IgM ) Bc that have emigrated from the bone marrow No 53
and express the preimmune repertoire of Ig
IgM mBc (CD27+ IgD+ IgM+) Bc that probably represent a heterogeneous Yes 11, 20, 53
subpopulation, an important part of which has
a prediversied repertoire of Ig; the CD27+
IgDlow IgMhi subset resembles marginal zone
Bc and are particularly enriched in RV-mBc
CD27- mBc (CD27- IgD- Atypical mBc that do not express CD27 and Yes in the IgG+ subset 53
IgM or IgG or IgA+) that are enriched in patients with autoimmune
diseases such as lupus
IgM-only mBc (CD27+ A minor mBc population that expresses IgM Yes 20
IgD- IgM+) and seems to be related to switch mBc
Switch mBc (CD27+ IgD- Classical mBc subset that expresses IgG or IgA Decreased frequency 11, 20, 53
IgM- IgG or IgA+)

have been shown to specifically bind GFP- participate in innate Ig responses to patho-
containing VLPs using flow cytometry-based gens (12).
assays. Between 6 months (when maternal anti-
Unexpectedly, children are born with a bodies wane) and 3 years of age, almost every
high number (approximately 1 to 2%) of RV- child will have been infected by RV, and
Bc with a naive phenotype (8). These Bc approximately 50% of them will have had at
secrete antibodies that have a low affinity to least one symptomatic infection that consists
VP6 (9). Intriguingly, by 2 months of age, primarily of diarrhea and vomiting (3). The
children will have a high number of RV-Bc primary Bc response in the peripheral blood of
that express IgM, IgD, and CD27 (generally these children is characterized acutely by the
considered a marker constellation for memory presence of circulating RV-IgM ASC (13).
Bc [mBc]), irrespective of whether the chil- Between 1 and 2 weeks after infection, the
dren have had an RV infection, as evidenced classical IgD- CD27+ mBc appear in blood (14),
by the presence of RV-IgA in serum (10). It is and concomitantly, RV-IgA and IgM can be
uncertain at present if these RV-mBc have detected in serum (15) and stool (16). Impor-
or have not been induced by asymptomatic or tantly, in the serum of these children, secreto-
symptomatic RV infections (either enteric ry antibodies (antibodies that have been
or systemic) that, for some reason, do not secreted to the intestinal lumen) can also be
induce serum RV-IgA. However, they may be identified (17). RV sIgs present in the stool and
playing a role in RV immunity. In experiments serum originate from intestinal lamina propria
in which human IgM-mBc (mostly IgD+) are RV ASCs that secrete polymeric antibodies.
passively transferred to RV-infected immuno- These antibodies are transported by the
deficient mice, they are able to switch to IgG polymeric Ig receptor, present on the baso-
ASC and mediate immunity against RV lateral membrane of enterocytes, to the lumen
antigenemia and viremia (11). Moreover, RV- of the intestine. In the lumen, proteases cleave
mBc from adults are enriched in the IgDlow the receptor, leaving part of it covalently
IgMhi CD27+ subset of Bc (11) (Fig. 1, Table 1). attached to the Ig. This portion of the receptor,
This phenotype is reminiscent of spleen known as the secretory component, serves as a
marginal zone Bc, a subset that has been marker of sIg and protects the Igs from
postulated to develop (by an unknown mech- degradation in the harsh intestinal lumen. By
anism) a prediversified Ig repertoire and to an unknown mechanism, some of these sIg
292 FRANCO AND GREENBERG

antibodies are retrotranscytosed and reach the and adults was unexpected, since it was
systemic circulation (18). Four months after thought that childrens Bc would have similar-
viral infection, RV-IgA persist in the circula- ities to Bc present during the fetal period,
tion of children but the RV-sIg have disap- which have a particular repertoire of VH and
peared (17). This finding has been associated VL gene usage and lack junctional diversity.
with the fact that the production of local These results suggested that the Bc repertoire
intestinal antibodies is short lived compared is not a limiting determinant of the quality of
to antibodies (especially RV-IgG) present in antibody responses to RV in children (21).
serum (see below for discussion on the per- Nonetheless, a subsequent report showed that
sistence of RV-Ig) (19). VH sequences of RV-Bc from children with
Children and adults can be reinfected with acute RV GE have a lower number of mutations
RV, with the majority of reinfections being less than those of the corresponding adult se-
symptomatic or asymptomatic compared to the quences (22). Besides, further studies that
first one or two. When caring for children with concentrated on VP6-specific antibodies en-
RV infections, up to 50% of parents become coded by the VH1-46 gene segment (an
infected and half of them will have mild GE immunodominant gene in RV-Bc of both
symptoms. Among the elderly, it is not uncom- children and adults) showed that the mutations
mon for symptomatic RV outbreaks to reoccur, detected in adults conferred functional advan-
presumably due to waning immunity and or tages to these antibodies (9). For instance,
immunesenescence in this population (1). somatic mutations in the H chain CDR2 region
of these antibodies generated a prolonged off-
rate in VP6 binding and increased antiviral
(ii) Ig Gene Usage of RV-Bc
activity in an in vitro intracellular neutraliza-
The relatively high number of circulating RV- tion assay. Of note, using three-dimensional
Bc and the availability of a flow cytometry cryoelectron microscopy, investigators demon-
assay (Fig. 1) to detect them have permitted strated that these antibodies bind VP6 where
extensive studies of the Ig genes used by these this protein forms viral type 1 channels,
cells in human peripheral blood and, to a much suggesting that the mechanism of intracellular
lesser degree, the intestine (9, 20, 21). The neutralization could involve inhibition of viral
general strategy to perform these studies has RNA release during replication (9).
been to clone and express the genes of single More recently, the Ig gene repertoire of
RV-Bc sorted (using the same strategy used to VP6-Bc in circulating naive (CD19+ IgD+
identify RV-Bc by flow cytometry described CD27-) or mBc subsets (CD19+ IgD+ CD27+
above and in Fig. 1) and in some cases ex- IgM mBc or CD19+ IgD- CD27+ switched mBc
panded in vitro. These studies are robust that in these experiments include IgM-only
because the genes amplified by PCR come mBc) from healthy adults has been evaluated
from single cells and the specificity and (20). As previously stated, compared to non-
functionality of the antibody they produce antigen-specific mBc, RV-mBc had an in-
can be further tested. creased frequency of IgM mBc (Table 1).
Initial findings showed that, compared to Also, IgM RV-mBc had a shorter CDR3 length
non-antigen-specific Bc, RV-Bc of healthy than naive and RV-switched mBc. This could
adults have a biased usage of VH genes (21). be explained by both lower numbers of N and
In addition, the RV-Bc VH, D, JH, VL, and JL P nucleotide additions and shorter D segment
segment usage, extent of junctional diver- length, due to increased exonuclease activity.
sity, and mean H chain complementarity- A comparable finding was observed for total
determining region 3 length of adults and non-antigen-specific IgM-mBc, suggesting
infants were found to be similar (21). The that IgM RV-mBc are probably selected by
genetic resemblance between cells from infants the same unidentified mechanism (20). In
CHAPTER 17 Rotavirus 293

addition, the authors observed that switched blood RV-Bc (23). Ten of the IgA antibodies
RV-mBc were enriched in IgM-only mBc at were expressed, and 8 were shown to bind RV
the expense of IgA mBc (Table 1) and had a but not a control antigen. In the second report,
lower frequency of somatic mutations. These the genes from total non-antigen-specific IgA
findings suggest that RV-mBc have undergone and IgG plasmablasts (CD38+ CD27+ cells still
less extensive maturation in germinal centers expressing the corresponding Ig isotype) from
(where switching and somatic hypermutation the lamina propria of the terminal ileum of
occur). Although the IgM mBc and switched healthy adult volunteers were cloned and
mBc subpopulations of RV-mBc thus seem to expressed in vitro (6). Although the IgG plas-
be selected by different mechanisms, they mablasts were approximately 10 times less
share a very intriguing difference from naive frequent than the IgA plasmablasts, the IgH,
RV-Bc. While the VH1 family is the dominant IgK, and IgL chain gene repertoire of both
VH gene family used by naive RV-mBc, the types of plasmablasts were similar to each
VH3 family is dominant in both subtypes of other and to their blood-circulating counter-
RV-mBc. Particularly, the dominance of the parts. The reactivity profile of Ig produced by
VH1-46 gene segment usage is high in naive the Bc clones against a large panel of self-
RV-Bc (28.6%) compared with IgM and antigens, intestinal bacteria, and RV was
switched RV-mBc (7.8 and 8.3%, respectively). characterized. Approximately 30% of intesti-
In contrast, the VH3 family is the predominant nal IgA and IgG plasmablast antibodies were
VH family used by naive mBc and the two polyreactive, and the majority of these recog-
subtypes of mBc in total non-antigen-specific nized RV 2/6 VLPs. Only 1 of 137 IgA and 2 of
Bc. Altogether, these findings indicate that, 85 IgG plasmablast clones were exclusively
unexpectedly, the dominant naive RV-VH1- specific for the RV VLPs (24). Thus, the
expressing Bc do not have an advantage in the majority of intestinal plasmablasts that recog-
two selection processes that give rise to IgM nize RV (most probably VP6) are polyreactive.
and switched RV-mBc. Hence, more studies This observation underscores the necessity to
are necessary at both the ontological and characterize the reactivity of RV-Ig obtained
functional levels to understand the relation- from blood-circulating Bc in the studies pre-
ship between the RV-specific naive and mem- viously described. Hence, future investigations
ory repertoires of Bc. to directly compare the blood and intestinal
The investigations described above have RV-Bc repertoires seem warranted.
been performed with blood-circulating Bc,
and as previously stated, RV-Bc are probably
(iii) Compartmentalization of RV-Bc
concentrated in the intestine (see below for
discussion on the compartmentalization of Although the largest amount of RV replication
RV-Bc). Accordingly, two recent studies ad- in both animals and humans occurs in the
dressed the genes used by intestinal RV-Bc in intestinal tract, infection is generally accom-
healthy adult subjects (23, 24). In the first panied by antigenemia and viremia. Low levels
report, IgG or IgM plasma cells from the small of systemic RV replication commonly occur in
bowel (CD138+ CD27+ that still express surface animals, and many children have elevated liver
Ig of the corresponding isotype) capable of enzyme levels, suggesting that mild hepatitis is
binding 2/6 RV-VLPs were purified by cell a common feature of RV infection (2). With
sorting (23). Ig genes from Bc derived from this pattern of infection, it is not surprising that
three donors were cloned, sequenced, and the distribution of RV-Bc in animals is biased to
expressed. The VH genes of 26 Bc clones intestinal localization but also has an important
obtained (22 IgA, 4 IgM) were highly mutated systemic component (25). For this reason, the
and preferentially expressed the VH4 family, RV model has been useful to test the hypoth-
which is not dominant among peripheral esis that the immune system functions in
294 FRANCO AND GREENBERG

compartments (1). According to this hypothe-


sis, an immune response will be tailored to best
function at the anatomical niche where it
develops, and lymphocytes specific for a
specific pathogen will concentrate in the
compartment of entry of that pathogen. For
RV, this is clearly the case. During an acute
infection, most viral reactive Bc are concen-
trated in the intestine (25). However, concom-
itant with viremia, RV-Bc also appear in the
spleen and bone marrow (25). Nine months
after primary RV infection in a mouse, effector
ASC predominantly secrete RV-IgA (the
isotype suited to survive in the harsh environ-
ment of the intestinal lumen), rather than
RV-IgG, and are concentrated in the small
intestinal lamina propria and, to a lesser extent,
in Peyers patches (Fig. 2, top panel). In
contrast, equal numbers of IgG and IgA RV-
ASC are present in the spleen and bone
marrow (Fig. 2, top panel). When comparing
RV-mBc (characterized by flow cytometry) at
these late time points after infection, it is
estimated that a mouse will have approximate-
ly the same number of RV-mBc in the spleen
and in the Peyers patches (for technical FIGURE 2 Distribution of RV-Bc 9 months after
reasons, RV-mBc were not evaluated in the primary oral infection of mice in selected organs
intestinal lamina propria) (Fig. 2, middle (data from experiments reported in reference 25).
The top panel shows numbers of IgG and IgA RV ASC
panel). However, the frequency of RV-mBc as
per mouse, in different organs, evaluated by enzyme-
a fraction of total cells in each organ is higher in linked immunospot assay. The middle panel shows
the Peyers patches (Fig. 2, bottom panel), numbers of RV-mBc (small IgD- Bc) that bind RV VLPs
indicating a selective accumulation in these per mouse, in different organs, evaluated by ow
organs. An important prediction of the com- cytometry. The bottom panel shows the same data as
the second panel but expressed as RV-mBc per 105
partmentalization hypothesis is that RV-Bc
cells of the respective organ. LN, lymph node; LP,
that have been primed in a given Peyers lamina propria. doi:10.1128/microbiolspec.AID-
patch will have the capacity to migrate to the 0011-2013.f2
blood and, from there, selectively to other
Peyers patches, lamina propria, and other
parts of the intestinal compartment to provide naive Bc that also express this receptor (1).
a thorough protection against the pathogen. Since the CCR9 ligand, the chemokine TECK,
This prediction has been directly demonstrat- is selectively expressed in the small intestine,
ed for RV (26), and the compartmentalized RV-Bc that express CCR9 will home specifi-
migration of the RV-Bc has been shown to cally to this portion of the intestine, where RV
depend on the expression of the integrin 47 predominantly replicates. In humans, a direct
(the intestinal homing receptor) and the relationship has been shown between circu-
chemokine receptor 9 (CCR9) (1). Of note, lating RV-ASCs and the presence of RV-
relatively high levels of 47 are expressed by specific ASCs in the human small intestinal
intestinally committed mBc/ASC compared to lamina propria (27). Moreover, in children
CHAPTER 17 Rotavirus 295

with an acute RV infection, the great majority persistence (30). In this model, both wild-type
of blood-circulating RV-ASC express both and IgA-deficient Bc expressing the intestinal
intestinal homing receptors (Fig. 3, left panel) homing receptor mediated viral immunity,
(14). In the convalescent phase of viral infec- although the latter did so with somewhat lesser
tion, approximately one-third of RV-mBc efficiency (31). This suggets that intestinal
express both receptors and are presumably localization and, to a lesser extent, Ig isotype
targeted to the small intestine. Another third are critical factors in the protection mecha-
only express 47 (presumably targeted to nism. Another factor that seems critical in
other parts of the intestine and other mucosal determining the capacity of a Bc to protect
surfaces), and the final third express neither against RV is the age at which the mice are
receptor (presumably targeted to the spleen immunized. The age of immunization is, itself,
and other systemic organs) (Fig. 3, right panel) related to the capacity of the mouse to develop
(13, 14). This result (Fig. 3) is consistent with a CD4+ T cells. Mouse pups immunized orally
compartmentalized mBc response to RV in with a heterologous, poorly replication-
humans, as observed in animals (Fig. 2). competent simian RV were less protected
than adult mice and produced less RV intestinal
IgA and neutralizing antibodies. This finding
(iv) Mechanisms of Protection and
was associated with the immature T-cell
Specicity of RV-Ig
response in the pups (32). In fact, more than
Although in mice RV-Bc can play a contribu- 90% of the intestinal IgA in adult mice is CD4+
tory role in viral clearance (28), they are T-cell dependent (29). Although the T-cell-
primarily necessary for preventing reinfection independent response may aid in viral clear-
(29). The localization of RV-Bc to the intestine ance (28), the mechanism of how the virus
seems to be critical for these cells to be able to stimulates it is not completely clear. In T-cell-
mediate their antiviral effect, since in immu- deficient mice, RV can induce a strong Bc
nodeficient mice chronically infected with RV, polyclonal activation (33), and this phenome-
the transfer of 47+ mBc is associated with an non has been linked, in one study, to the
antiviral response in the intestine and viral capacity of VP7 to stimulate Bc via Toll 4
clearance, while the transfer of 47- mBc is receptor (34). In vitro, RV has been shown to
associated with a serum response and viral induce activation and differentiation of human
Bc, present in peripheral blood mononuclear
cells, into ASC (35). However, this effect was
not observed with purified Bc, suggesting the
participation of other cells in activating the Bc;
most likely dendritic cells that produce alpha
interferon. In children, intestinal IgA responses
are frequently short lived (16), while systemic
IgG responses tend to persist (19). It has also
been shown that children, like mice, have an
age-related delay in the development of neu-
tralizing Ig (54). A primary infection in young
FIGURE 3 Expression of the integrin 47 (the children thus develops in the context of an
intestinal homing receptor) and CCR9 (chemokine immature immune system, with a poor capacity
receptor 9 whose ligand, TECK or CCL25, is selectively to stimulate memory CD4+ T cells and with the
expressed in the small intestine) on RV-Bc (Bc that bind
development of a T-cell-independent/innate
uorescent RV VLPs) in children with acute (left dot
plot)- or convalescent (right dot plot)-phase RV infec- Bc response that may, in some cases, not persist.
tion. The gure is from reference 14, with modica- The great majority of RV-Igs both in
tions. doi:10.1128/microbiolspec.AID-0011-2013.f3 animals (36) and humans (1) are directed
296 FRANCO AND GREENBERG

against VP6. The role played by most of these which constitutes the stalk of the spike, and
antibodies in RV immunity is incompletely VP8*, localized on the tip of the stalk, which
understood, but it has been postulated that mediates initial cellular attachment. At least
some of these antibodies mediate intracel- four structurally defined epitopes have been
lular neutralization (37). It is hypothesized identified in VP8*, and antibodies against them
that during the transcytosis of polymeric IgA mediate neutralization by blocking virus
from the basolateral membrane of enterocytes attachment to cells (43). Five structurally
to the gut lumen, these Ig bind virus VP6 and defined epitopes have been identified for
expulse it to the gut lumen. Loading of the VP5* (43). The VP5-1 epitope is located in the
sIgA may occur in the crypt enterocytes that apical hydrophobic loops of the protein and
express the polymeric Ig receptor. When these seems to block the association of VP5* with
enterocytes reach the tip of the villi (site of RV membranes, probably blocking membrane
infection), they still may have some IgA that penetration (44). The mechanisms of neutral-
can mediate the proposed viral expulsion. This ization of antibodies directed to the other
model has been supported by in vitro exper- epitopes are not completely clear at present.
iments in which neutralization of RV can be
achieved if the anti-VP6 dimeric IgA anti-
bodies are delivered intracellularly to infected THERAPEUTIC AND PROPHYLACTIC
cells (38). Furthermore, in mice, protection by APPLICATIONS OF RV ANTIBODIES
these antibodies is dependent on transcytosis
of mucosal Igs by the polymeric Ig receptor RV-Ig can have therapeutic/prophylactic appli-
(39) and not mediated via immune exclusion cations and has been used as a correlate of
(see below) (40). Of note, several recent protection for RV vaccines. The latter have
reports have identified llama VP6-directed been recently reviewed (3) and will only be
monoclonal antibodies with substantial tradi- briefly mentioned here.
tional neutralization activity (41, 42). The Passive administration of RV-Ig can short-
mechanistic and structural basis of these en the duration of RV infection in animals (1).
findings has not yet been determined, and Oral administration of commercial human Ig
numerous attempts to find comparable VP6 preparations, with high titers of neutralizing
antibodies in mice and humans have thus far RV-Ig, have been shown to be of clinical
been unsuccessful. benefit in children with RV GE (45). This
The classical mechanism of Ig-mediated type of treatment may be helpful for children
protection against RV involves viral exclusion with primary and acquired immunodeficiency
(block of enterocyte infection) by neutralizing and prolonged RV infection (1, 46). Careful
antibodies directed against the virus outer kinetic studies of children suffering primary
proteins VP4 and VP7 (1). The mechanism of immunodeficiency, who presented with
neutralization for both types of antibodies has chronic diarrhea and RV excretion, confirmed
been described in detail. Based on structural that RV-Ig can survive passage in the gastro-
studies, two main neutralizing epitopes have intestinal tract and temporarily reduce viral
been described for VP7. The VP7-1 epitope excretion (46). However, preparations of
lies at the three corners of VP7 trimers and human Ig are expensive, and multiple al-
involves at least two VP7 molecules. Anti- ternatives have been investigated. Treatment
bodies against this epitope neutralize the virus with Igs extracted from colostrum from
by stabilizing the capsid and preventing viral immunized bovines was shown to reduce
uncoating. The VP7-2 epitope is in the center stool frequency and to accelerate viral clear-
of the protein, and the mechanism of neutral- ance in children with RV GE (47). An even less
ization is unknown (43, 44). VP4, the virus expensive strategy has been to treat children
spike protein, is cleaved in two proteins, VP5*, with Ig extracted from eggs of chickens
CHAPTER 17 Rotavirus 297

immunized with human RV (48). Further very unlikely that total RV IgA is actually the
investigation of strategies to improve the effector of protection but rather a surrogate
efficacy of orally administered antibodies marker of other more specific immune
includes studies of single-chain antibody frag- effectors. Given this indirect measure, it should
ments expressed from llama antibody genes not be unexpected that some vaccinees with
(42, 49). The advantages of this approach are serum RV-IgA responses develop mild RV GE,
that the antibodies are relatively small, soluble, whereas in others, the protection provided by
acid- and heat-resistant, and easy to express in the vaccine can exceed the levels predicted by
multiple vectors, including lactobacilli. Sur- the serum RV-IgA response (2, 3). The first
prisingly, in two such studies, antibodies incongruity can be best explained because of
directed to RV VP6 possessed broad neutral- the induction of serum RV-IgA without intes-
izing activity in vitro and conferred protection tinal RV-IgA. This occurs in mice infected with
against diarrhea in mice (41, 42). It is possible some heterologous RV, for which the viral dose
that the small size of the VHH antibodies may that induces antigenemia is lower than the
enable access to VP6 on the complete triple- dose that induces intestinal infection. When
layered viral particle that is covered by VP4 low doses of these RV are used for infection,
and VP7. Testing of these VHH constructs in a serum RV-IgA can be detected in the absence
clinical setting and further evaluation of the of intestinal RV-IgA and these mice are
preparations mentioned before (bovine colos- susceptible to infection with a homologous
trum and chicken IgY) are necessary to RV (1). In children, this has been hypothesized
determine their practical utility. Furthermore, to occur with the vaccines based on rhesus RV
testing the value of human Ig in relevant (1). A similar situation may exist with the RV5
clinical settings is necessary; for example, a bovine reassortant vaccine, for which the level
recent Cochrane Database systematic review of serum RV-IgA exceeds the level of protec-
concluded that no randomized controlled tion in several clinical studies in developing
trials assessing the effectiveness or safety of countries (2). The second incongruity, the
oral immunoglobulin preparations for the excess actual protection observed when com-
treatment of RV diarrhea in hospitalized low- pared to serum RV-IgA responses (or neutral-
birth-weight infants have been published (50). ization responses), occurs most frequently
Two safe and effective RV vaccines, the with homologous vaccines, like RV1. In this
monovalent G1P[8] Rotarix vaccine (RV1) and case, it is possible that the excess protection is
the bovine reassortant pentavalent RotaTeq explained by an antibody response in the
vaccine (RV5) are recommended by WHO for intestine (where the vaccine virus has been
worldwide use (3). Nonetheless, they are shown to replicate) that is not strong enough
clearly less efficacious in the poorer countries to be reflected in the circulation.
of Africa and Asia, where they would be most Importantly, RV1 and RV5 have similar
useful, than in the Americas and Europe. For efficacy against severe RV GE in countries
this and other practical reasons, these vaccines where a high diversity of strains cocirculate,
need to be improved and/or better ones supporting the conclusion that immunity to
developed (3). Our poor understanding of the RV has a substantial heterotypic component
mechanisms of protection induced by RV (3). For this reason, the specificity of the Bc
vaccines, and in particular the lack of good response is probably only a secondary aspect
correlates of protection, hamper this process, of an ideal correlate of protection and probably
as does our lack of insight as to why the explains why serum RV-IgA is a better corre-
vaccines perform less effectively in the poorest late than the homotypic neutralization re-
countries. Total serum RV-IgA measured sponse. Thus, efforts to identify stronger
shortly after vaccination is the correlate of correlates of protection may have to concen-
protection currently most widely used (1). It is trate on strategies to better evaluate the
298 FRANCO AND GREENBERG

intestinal RV-Ig responses that have the naive Bc, make RV one of the few human
capacity to persist over time. Although stool pathogens in which this question has started
RV-IgA has been shown to be a good correlate to be addressed and need to be pursued.
of protection for natural RV infection (16), this (ii) Currently, immunologists are puzzled by
parameter may not be useful for evaluating the mechanism of selection, function, and
protection induced by RV vaccines because of biological relevance of the IgM mBc (12).
the interference in the measurement of vac- Since RV-mBc are enriched in this subset, RV
cine-induced RV-Ig by maternal antibodies antigen may be used as a tool to unravel the
from breast-feeding (10). This is particularly mystery of IgM mBc. Further studies of this
true in developing countries, where the fre- mBc subset and of the innate and T-cell-
quency of breast-feeding is high and the basal independent antibody response against RV
levels of preimmune stool RV-IgA mask will likely expand our understanding of RV
vaccine-induced coproconversions. As an al- immunity. (iii) The study of RV-Bc may help
ternative, measurement of blood-circulating clarify the extent and mechanisms that deter-
intestinally induced RV-Bc has been proposed mine compartmentalization of the immune
(3). In a gnotobiotic pig model, for example, system. As discussed, limited studies of intes-
both systemic and intestinal RV ASCs have tinal RV-Bc suggest that they use a different Ig
been shown to be a good correlate of protective gene repertoire than RV-Bc in circulation, and
immunity to human RV challenge (51). In spite a direct comparison of both compartments
of this, it is not clear if measurement of RV ASC would help confirm that this is the case.
can predict long-term persistence of immuni- From a practical point of view, ongoing
ty, and alternative efforts have concentrated clinical studies are evaluating whether llama-
on the study of RV-mBc. As previously men- derived single molecule antibodies against VP6
tioned, during an acute RV infection in can provide broad protection against RV, as
children, circulating IgD- RV-mBc express predicted from animal studies. If this is the
intestinal-homing receptors (47+, CCR9+) case, future studies should be aimed at exam-
and thus probably reflect mucosal immunity ining the possibility that humans are also able
(14). In agreement with this finding, a corre- to generate such antibodies and that this
lation between protection from disease and phenomenon might help explain heterotypic
plasma RV-IgA and 47+ CCR9+ IgD- circu- immunity. An alternative option for passive
lating RV-mBc was found in a trial of the protection could be to design antibodies against
attenuated human RV1 vaccine precursor. conserved heterotypic neutralizing epitopes on
However, the correlation coefficients with VP7 and/or VP4. In this respect, antibodies
protection for both parameters were low, against the VP5-1 epitope seem appealing
making them of little practical use (10). because of their conserved nature and essential
functional role of the VP5 domain in viral
penetration.
FUTURE CHALLENGES Concerning correlates of protection for RV
vaccines, it is possible that formal statistical
RV-Bc have been studied at many levels, analysis of serum RV-IgA responses from
opening several fruitful areas of basic research children in the large clinical trials that have
on the interaction between Bc and an enteric been performed to date will establish this
human viral pathogen. Critical questions that parameter as a practical surrogate marker of
remain to be addressed include (i) the rela- protection. Even if so, as discussed above, the
tionship between the preimmune repertoire development of better correlates seems desir-
and the mBc repertoire, especially in the able, and in this respect, two areas of further
intestine. The studies described above, which study seem promising. (i) The characterization
take advantage of the high numbers of RV- of serum RV-sIg induced by RV vaccines may
CHAPTER 17 Rotavirus 299

be a useful way to obtain an indirect measure of 3. Angel J, Franco MA, Greenberg HB. 2012.
the immune response generated in the intes- Rotavirus immune responses and correlates of
protection. Curr Opin Virol 2:419425.
tine. Preliminary studies have shown that this is 4. Ray PG, Kelkar SD, Walimbe AM, Biniwale V,
possible (J. Angel and M. Franco, unpublished Mehendale S. 2007. Rotavirus immunoglobulin
data). However, an important number (approx- levels among Indian mothers of two socio-
imately one-third) of placebo recipients appear economic groups and occurrence of rotavirus
to have plasma RV-sIg in the absence of infections among their infants up to six months.
J Med Virol 79:341349.
detectable RV-IgA. This can be explained
5. Ogra SS, Weintraub D, Ogra PL. 1977. Immuno-
because, at this age, RV-sIg is mostly composed logic aspects of human colostrum and milk. III.
of IgM RV-sIg (Angel and Franco, unpub- Fate and absorption of cellular and soluble com-
lished). The best (but not exclusive) explana- ponents in the gastrointestinal tract of the new-
tion for these RV-sIg (presumably IgM) born. J Immunol 119:245248.
responses in nonvaccinated children are low- 6. Nguyen TV, Yuan L, Azevedo MS, Jeong KI,
Gonzalez AM, Iosef C, Lovgren-Bengtsson K,
level asymptomatic infections: at the age of RV Morein B, Lewis P, Saif LJ. 2006. High titers of
vaccination (2 to 3 months), RV RNA may be circulating maternal antibodies suppress effector
detected by a sensitive PCR in the stools of up to and memory B-cell responses induced by an
one-third of healthy children (52). (ii) Further attenuated rotavirus priming and rotavirus-like
studies of circulating RV-mBc (particularly particle-immunostimulating complex boosting
vaccine regimen. Clin Vaccine Immunol 13:475
those primed in the intestine) and their rela- 485.
tionship with levels of intestinal and serum Ig 7. Nguyen TV, Yuan L, Azevedo MS, Jeong KI,
(10, 53) may also be helpful to identify improved Gonzalez AM, Iosef C, Lovgren-Bengtsson K,
correlates of protection. Our poor understand- Morein B, Lewis P, Saif LJ. 2006. Low titer
ing of IgM RV-mBc, and the fact that they are maternal antibodies can both enhance and sup-
press B cell responses to a combined live attenu-
probably present in children that have not been ated human rotavirus and VLP-ISCOM vaccine.
infected with RV (defined by the absence of Vaccine 24:23022316.
serum RV-IgA), hinder these studies. 8. Parez N, Garbarg-Chenon A, Fourgeux C, Le
Deist F, Servant-Delmas A, Charpilienne A,
Cohen J, Schwartz-Cornil I. 2004. The VP6
ACKNOWLEDGMENTS
protein of rotavirus interacts with a large fraction
This work was supported by funds from the of human naive B cells via surface immuno-
globulins. J Virol 78:1248912496.
Pontificia Universidad Javeriana, Colciencias
9. Kallewaard NL, McKinney BA, Gu Y, Chen A,
grant 1203-521-28212, by NIH grants R01 Prasad BV, Crowe JE, Jr. 2008. Functional
AI012362-24 and P30DK56339 to H.B.G., maturation of the human antibody response to
and by a merit review Veterans Affairs grant rotavirus. J Immunol 180:39803989.
to H.B.G. 10. Rojas OL, Caicedo L, Guzman C, Rodriguez LS,
Conflicts of interest: We disclose no conflicts. Castaneda J, Uribe L, Andrade Y, Pinzon R,
Narvaez CF, Lozano JM, De Vos B, Franco MA,
Angel J. 2007. Evaluation of circulating intesti-
CITATION nally committed memory B cells in children
vaccinated with attenuated human rotavirus
Franco MA, and Greenberg HB. 2013. Rotavi- vaccine. Viral Immunol 20:300311.
rus. Microbiol Spectrum 1(2):AID-0011-2013 11. Narvaez CF, Feng N, Vasquez C, Sen A, Angel
J, Greenberg HB, Franco MA. 2012. Human
rotavirus-specific IgM memory B cells have
REFERENCES
differential cloning efficiencies and switch capac-
1. Franco MA, Angel J, Greenberg HB. 2006. ities and play a role in antiviral immunity in vivo.
Immunity and correlates of protection for rota- J Virol 86:1082910840.
virus vaccines. Vaccine 24:27182731. 12. Reynaud CA, Descatoire M, Dogan I, Huetz F,
2. Angel J, Franco MA, Greenberg HB. 2007. Weller S, Weill JC. 2012. IgM memory B cells: a
Rotavirus vaccines: recent developments and fu- mouse/human paradox. Cell Mol Life Sci 69:1625
ture considerations. Nat Rev Microbiol 5:529539. 1634.
300 FRANCO AND GREENBERG

13. Gonzalez AM, Jaimes MC, Cajiao I, Rojas OL, bodies from small-intestinal mucosa. J Immunol
Cohen J, Pothier P, Kohli E, Butcher EC, 185:53775383.
Greenberg HB, Angel J, Franco MA. 2003. 24. Benckert J, Schmolka N, Kreschel C, Zoller MJ,
Rotavirus-specific B cells induced by recent Sturm A, Wiedenmann B, Wardemann H. 2011.
infection in adults and children predominantly The majority of intestinal IgA+ and IgG+ plas-
express the intestinal homing receptor 47. mablasts in the human gut are antigen-specific.
Virology 305:93105. J Clin Investig 121:19461955.
14. Jaimes MC, Rojas OL, Kunkel EJ, Lazarus NH, 25. Youngman KR, Franco MA, Kuklin NA, Rott LS,
Soler D, Butcher EC, Bass D, Angel J, Franco Butcher EC, Greenberg HB. 2002. Correlation of
MA, Greenberg HB. 2004. Maturation and tissue distribution, developmental phenotype, and
trafficking markers on rotavirus-specific B cells intestinal homing receptor expression of antigen-
during acute infection and convalescence in specific B cells during the murine anti-rotavirus
children. J Virol 78:1096710976. immune response. J Immunol 168:21732181.
15. Ray PG, Kelkar SD. 2004. Measurement of 26. Bowman EP, Kuklin NA, Youngman KR, Lazarus
antirotavirus IgM/IgA/IgG responses in the NH, Kunkel EJ, Pan J, Greenberg HB, Butcher
serum samples of Indian children following EC. 2002. The intestinal chemokine thymus-
rotavirus diarrhoea and their mothers. J Med expressed chemokine (CCL25) attracts IgA anti-
Virol 72:416423. body-secreting cells. J Exp Med 195:269275.
16. Coulson BS, Grimwood K, Hudson IL, Barnes 27. Brown KA, Kriss JA, Moser CA, Wenner WJ,
GL, Bishop RF. 1992. Role of coproantibody in Offit PA. 2000. Circulating rotavirus-specific
clinical protection of children during reinfection antibody-secreting cells (ASCs) predict the pre-
with rotavirus. J Clin Microbiol 30:16781684. sence of rotavirus-specific ASCs in the human
17. Hjelt K, Grauballe PC, Andersen L, Schiotz PO, small intestinal lamina propria. J Infect Dis
Howitz P, Krasilnikoff PA. 1986. Antibody re- 182:10391043.
sponse in serum and intestine in children up to six 28. VanCott JL, McNeal MM, Flint J, Bailey SA,
months after a naturally acquired rotavirus gas- Choi AH, Ward RL. 2001. Role for T cell-
troenteritis. J Pediatr Gastroenterol Nutr 5:7480. independent B cell activity in the resolution
18. Mantis NJ, Forbes SJ. 2010. Secretory IgA: of primary rotavirus infection in mice. Eur J
arresting microbial pathogens at epithelial borders. Immunol 31:33803387.
Immunol Investig 39:383406. 29. Franco MA, Greenberg HB. 1995. Role of B cells
19. Bernstein DI, McNeal MM, Schiff GM, Ward and cytotoxic T lymphocytes in clearance of and
RL. 1989. Induction and persistence of local immunity to rotavirus infection in mice. J Virol
rotavirus antibodies in relation to serum anti- 69:78007806.
bodies. J Med Virol 28:9095. 30. Williams MB, Rose JR, Rott LS, Franco MA,
20. Tian C, Luskin GK, Dischert KM, Higginbotham Greenberg HB, Butcher EC. 1998. The memory B
JN, Shepherd BE, Crowe JE, Jr. 2008. Immuno- cell subset responsible for the secretory IgA
dominance of the VH1-46 antibody gene segment response and protective humoral immunity to
in the primary repertoire of human rotavirus- rotavirus expresses the intestinal homing recep-
specific B cells is reduced in the memory compart- tor, alpha4beta7. J Immunol 161:42274235.
ment through somatic mutation of nondominant 31. Kuklin NA, Rott L, Feng N, Conner ME, Wagner
clones. J Immunol 180:32793288. N, Muller W, Greenberg HB. 2001. Protective
21. Weitkamp JH, Kallewaard N, Kusuhara K, intestinal anti-rotavirus B cell immunity is depen-
Bures E, Williams JV, LaFleur B, Greenberg dent on alpha 4 beta 7 integrin expression but does
HB, Crowe JE, Jr. 2003. Infant and adult not require IgA antibody production. J Immunol
human B cell responses to rotavirus share common 166:18941902.
immunodominant variable gene repertoires. 32. VanCott JL, Prada AE, McNeal MM, Stone SC,
J Immunol 171:46804688. Basu M, Huffer B, Jr, Smiley KL, Shao M, Bean
22. Weitkamp JH, Lafleur BJ, Greenberg HB, JA, Clements JD, Choi AH, Ward RL. 2006. Mice
Crowe JE Jr. 2005. Natural evolution of a develop effective but delayed protective immune
human virus-specific antibody gene repertoire responses when immunized as neonates either
by somatic hypermutation requires both hotspot- intranasally with nonliving VP6/LT(R192G) or
directed and randomly-directed processes. Hum orally with live rhesus rotavirus vaccine can-
Immunol 66:666676. didates. J Virol 80:49494961.
23. Di Niro R, Mesin L, Raki M, Zheng NY, Lund- 33. Blutt SE, Warfield KL, Lewis DE, Conner ME.
Johansen F, Lundin KE, Charpilienne A, Poncet 2002. Early response to rotavirus infection
D, Wilson PC, Sollid LM. 2010. Rapid generation involves massive B cell activation. J Immunol
of rotavirus-specific human monoclonal anti- 168:57165721.
CHAPTER 17 Rotavirus 301

34. Blutt SE, Crawford SE, Warfield KL, Lewis DE, 45. Guarino A, Canani RB, Russo S, Albano F,
Estes MK, Conner ME. 2004. The VP7 outer Canani MB, Ruggeri FM, Donelli G, Rubino A.
capsid protein of rotavirus induces polyclonal B- 1994. Oral immunoglobulins for treatment of
cell activation. J Virol 78:69746981. acute rotaviral gastroenteritis. Pediatrics 93:1216.
35. Narvaez CF, Franco MA, Angel J, Morton JM, 46. Losonsky GA, Johnson JP, Winkelstein JA,
Greenberg HB. 2010. Rotavirus differentially Yolken RH. 1985. Oral administration of human
infects and polyclonally stimulates human B serum immunoglobulin in immunodeficient
cells depending on their differentiation state and patients with viral gastroenteritis. A pharmacoki-
tissue of origin. J Virol 84:45434555. netic and functional analysis. J Clin Investig
36. Ishida SI, Feng N, Gilbert JM, Tang B, 76:23622367.
Greenberg HB. 1997. Immune responses to 47. Sarker SA, Casswall TH, Mahalanabis D, Alam
individual rotavirus proteins following heterolo- NH, Albert MJ, Brussow H, Fuchs GJ,
gous and homologous rotavirus infection in mice. Hammerstrom L. 1998. Successful treatment of
J Infect Dis 175:13171323. rotavirus diarrhea in children with immunoglob-
37. Burns JW, Siadat-Pajouh M, Krishnaney AA, ulin from immunized bovine colostrum. Pediatr
Greenberg HB. 1996. Protective effect of rotavi- Infect Dis J 17:11491154.
rus VP6-specific IgA monoclonal antibodies that 48. Sarker SA, Casswall TH, Juneja LR, Hoq E,
lack neutralizing activity. Science 272:104107. Hossain I, Fuchs GJ, Hammarstrom L. 2001.
38. Feng N, Lawton JA, Gilbert J, Kuklin N, Vo P, Randomized, placebo-controlled, clinical trial of
Prasad BV, Greenberg HB. 2002. Inhibition of hyperimmunized chicken egg yolk immunoglob-
rotavirus replication by a non-neutralizing, rota- ulin in children with rotavirus diarrhea. J Pediatr
virus VP6-specific IgA mAb. J Clin Investig Gastroenterol Nutr 32:1925.
109:12031213. 49. Pant N, Hultberg A, Zhao Y, Svensson L, Pan-
39. Schwartz-Cornil I, Benureau Y, Greenberg H, Hammarstrom Q, Johansen K, Pouwels PH,
Hendrickson BA, Cohen J. 2002. Heterologous Ruggeri FM, Hermans P, Frenken L, Boren T,
protection induced by the inner capsid proteins of Marcotte H, Hammarstrom L. 2006. Lactobacilli
rotavirus requires transcytosis of mucosal immu- expressing variable domain of llama heavy-chain
noglobulins. J Virol 76:81108117. antibody fragments (lactobodies) confer protec-
40. Corthesy B, Benureau Y, Perrier C, Fourgeux C, tion against rotavirus-induced diarrhea. J Infect
Parez N, Greenberg H, Schwartz-Cornil I. 2006. Dis 194:15801588.
Rotavirus anti-VP6 secretory immunoglobulin A 50. Pammi M, Haque KN. 2011. Oral immunoglobu-
contributes to protection via intracellular neutral- lin for the treatment of rotavirus diarrhea in low
ization but not via immune exclusion. J Virol birth weight infants. Cochrane Database Syst Rev
80:1069210699. 2011:CD003742.
41. Aladin F, Einerhand AW, Bouma J, Bezemer S, 51. Yuan L, Ward LA, Rosen BI, To TL, Saif LJ.
Hermans P, Wolvers D, Bellamy K, Frenken LG, 1996. Systematic and intestinal antibody-secreting
Gray J, Iturriza-Gomara M. 2012. In vitro cell responses and correlates of protective immu-
neutralisation of rotavirus infection by two broad- nity to human rotavirus in a gnotobiotic pig model
ly specific recombinant monovalent llama- of disease. J Virol 70:30753083.
derived antibody fragments. PLoS One 7:e32949. 52. Amar CF, East CL, Gray J, Iturriza-Gomara M,
42. Garaicoechea L, Olichon A, Marcoppido G, Maclure EA, McLauchlin J. 2007. Detection by
Wigdorovitz A, Mozgovoj M, Saif L, Surrey T, PCR of eight groups of enteric pathogens in 4,627
Parreno V. 2008. Llama-derived single-chain faecal samples: re-examination of the English case-
antibody fragments directed to rotavirus VP6 control Infectious Intestinal Disease Study (1993
protein possess broad neutralizing activity in 1996). Eur J Clin Microbiol Infect Dis 26:311323.
vitro and confer protection against diarrhea in 53. Rojas OL, Narvaez CF, Greenberg HB, Angel J,
mice. J Virol 82:97539764. Franco MA. 2008. Characterization of rotavirus
43. Aoki ST, Settembre EC, Trask SD, Greenberg specific B cells and their relation with serological
HB, Harrison SC, Dormitzer PR. 2009. Structure memory. Virology 380:234242.
of rotavirus outer-layer protein VP7 bound with a 54. Ward RL, Kirkwood CD, Sander DS, Smith VE,
neutralizing Fab. Science 324:14441447. Shao M, Bean JA, Sack DA, Bernstein DI. 2006.
44. Trask SD, McDonald SM, Patton JT. 2012. Reductions in cross-neutralizing antibody re-
Structural insights into the coupling of virion sponses in infants after attenuation of the human
assembly and rotavirus replication. Nat Rev rotavirus vaccine candidate 89-12. J Infect Dis
Microbiol 10:165177. 194:17291736.
Bacterial ToxinsStaphylococcal
Enterotoxin B

BETTINA C. FRIES1 and AVANISH K. VARSHNEY1


18
INTRODUCTION

Staphylococcal Enterotoxins
Staphylococcus aureus is a nonmotile, ubiquitous, gram-positive coccus which is a
major human pathogen responsible for a wide range of infections, including skin
and soft tissue infections, bacteremia, pneumonia, and several toxin-mediated
diseases. Among many extracellular proteins, S. aureus strains also secrete a
variety of potent toxins which include alpha hemolysin, enterotoxins, leukocidins,
and exfoliative toxins, all of which are directly associated with particular disease
manifestations. To date, more than 33 enterotoxin sequences have been described
in various S. aureus genomes. Enterotoxins are heat stable and exert their effect on
the epithelium of the intestinal tract when ingested, and thus, they are a common
cause of food poisoning. Several enterotoxins are potent superantigens (SAgs)
that, in a non-antigen (Ag)-dependent way, predominantly activate CD4+ T cells (1)
but also activate other immune cells. The SAgs of S. aureus include toxic shock
syndrome toxin 1 (TSST-1), enterotoxin serotypes A to E and I (sea, seb, sec, sed,
see, and sei), and enterotoxin-like serotypes G (selG), H (selH), and J to U (selJ to
selU). Of these SAgs, sea to see have the ability to induce emesis in monkeys and

1
Department of Medicine/Infectious Diseases and Department of Microbiology and Immunology, Albert Einstein
College of Medicine, Bronx, NY 10461.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0002-2012

303
304 FRIES AND VARSHNEY

arethus referred to as classic enterotoxins. The estimated to be 0.02 g/kg of body weight for
remaining SAgs either have not been tested for humans exposed by the inhalation route. A
emetic activity or lack emetic activity and are convention on the Prohibition of the Devel-
therefore referred to as enterotoxin-like pro- opment, Production and Stockpiling of Bacte-
teins (selG, selH, and selJ to selU). For the most riological (Biological) and Toxin Weapons and
part, staphylococcal SAgs are encoded by on Their Destruction was signed by the
mobile genetic elements, which include extra- United Kingdom, U.S., and U.S.S.R. govern-
chromosomal plasmids as well as chromosomal ments in 1972. The U.S. government opted to
prophages, transposons, and pathogenicity reestablish research programs for vaccine and
islands. It is noteworthy that a chromosomally therapeutic development against biological
carried enterotoxin-like gene (selX) was recently weapons after suspicion arose that the U.S.S.
identified (2). The seb gene is carried on the R. was continuing the stockpiling and testing of
pathogenicity island SaPI3. The genes of SAgs biological weapons. Major NIH grant funding
selG, selI, selM, selO, and selU are located in the reinvigorated research on biological warfare
enterotoxin gene cluster (egc) and are among agents after 11 September 2001 and especially
the most prevalent SAgs in clinical S. aureus after the anthrax attacks occurred in the U.S.
isolates. They are expressed by S. aureus during mail system. Despite extensive efforts, how-
logarithmic growth and shut off expression once ever, there is no therapy or vaccine approved
a certain bacterial density is reached. Conse- for human use against SEB to date.
quently, they do not induce a humoral response
in the human host. In contrast, non-egc-associ-
ated SAgs (e.g., sea, seb, sec, and tsst-1) are
THE INFECTION
expressed in late-logarithmic and stationary
growth, induce a specific antibody (Ab) re-
Description of Agent
sponse in the human host, and are a prominent
cause of cause toxic shock (3). SEB is a well-characterized 28-kDa protein that
consists of 239 amino acids and is most closely
related to SEC1, with whom it shares structural
SEB
as well as 67% amino acid homology (5, 6). SEB
Staphylococcal enterotoxin B (SEB) is the proto- is water soluble, heat labile, and resistant to
type of a non-egc-associated potent SAg. It is proteolytic enzymes, including pepsin, trypsin,
categorized as a category B select agent and papain. The crystal structure of SEB was
because it is the most potent staphylococcal first determined in 1992 to a resolution of 2.5
enterotoxin, and much lower quantities are (6) and later (7) to a resolution of 1.5 . The
sufficient to produce a toxic effect than with refined model contained 1,948 protein atoms
synthetic chemicals. Furthermore, SEB is and 177 water molecules and had an excellent
extremely stable and easily produced in large geometry with root-mean-square (rms) devia-
quantities. At low concentrations, SEB can tion of 0.007 and 1.73 in bond lengths and
cause multi-organ system failure and death. bond angles, respectively. As a SAg, SEB cross-
During the 1960s, when the United States had links Ag-presenting cells (APCs) and T cells by
an offensive biological warfare program, SEB forming a ternary complex between the im-
was studied as a biological weapon and mune receptors major histocompatibility com-
stockpiled with various other bioweapons plex (MHC) class II and specific V chains of
prior to its destruction in 1972 (4). Based on the T-cell receptors (TcR) (6, 8, 9, 10). SEB
those investigations, the effective dose of SEB protein is ellipsoidal, tightly and compactly
that would incapacitate 50% of the exposed folded into two unequal-sized domains of
population was estimated to be 0.0004 g/kg of mixed / structure. Although the overall fold
body weight, whereas the 50% lethal dose was of SEB is similar to those of other microbial
CHAPTER 18 Bacterial ToxinsStaphylococcal Enterotoxin B 305

SAgs, it lacks the zinc-binding site and only II provides a mechanism for the ability of SEB
possesses one MHC class II binding site. The to induce anergy in some cases and activation
SEB residues implicated in TcR binding are 18, in others (11).
19, 20, 22, 23, 26, 60, 90, 91, 177, 178, and 210. SEB is excreted by S. aureus strains from
The suggested residues involved in binding of diverse clonal complexes. Most, if not all,
SEB to the MHC binding site are 43 to 47, 65, staphylococcal strains designated as part of
67, 89, 92, 94, 96, 98, 115, 209, 211, and 215. the CDC USA400 clonal group (by pulsed-field
Furthermore, the C-terminal disulfide loop gel electrophoresis) produce large amounts of
(residues 113 to 126) in SEB has high flexibility, SEB or SEC. One study with isolates derived
and it has been suggested to be responsible for from New York identified SEB in four clonal
emetic properties (6). Additional data from complexes, with CC8 being the most common,
native gel electrophoresis and plasmon reso- followed by CC59, CC20, and one unassigned
nance affinity measurements indicate that the strain (12). Sequence analysis of 20 different S.
SEB-TcR complex can even form in the absence aureus strains identified amino acid substitu-
of MHC class II and that SEB-TcR interaction tions when compared to the SEB of strains
increases the binding to the MHC class II COL and MNHO. These amino acid mutations
molecule DR1. It has been proposed that the involve positions 7 (lysine-asparagine), 14
finding that SEB can form complexes with TcR (serine-alanine), 35 (alanine-serine), 125 (glu-
in both the absence and presence of MHC class tamine-histidine), 192 (asparagine-serine), and

FIGURE 1 Alignment of amino acid sequences of SEB derived from S. aureus clinical isolates. Amino acid
mutations are highlighted in green. MHC- and TcR-interacting residues are shown in blue and magenta,
respectively. doi:10.1128/microbiolspec.AID-0002-2012.f1
306 FRIES AND VARSHNEY

222 (methionine-leucine) (13) (Fig. 1). It is TcR (6, 8, 9, 10) formed by this cross-linking.
noteworthy that these amino acid sequences SEB binds to the MHC molecule outside the
lie outside the residues that are responsible for peptide-binding groove without prior
binding to MHC class II molecule and the TcR processing, stimulating one of the seven Vh
(Fig. 2). Investigations with purified, variant subclasses of the TcR (3, 12, 13.2, 14, 15, 17, or
SEBs indicated that they varied in inducing 20). Stimulated T cells then release large
proliferation of rabbit splenocytes in vitro as amounts of cytokines, namely interleukin-2
well as in lethality in a rabbit model of toxic (IL-2), tumor necrosis factor alpha (TNF-),
shock syndrome (TSS) (13). Enzyme-linked and gamma interferon (IFN-), and undergo
immunosorbent assay (ELISA)-based quanti- hyperproliferation and ultimately depletion.
fication of SEB in supernatants of cultures Cell adhesion molecules such as CD2 and
in log phase demonstrates great variability ELAM on endothelial cells can also function
among clinical S. aureus isolates, including as coreceptors for SEB-induced T-cell activa-
sequential isolates derived from the same tion and cytokine production (14). The trimer
patient (12). complex activates intracellular signaling,
which elicits phosphotidylinositol production
and intracellular Ca2+ flux. This is followed by
Interaction of Immune Cells with SEB
a rapid activation of membrane-associated
The primary targets of SEB are the TcR on T protein tyrosine kinase and protein kinase C
cells and the MHC class II molecules on APCs, (15). Activation of the CD28-regulated signal
resulting in a ternary complex between MHC transduction pathway is required for SAg
class II molecules and specific V chains of the stimulation in T cells and subsequent IL-2

FIGURE 2 (A) Ribbon structure of SEB protein showing amino acid mutations in S. aureus isolates.
Residues which interact with MHC and TcR are shown in blue and magenta, respectively. (B) View after
rotating 180 degrees around vertical axis. doi:10.1128/microbiolspec.AID-0002-2012.f2
CHAPTER 18 Bacterial ToxinsStaphylococcal Enterotoxin B 307

production. Activation of transcriptional ANIMAL MODELS TO STUDY THE


factors NF-B and AP-1 result in high-level PATHOGENESIS OF SEB IN VIVO
expression of cytokines, including IL-1 and
TNF- from macrophages and TNF-, IL-2, Murine Models
and IFN- from T cells. Excreted cytokines
Clearance of SEB has been investigated in mice
have potent effects and cause fever, hypoten-
after intravenous (i.v.) injection. SEB becomes
sion, multiorgan dysfunction, and ultimately,
systemically distributed within 5 to 30 min in
lethal shock. Table 1 summarizes the biological
blood and in lymph nodes (16). Clearance
and pathological effects of SEB.
occurs within 10 to 24 h via glomerular
filtration in the kidney. Small amounts of
In Vitro Models to Study SAg
SEB are detectable also in the spleen. Mani-
Activity of SEB
festation of functional outcomes such as anergy,
In vitro cellular responses of SEB have been
clonal expansion, and clonal deletion begins
extensively studied in human peripheral
after 24 h.
blood mononuclear cells (PBMCs) and mu-
rine splenocytes. The MHC of murine cells
has a lower affinity to SEB than the human Murine Models with Potentiating Agents
HLA complex. Therefore, humans are many Standard mice are not very sensitive to SEB
times more sensitive to SEB. Human PBMCs due to low-affinity binding of SEB to murine
are sensitive to picomolar concentrations of MHC class II. Therefore, a potentiating
SEB, whereas mouse splenocytes require agent is required to amplify the toxic effect
nanomolar concentrations for stimulation. of SEB. The list of potentiating agents
SEB-induced T-cell proliferation can be includes lipopolysaccharide (LPS), D-galac-
measured with different methods and usu- tosamine, and actinomycin D (10, 17). The
ally peaks at 96 h in the employed in vitro hepatotoxin D-galactosamine induces TNF-
systems. Most cytokine stimulation assays and produces fulminant liver failure and
focus on quantification of IFN- and IL-2, shock when given in combination with SEB,
but other cytokines are also induced, in- and higher levels of TNF- are measured
cluding IL-1, IL-6, IL-12, TNF-, and IL-8. when compared to SEB alone. IL-2-defi-
For optimal induction of proliferation, both cient mice are more resistant to SEB-
T cells and monocytes are required. In vitro induced lethal shock (SEBILS), supporting
studies have demonstrated that SEB can also the importance of IL-2 in the pathogenesis
interact directly with TcR in the absence of of SEBILS (18). The lethal shock in this
MHC class II molecules, which results in an model is associated with high concentra-
anergic T-cell response (11). tions of IL-1, IL-2, TNF-, and INF- in the
serum, which results in shock in mice.
Studies that analyzed the cytokine level in
TABLE 1 Major biological and pathological
activities of SEB
the serum of mice treated with SEB or LPS
alone or in combination (19) demonstrated
Superantigenicity: proliferation of CD4 T cells following
binding with the V motif of TcR and MHC class II higher TNF-, IL-6, macrophage inflamma-
molecules on the surface of APCs tory protein 2, and monocyte chemoattract-
Induction and release of several cytokines ant protein 1 (MCP-1) levels in mice treated
Lethality and shock in experimental animals, including with both toxins than in those treated with
mice, rabbits, piglets, and monkeys either alone. Significantly higher levels of
Emetic activity IFN- and IL-2 were observed at the later
Direct or indirect involvement in pathogenesis of time point. However, it remains difficult to
severe diseases, including TSS, nonmenstrual TSS, discern which cytokine induction is specif-
atopic dermatitis, asthma, and chronic rhinitis
ically caused by SEB in this model.
308 FRIES AND VARSHNEY

Murine Models Without also show a toxicity to SEB similar to those


Potentiating Agents observed in staphylococcal food-borne illness,
A dual-dose SEB model has been described for namely emesis when ingested orally, although
C3H/HeJ, a Toll-like receptor 4-defective this is not observed with the streptococcal
mouse, and involves giving one dose of SEB SAgs or TSST-1. The role of SAgs (TSST-1,
(5 g) administered intranasally followed by SEB, and SEC) in the setting of staphylococcal
another dose of SEB (2 g) intraperitoneally sepsis has been successfully investigated in a
(i.p.) 2 h later, which results in high serum rabbit model of lethal pulmonary infection
concentrations of IL-2, IL-6, and MCP-1 as (25).
well as elevated MCP-1 levels in the lung
(20). The increased concentration of MCP-1,
Piglet Model
a potent activator and chemotactic factor for
T cells and monocytes, may contribute to A piglet model has also been used in assessing
leukocyte recruitment into the lungs. Lethal- and understanding pathology and toxicity
ity and clinical signs of intoxication, such as following native SEB challenge (26). For
ruffled fur and hypothermia, are similar to these models, weaning 7- to 14-day-old York-
those observed in transgenic mice and non- shire piglets are injected with SEB i.v. The
human primates. clinical signs are biphasic, with pyrexia, vom-
Another dual-dosing model for SEBILS is iting, and diarrhea within 4 h, followed by
the HLA transgenic mouse model. These terminal hypotension and shock by 96 h. Mild
MHC class II knockout mice express the lymphoid lesions are identified as early as 24 h,
human MHC class II determinant DR3 in with severe lymphadenopathy, splenomegaly,
trans and are thus more sensitive to SEBILS. and prominent Peyers patches by 72 h.
Two doses of SEB (50 g) given 48 h apart Widespread edemamost prominent in the
i.p. induce toxic shock and 100% mortality mesentery, between loops of spiral colon, and
(21). These mice also exhibit high levels of in retroperitoneal connective tissueis found
IFN-, IL-2, and IL-6 when SEB is admin- at 72 h. Additional histologic changes included
istered by aerosol (22). perivascular aggregates of large lymphocytes
variably present in the lung and brain, circu-
lating lymphoblasts, and lymphocytic portal
Rat Model
hepatitis. The piglet model has also been
Rats have been used to study the effect of SEB successfully used in oral vaccine studies. Like
in the central nervous system and to specifi- human cells, pig leukocytes readily respond to
cally clarify the role of the vagus nerve in native SEB. Therefore, the piglet model is
sensation and transmission of abdominal SEB superior to mouse models which require
stimulation. In this model, i.p. administration potentiation of SEB toxicity (27) and the DR3
of SEB (1 mg/kg of body weight) induced a transgenic mouse model in mirroring the
robust Fos expression and induced activation biphasic clinical response and overall pathol-
of neurons in widespread brain areas, trans- ogy observed in humans. It is considerably
mitting the signal of abdominal immune cheaper than the rhesus macaque model.
stimulation to the brain (23).
Nonhuman Primate Model
Rabbit Model
In the late 1960s, rhesus monkeys (Macaca
Rabbits are more sensitive to many staphylo- mulatta) were used to study SEB pathogenesis
coccal toxins and develop pyrogenic symp- because they exhibit disease progression sim-
toms similar to those of humans when SAgs ilar to that observed in humans (28). They
are given by continuous perfusion (24). They manifest multiorgan failure and shock when
CHAPTER 18 Bacterial ToxinsStaphylococcal Enterotoxin B 309

injected i.v. Specifically, they exhibit acute Research Institute who developed conjunctivi-
renal failure, terminal depression of electro- tis with localized cutaneous swelling within 1 to
encephalographic patterns, fever, and emesis. 6 h, followed by gastrointestinal symptoms in
A SEB-induced immediate-type skin reaction two of the three workers after accidental
was also investigated in unsensitized monkeys. cutaneous or ocular exposure to SEB (30).
Substance P plays a predominant role in
mediating intradermal SEB challenge and
Toxic Shock Syndrome (TSS)
exerts its effect on cutaneous mast cells via
stimulation of primary sensory neurons that TSS is characterized by the occurrence of
contain substance P (29). The prohibitively fever, hypotension, multiple organ system
high expenses and the limited number of dysfunction, rash, and desquamation, and it is
monkeys that can be used per group limit the classified as nonmenstrual or menstrual. The
use of this model. latter was first characterized in 1978 (31) in
In summary, several animal models are women that used tampons and is associated
available to test neutralization of SEB in vivo. with TSST-1. The incidence has decreased
It should be pointed out that SEB quantities significantly in past years (32), and nonmen-
differ between the models, as do the inherent strual cases account for 55% of all cases (33).
sensitivities of the animals. This has to be Those latter syndromes are commonly associ-
taken into consideration when the efficacies of ated with SEB. TSS has been reported to occur
Abs are compared. in association with use of barrier contra-
ceptives and after vaginal and cesarean deliv-
ery. It has also been reported in the setting of
CLINICAL MANIFESTATION AND soft tissue infection, endovascular infection,
EPIDEMIOLOGY and visceral abscesses as well as upper and
lower respiratory tract infection (34). Up to
SEB can cause several clinical symptoms in one-third of patients who have TSS develop
exposed humans. Manifestations of intoxica- recurrent disease. This requires persistent
tion depend on the dose as well as the route of colonization with a toxigenic strain of S.
exposure. aureus and only develops in patients who do
not mount a humoral immune response to the
implicated staphylococcal toxin (35). It is
Food Poisoning
noteworthy that several case reports of staph-
SEB is one of the most common toxins impli- ylococcal TSS without rash have been de-
cated in toxin-mediated food-borne disease. scribed, which can make the diagnosis very
Typically, heavily colonized food handlers difficult. It has been suggested that rash and
contaminate food products with S. aureus via desquamation result from delayed hypersen-
manual contact, coughing, or sneezing. S. sitivity, which is amplified by SAgs (36).
aureus grows rapidly and excretes enterotoxins,
especially in food products such as cream,
Atopic Dermatitis (AD)
mayonnaise, unrefrigerated meats, dairy, and
bakery products. Heating the contaminated AD is a common skin disorder that affects
food only kills the bacteria but does not destroy children during early childhood as well as
the heat-stable, preformed SEB toxin. After adults. Patients with AD are frequently colo-
ingestion of the toxin, the incubation period nized with S. aureus strains. Comparison of
before patients become symptomatic is only colonizing S. aureus strains derived from
approximately 4 to 6 h. This is also supported by patients with uncomplicated AD versus those
data on occupational exposures in three labo- derived from patients with chronic steroid-
ratory workers at the U.S. Army Medical resistant AD indicate that the latter more
310 FRIES AND VARSHNEY

commonly excrete SAg SEB. SAgs induce fever, vomiting, myalgia, diarrhea, headache,
immunoglobulin E (IgE) Abs that are thought and in severe cases, lethal shock. Laboratory
to exacerbate the skin and allergic inflamma- findings are not specific for the diagnosis of
tion in AD. Approximately 50 to 80% of SEB intoxication, as nonspecific neutrophilic
patients with chronic AD have IgE Abs specific leukocytosis and an elevated erythrocyte sed-
to SEA and SEB (37, 38). This hypothesis is imentation rate are present in many illnesses. A
further supported by data derived from a rising Ab titer response to SEB can be helpful
murine model of atopic dermatitis, where to validate the diagnosis retrospectively. Sev-
topical SAg exposure induces epidermal accu- eral methods to directly detect and quantify
mulation of CD8+ T cells, a mixed Th1/Th2 SEB have been developed in recent years. They
type dermatitis, and production of IgE Abs include immunological assays such as immu-
(39). A recent study observed predominance of nodiffusion assays, radioimmunoassay, and
SEB and SED in S. aureus isolates from AD ELISA, which have been applied for detection
patients with low IgE titers characterized by of SEB. Radioimmunoassay can detect up to 1
the prevalence of CD8+ lymphocytes and a ng of SEB/ml in food extract; and ELISAs can
dominant Th1 profile induced by SAgs and detect less than 0.1 ng of SEB/ml in urine,
elevated IFN- expression (40). blood, or food extract. Development of better
instruments for mass spectrometry (MS)
techniques has enhanced possibilities of more-
Respiratory Diseases, Including Asthma
precise structure identification and confirma-
and Nasal Polyps
tion of proteins. Using this technique in
Several studies suggest an association of combination with the Abs surface plasmon
colonization with SEB-excreting S. aureus resonance chip has further enhanced the
and chronic rhinitis. A small clinical study sensitivity and feasibility of these techniques.
comprised of 32 patients indicated a possible Currently, matrix-assisted laser desorption ion-
association between chronic rhinosinusitis izationtime of flight and electrospray ioniza-
and ulcerative colitis (UC). After functional tion (ESI)-time of flight MS-based analysis can
endoscopic sinus surgery, the clinical symp- be completed within 1 h and have a very low
tom scores of chronic rhinosinusitis and UC detection limit of 3 pmol/ml of water (41). The
severe scores were significantly reduced in combination of liquid chromatography-ESI
these patients. Interestingly, the number of MS/MS allows accurate determination by
cultured S. aureus colonies from the surgically molecular mass and also by amino acid se-
removed sinus mucosa significantly correlated quencing after enzymatic digestion. A sensitive
with the decrease in UC severe scores, and laser nephelometric assay was developed to
high levels of SEB were detected in the sinus detect SEB in plasma of healthy volunteers as
wash fluids of these patients. well as patients (42). Despite novel diagnostic
In summary, SEB-mediated disease is di- approaches, evidence that SEB has actually
verse and extends beyond TSS. Recent studies been detected in human body fluids of either
strongly suggest that SEB excretion by colo- infected or intoxicated patients is scarce (43).
nizing strains may worsen inflammatory re-
sponses of allergic diseases.
TREATMENT

DIAGNOSIS There is no treatment available for SEB-


mediated shock other than symptomatic sup-
SEB-mediated intoxication is usually diag- port. The disease in the setting of food
nosed based on clinical suspicion and symp- intoxication is usually self-limiting, and patients
toms. The clinical signs of SEB intoxication are recover with active hydration and supportive
CHAPTER 18 Bacterial ToxinsStaphylococcal Enterotoxin B 311

measures. Steroids and antibiotics have not V Domains


been shown to be effective for SEB intoxication
Another method to neutralize SEB action is
(44). Many approaches to prophylaxis and
by employing soluble forms of genetically
therapy of SEB-mediated diseases have been
engineered V domains. These are high-
explored and are outlined below. They include
affinity toxin-binding agents. A soluble G5-
active immunization with inactivated recom-
8 mouse V (V8.2) mutant was generated and
binant SEB vaccines, synthetic peptides, and
shown to be a promising therapeutic agent.
proteasome-SEB toxoid. Furthermore, Ab-
Both administration of V-TcR G5-8 as well as
based passive immunoprophylaxis/immuno-
prior hyperimmunization to raise neutralizing
therapy, as well as synthetic peptide antagonists
Abs to SEB dramatically increase survival in a
and receptor mimics, such as chimeric mimics
lethal pulmonary disease model in rabbits (49).
of MHC class II-TcR and of the TcR-V,
Only equimolar amounts of these molecules
have been investigated. To date, the FDA has
are required to neutralize SEB, which indi-
licensed no vaccine and antitoxin. However,
cates that they could have beneficial pharma-
numerous studies on various animal models for
codynamic qualities.
SEBILS have shown a favorable outcome with
these diverse ranges of reagents that inhibit the
proliferation of T cells and downregulate the Cytokine Inhibitors
expression of cytokine.
Several drugs can interfere with cytokine
induction and T-cell proliferation. They in-
Peptide Antagonists clude the antibiotic doxycycline, which down-
SEBILS can be successfully blocked with small regulates the SEB-induced proinflammatory
overlapping antagonist peptides that inhibit cytokine and chemokine response as well as
the initial step of toxin-receptor interactions. SEB-induced T-cell proliferation in human
Peptides directed to SEB amino acids 150 to PBMCs (50). Furthermore, pentoxifylline, a
161 showed antagonist activity and protect mice methylxanthine derivative, and dexametha-
from lethal shock against SEA, SEB, and TSST- sone also inhibit SEB-induced activation of
1 when given i.v. 30 min after a lethal toxin dose human PBMCs in vitro and also SEBILS in
(45). This conserved domain of SEB is not mice (51, 52). Recently, rapamycin, an immu-
directly involved in MHC class II or TcR nosuppressant, has also been shown to inhibit
binding; however, it may be involved in cytokine release in vitro and toxin-mediated
interactions with coligands or cytotoxic T- shock in mice (53). All of these therapeutic
lymphocyte antigen, which are necessary for agents indirectly inhibit SEB-induced effects
superantigenic activity. A subsequent study also by downregulating the cytokine responses.
showed that peptides interfering with SEB
domain residues 140 to 151 can block the
Immunotherapy
proliferative effects against all staphylococcal
and streptococcal SAgs and antipeptide Ab Von Behring and Kitasato first established
can protect passively against toxic shock in a immunotherapy by demonstrating that pas-
rabbit model (46). However, a subsequent study sive transfer of Abs from immunized animals
indicated that these peptides were not effective could protect nonimmune animals against
in blocking T-cell activation, cytokine produc- diphtheria. Before the discovery of sulfon-
tion, and SEB-induced toxic shock in HLA class amide in the 1930s, serum therapy was a
II transgenic mice as well as human T lympho- common option to treat infectious diseases
cytes in vitro (47). Recently, another study (54). Serum therapy remains the only prophy-
demonstrated inhibition using dodecapeptide lactic and therapeutic option against many
P72, which does not bind to MHC class II (48). toxin-mediated and viral diseases. Toxins are
312 FRIES AND VARSHNEY

usually structurally distinct from the self- ever, would be preferable, as it prevents rather
antigens expressed by the host cells and than disrupts the cytokine induction.
therefore safe targets for Ab therapy. One Epidemiological data is consistent with the
problem is that most investigators only notion that Abs matter because older patients
screened monoclonal Abs (MAbs) alone and and healthy blood donors are more likely to
characterized them as protective, indifferent, exhibit Abs against SEB- and TSST-1-induced
and disease enhancing. This approach neglects shock and recurrence is more common in
the fact that naturally occurring Ab responses younger patients who do not have Abs (34).
are complex polyclonal mixtures of Abs. Titers in patients vary and predict susceptibil-
However, in part, this simplified approach is ity to presumed toxin-mediated disease. In
chosen because the interaction between mul- addition, investigations with serum from
tiple Abs and toxins is complex and not easily healthy blood donors demonstrated that im-
predictable, let alone reproducible. Thus, FDA munoglobulin counteracted SEB stimulation
approval would be more difficult to obtain for in T-cell assays (57).
mixtures of Abs. Currently, 39 MAbs are
licensed by the FDA for human use for diverse
Vaccine Data
indications, in addition to several polyclonal
sera from diverse sources. Based on decades of Several SEB vaccines have been tested in the
successful use, Abs are considered valuable past. The U.S. Army Medical Research Insti-
candidates for novel drug development be- tute first started but ultimately abandoned the
cause of their unique pharmacological quali- development of a vaccine to SEB using forma-
ties and safety profiles. lin-inactivated SEB toxin. SEB toxoid can be
generated by prolonged incubation of SEB in
formalin at pH 7.5. Despite retained immuno-
Epidemiological and Clinical Evidence
genicity and its ability to induce protective Abs
Since 11 September 2001, the development of in monkeys and rabbits, repeated oral doses of
Abs to neutralize toxins that could potentially SEB toxoid proved to be poor mucosal immu-
be used in biological warfare has substantially nogens and were thus not efficacious against
increased, specifically for toxins like ricin, the enteric ill effects of orally given SEB. Later,
anthrax, Shiga toxin, pertussis, and SEB (55). SEB toxoids containing a nontoxic biodegrad-
For SEB, this research is highly justified able adjuvant, poly (DL-lactide-co-glycolide)
because both clinical and experimental data microspheres or proteasomes were shown
strongly support the concept that immuno- to be capable of inducing long-lasting, high-
globulins can be used to treat SEB-mediated titered Abs. This toxoid-elicited immunity
disease. SEBILS in animals and humans in- promotes neutralization of toxin in vivo and
volve induction of several proinflammatory aborts lethality in mice and rhesus monkeys
cytokines, including, e.g., TNF-. Passive im- (58, 59). Mucosal vaccination with attenuated
munization with neutralizing anti-TNF- recombinant SEB vaccine in conjunction with
MAb can prevent SEB-induced lethality. Al- cholera toxin was explored in mice and non-
though this establishes for TNF- a pivotal role human primate models and shown to be
in SEB-mediated disease (10), this MAb does effective after challenge with wild-type SEB
not neutralize SEB but only the effects of SEB toxin (27, 60, 61). Mice immunized intranasally
induction. Similarly, Abs to costimulatory were fully protected against a lethal dose of
molecules, like anti-B7.2 MAbs, significantly wild-type SEB, whereas partial (75%) protec-
inhibited T-cell activation by lowering system- tion was seen when mice were immunized
ic IL-2 release, blastogenesis, and IL-2 recep- intragastrically. Site-directed mutagenesis of
tor expression and thus improved SEBILS conserved receptor-binding surfaces of SEA
survival in mice (56). Specific therapy, how- and SEB has been employed to generate
CHAPTER 18 Bacterial ToxinsStaphylococcal Enterotoxin B 313

toxoids for vaccination. Key amino acid resi- their ultimate neutralization capacity is dif-
dues involved in binding to the TcR V chain ficult to judge (21, 56, 63, 64). Table 2 sum-
(N23) and MHC class II (F44) were substitut- marizes the list of MAbs generated against
ed. Amino acid substitutions result in toxins SEB toxin.
with reduced SAg activity (61). A SEB triple Four murine MAbs, B334 (IgG1), B327
mutant with three critical amino acid substi- (IgG2b), B87 (IgG1), and 2B33 (IgG1), which
tutions in the MHC class II binding portion exhibit nanomolar range affinity have been
(L45, Y89) and TcR V chain-binding portion described, and they recognized different, non-
(Y94) also manifests reduced T-cell activation overlapping epitopes to SEB (64). Of those,
without altering the structure of the Ag (62). B87 and 2B33 inhibited the KS-6.1 (V8.2) T-
This toxoid was also explored as a vaccine cell response to SEB. To our knowledge, these
candidate in piglets where it induced an MAbs have not been further tested in in vivo
adequate Ab response even without the addi- models.
tion of the cholera toxin adjuvant. In summary, A neutralizing murine anti-TSST-1 MAb
results of vaccine studies underscore the (MAb5) which cross-reacts with SEB was
importance of the humoral immune response evaluated for neutralization of SEB-induced
and encourage efforts to generate Abs for superantigenic activities in vitro (63). The Ab
passive immunotherapy (62). was found to partially inhibit SEB-induced T-
cell mitogenesis (63%) and TNF secretion
(70%) in human PBMCs. Epitope mapping
Passive Immunotherapy
revealed that this Ab bound to TSST-1 residues
Murine MAb 47 to 56 (47FPSPYYSPAF56) and to SEB
Several SEB-specific murine MAbs have residues 83 to 92 (83DVFGANYYYQ92), se-
been described in the literature. However, quences that are structurally dissimilar. These
the majority of them have not been rigor- studies were also not analyzed further in in
ously tested in animal models, and thus, vivo models.

TABLE 2 List of MAbs generated against SEB toxins


Afnity
Source Ab(s) unit(s) In vitro model(s) In vivo model(s) Reference

Murine lFD7, 2DA3 and 2HA10, BALB/c 72


2EG5 and 2GD9 splenocytes
Murine B334, B327, B87, 2B33 nM Mouse V8.2 + 64
T cell (KS-6.1)
Murine MAb5 (anti-TSST MAb) Human PBMCs 63
Chicken IgY BALB/c Rhesus monkeys 66
Murine phage display Soluble SEB-ScFv nM 73

Chimeric Ch82M and Ch63 pM HLA-DR3 mouse 67


splenocytes
Human 10 Fab and 4 full-length nM Human PBMCs BALB/c 69
MAbs
Human Human MAb79G9, human pMnM Human PBMCs BALB/c 70
MAb154
Murine 20B1, 14G8, 6D3, 4C7 Human T cells BALB/c, 21
HLA-DR3
Chimeric + lovastatin Ch82M and Ch63 with BALB/c mouse HLA-DR3 68
lovastatin splenocytes
Murine 3F3 (anti-SEA and -SEB) 74
Synthetic human MAb IgG 075, IgG 079, IgG 079-P, nM Human PBMCs BALB/c 71
IgG 119, IgG 120, IgG 121
314 FRIES AND VARSHNEY

Recently, four murine MAbs (20B1, 14G8, Chimeric and Human Abs
6D3, and 4C7) specific to SEB were investigat- In the more recent era of Ab development,
ed, and three of four MAbs showed significant construction of chimeric and humanized Abs
inhibition of SEB-induced T-cell proliferation have been aggressively pursued, as they can be
as well as IL-2 and IFN- production by human expected to be less immunogenic and can
T cells in vitro (21). These MAbs bind to potentially also confer human constant region
different conformational epitopes that are function. A group of investigators generated
destroyed by deletion of the distal C terminus chimeric human-mouse SEB-specific Abs (67).
of SEB. In spite of inhibition of T-cell prolif- Two good candidates were identified and
eration in vitro, these MAbs differed in further investigated. At all SEB concentrations,
protective efficacy in a SEBILS mouse model. significant neutralization of SEB-induced T-
MAbs 14G8 and 4C7 were not effective in in cell proliferation (human and mouse) was
vivo BALB/c and HLA-DR3 mouse models. achieved with the chimeric Abs Ch82M and
MAb 20B1 was 100% protective in both mouse Ch63. Interestingly, improved neutralization
models, whereas 6D3 was partially protective between the combination of anti-SEBs and
in the BALB/c model but nonprotective in the either Ab used alone was also noted. These
HLA-DR3 model. In addition, enhanced pro- chimeric Abs manifested affinities in the
tection against SEBILS was demonstrated picomolar range. The chimeric Abs have also
when two nonprotective MAbs, such as 14G8 been tested in HLA-DR3 transgenic mice and
and 6D3, were combined in vivo even if they achieved partial protection with one and
were less protective or nonprotective in complete protection with a combination of
monotherapy in the HLA-DR3 model. This MAbs. In addition, these chimeric Abs were
study was important, as it demonstrated the also shown to be effective both in vitro and in
superiority of combination therapy, possibly vivo when combined with lovastatin (68).
because of altered toxin clearance via Fc More-recent work has focused on develop-
receptor-mediated uptake. ing human MAbs to SEB. The inhibitory and
The SEB-neutralizing MAb-20B1 has also biophysical properties of 10 human Fabs,
been shown to be an effective treatment in derived by panning after vaccination with
methicillin-resistant S. aureus infection in STEBVax, were examined. These Fabs ex-
three mouse models. Administration of mAb- hibited binding affinities equal to polyclonal
20B1 protects mice from lethal sepsis and IgG, had low-nanomolar 50% inhibitory con-
reduces invasion of skin tissue and deep centrations against SEB in cell culture assays,
abscess formation. This study demonstrates and partially protected mice from SEBILS.
further evidence for the role of SEB in S. This study used an LPS-potentiated model
aureus infections and a rationale for anti-SEB with fairly low doses of SEB (2.5 g). Fabs
IgG as an immunotherapeutic agent for treat- also bound to SEC1 and SEC2 as well as
ment of severe staphylococcal infections (65). streptococcal pyrogenic exotoxin C. Four
Fabs against SEB, with the lowest 50% inhib-
Chicken itory concentrations, were converted into
SEB-specific Abs generated in chickens (IgY) native full-length MAbs. Of note is that a 250-
successfully inhibited SEB-induced T-cell pro- fold-greater inhibition of SEB-induced T-cell
liferation and cytokine responses in vitro and activation was observed with two MAbs than
in passive transfer-protected mice. Rhesus with their respective Fab fragments, which had
monkeys were also protected from lethal SEB equal binding affinities (69). SEB-specific fully
aerosol exposure when treated with the IgY human MAbs were also generated using the
specific for SEB up to 4 h after challenge. The human MORPHO-DOMA technology after
advantage of chicken Abs would be substantial isolating B cells from healthy donors whose
cost savings (66). sera showed preexisting high immune reactiv-
CHAPTER 18 Bacterial ToxinsStaphylococcal Enterotoxin B 315

ity to SEB. Human MAb 154 showed an importantly, to improve the management of
inhibitory effect on SEB-induced secretion of SAg-associated diseases. In this regard, care
proinflammatory cytokines specifically tested will have to be taken to monitor for the
for IFN- and TNF- by human PBMCs and emergence of SEB variants that may not be
protected mice prophylactically from a chal- effectively neutralized by all Abs.
lenge of up to 100 g of SEB injected i.p.
potentiated by LPS (70).
In addition, using phage display technology, ACKNOWLEDGMENTS
human Ag-binding fragments have been syn- We thank Kaushik Dutta for generating the
thesized and converted into fully human IgG figure.
Ab. These synthetic human MAbs display BCF was supported by NIH grant U54-
affinities in the nanomolar range. They were AI057158 and in part by an award from Pfizers
effective at a dose of 200 g in the murine Centers for Therapeutic Innovation, New York.
SEBILS model using different challenge doses Conflicts of interest: We disclose no conflicts.
of SEB (71).

CITATION
FUTURE CHALLENGE
Fries BC, Varshney AK. 2013. Bacterial
toxinsstaphylococcal enterotoxin B. Micro-
Current data from various studies are encour-
biol Spectrum 1(2):AID-0002-2012
aging and predict that neutralization of SAgs
like SEB is feasible. Future developments
should focus on developing high-affinity REFERENCES
MAbs that exhibit good pharmacokinetic
1. Fleischer B. 1994. Superantigens. APMIS 102:3
parameters and will permit treatment with
12.
lower doses. Several studies indicate that 2. Wilson GJ, Seo KS, Cartwright RA, Connelley T,
combination of MAbs may result in more Chuang-Smith ON, Merriman JA, Guinane CM,
potent neutralization. A major challenge is to Park JY, Bohach GA, Schlievert PM, Morrison
define an adequate setting in which these Abs WI, Fitzgerald JR. 2011. A novel core genome-
encoded superantigen contributes to lethality of
can be tested, as patients with toxic shock are
community-associated MRSA necrotizing pneu-
difficult to identify, let alone provide consent monia. PLoS Pathog 7:e1002271.
for a study. The FDA has recently approved 3. Grumann D, Scharf SS, Holtfreter S, Kohler C,
raxibacumab to treat inhalational anthrax. Steil L, Engelmann S, Hecker M, Volker U,
This is the first MAb approved using the Broker BM. 2008. Immune cell activation by
animal efficacy rule. So one option would be enterotoxin gene cluster (egc)-encoded and non-
egc superantigens from Staphylococcus aureus.
to test these MAbs only in nonhuman primate J Immunol 181:50545061.
models, which is possible under the new FDA 4. Franz DR, Parrott CD, Takafuji ET. 1997. The
animal efficacy rule. Another option would be U.S. biological warfare and biological defense
to better define the SEB-induced allergic programs, p 425436. In Sidell FR, Takafuji E,
Franz DR (ed), Textbook of Military Medicine. Part
syndromes that could constitute human
I. Warfare, Weaponry and the Casualty, vol 3. U.S.
patients to test the effect of neutralizing Abs. Government. Printing Office, Washington, DC.
Another challenge will be to identify the 5. Iandolo JJ, Shafer WM. 1977. Regulation of
selective pressures on bacteria expressing staphylococcal enterotoxin B. Infect Immun 16:
SAgs and how the selective pressures influ- 610616.
ence the interaction between SAg-bearing 6. Swaminathan S, Furey W, Pletcher J, Sax M.
1992. Crystal structure of staphylococcal entero-
bacteria and the human immune system. This toxin B, a superantigen. Nature 359:801806.
knowledge may prove invaluable to prevent 7. Papageorgiou AC, Tranter HS, Acharya KR.
emerging diseases mediated by SAgs and, most 1998. Crystal structure of microbial superantigen
316 FRIES AND VARSHNEY

staphylococcal enterotoxin B at 1.5 A resolution: 19. Krakauer T, Buckley MJ, Fisher D. 2010.
implications for superantigen recognition by Proinflammatory mediators of toxic shock and
MHC class II molecules and T-cell receptors. their correlation to lethality. Mediators Inflamm
J Mol Biol 277:6179. 2010:517594.
8. Kappler JW, Herman A, Clements J, Marrack P. 20. Huzella LM, Buckley MJ, Alves DA, Stiles BG,
1992. Mutations defining functional regions of Krakauer T. 2009. Central roles for IL-2 and
the superantigen staphylococcal enterotoxin B. MCP-1 following intranasal exposure to SEB: a
J Exp Med 175:387396. new mouse model. Res Vet Sci 86:241247.
9. Faulkner L, Cooper A, Fantino C, Altmann DM, 21. Varshney AK, Wang X, Cook E, Dutta K, Scharff
Sriskandan S. 2005. The mechanism of superantigen- MD, Goger MJ, Fries BC. 2011. Generation,
mediated toxic shock: not a simple Th1 cytokine characterization, and epitope mapping of neutral-
storm. J Immunol 175:68706877. izing and protective monoclonal antibodies
10. Miethke T, Wahl C, Heeg K, Echtenacher B, against staphylococcal enterotoxin B-induced
Krammer PH, Wagner H. 1992. T cell-mediated lethal shock. J Biol Chem 286:97379747.
lethal shock triggered in mice by the superantigen 22. Roy CJ, Warfield KL, Welcher BC, Gonzales RF,
staphylococcal enterotoxin B: critical role of Larsen T, Hanson J, David CS, Krakauer T,
tumor necrosis factor. J Exp Med 175:9198. Bavari S. 2005. Human leukocyte antigen-DQ8
11. Hewitt CR, Lamb JR, Hayball J, Hill M, Owen transgenic mice: a model to examine the toxicity
MJ, OHehir RE. 1992. Major histocompatibility of aerosolized staphylococcal enterotoxin B. Infect
complex independent clonal T cell anergy by Immun 73:24522460.
direct interaction of Staphylococcus aureus 23. Wang X, Wang BR, Zhang XJ, Duan XL, Guo
enterotoxin B with the T cell antigen receptor. X, Ju G. 2004. Fos expression in the rat brain
J Exp Med 175:14931499. after intraperitoneal injection of Staphylococcus
12. Varshney AK, Mediavilla JR, Robiou N, Guh A, enterotoxin B and the effect of vagotomy. Neuro-
Wang X, Gialanella P, Levi MH, Kreiswirth BN, chem Res 29:16671674.
Fries BC. 2009. Diverse enterotoxin gene pro- 24. McCormick JK, Bohach GA, Schlievert PM.
files among clonal complexes of Staphylococcus 2003. Pyrogenic, lethal, and emetic properties of
aureus isolates from the Bronx, New York. Appl superantigens in rabbits and primates. Methods
Environ Microbiol 75:68396849. Mol Biol 214:245253.
13. Kohler PL, Greenwood SD, Nookala S, Kotb M, 25. Strandberg KL, Rotschafer JH, Vetter SM,
Kranz DM, Schlievert PM. 2012. Staphylococcus Buonpane RA, Kranz DM, Schlievert PM.
aureus isolates encode variant staphylococcal 2010. Staphylococcal superantigens cause lethal
enterotoxin B proteins that are diverse in super- pulmonary disease in rabbits. J Infect Dis 202:
antigenicity and lethality. PLoS One 7:e41157. 16901697.
14. Krakauer T. 1994. Cell adhesion molecules 26. Bi S, Das R, Zelazowska E, Mani S, Neill R,
are co-receptors for staphylococcal enterotoxin Coleman GD, Yang DC, Hammamieh R, Shupp
B-induced T-cell activation and cytokine produc- JW, Jett M. 2009. The cellular and molecular
tion. Immunol Lett 39:121125. immune response of the weanling piglet to staphy-
15. Matsuyama S, Koide Y, Yoshida TO. 1993. HLA lococcal enterotoxin B. Exp Biol Med (Maywood)
class II molecule-mediated signal transduction 234:13051315.
mechanism responsible for the expression of 27. Boles JW, Pitt ML, LeClaire RD, Gibbs PH,
interleukin-1 beta and tumor necrosis factor- Torres E, Dyas B, Ulrich RG, Bavari S. 2003.
alpha genes induced by a staphylococcal super- Generation of protective immunity by inactivated
antigen. Eur J Immunol 23:31943202. recombinant staphylococcal enterotoxin B vac-
16. Vabulas R, Bittlingmaier R, Heeg K, Wagner H, cine in nonhuman primates and identification of
Miethke T. 1996. Rapid clearance of the bacterial correlates of immunity. Clin Immunol 108:5159.
superantigen staphylococcal enterotoxin B in 28. Hodoval LF, Morris EL, Crawley GJ, Beisel WR.
vivo. Infect Immun 64:45674573. 1968. Pathogenesis of lethal shock after intrave-
17. Stiles BG, Bavari S, Krakauer T, Ulrich RG. 1993. nous staphylococcal enterotoxin B in monkeys.
Toxicity of staphylococcal enterotoxins potenti- Appl Microbiol 16:187192.
ated by lipopolysaccharide: major histocompati- 29. Alber G, Scheuber PH, Reck B, Sailer-Kramer
bility complex class II molecule dependency and B, Hartmann A, Hammer DK. 1989. Role of
cytokine release. Infect Immun 61:53335338. substance P in immediate-type skin reactions
18. Khan AA, Priya S, Saha B. 2009. IL-2 regulates induced by staphylococcal enterotoxin B in unsen-
SEB induced toxic shock syndrome in BALB/c sitized monkeys. J Allergy Clin Immunol 84:880
mice. PLoS One 4:e8473. 885.
CHAPTER 18 Bacterial ToxinsStaphylococcal Enterotoxin B 317

30. Rusnak JM, Kortepeter M, Ulrich R, Poli M, 42. Hosotsubo KK, Hosotsubo H, Nishijima MK,
Boudreau E. 2004. Laboratory exposures to Nishimura M, Taenaka N, Yoshiya I. 1989. Rapid
staphylococcal enterotoxin B. Emerg Infect Dis screening for Staphylococcus aureus infection
10:15441549. by measuring enterotoxin B. J Clin Microbiol 27:
31. Todd J, Fishaut M, Kapral F, Welch T. 1978. 27942798.
Toxic-shock syndrome associated with phage- 43. Azuma K, Koike K, Kobayashi T, Mochizuki T,
group-I staphylococci. Lancet ii:11161118. Mashiko K, Yamamoto Y. 2004. Detection of
32. Hajjeh RA, Reingold A, Weil A, Shutt K, circulating superantigens in an intensive care unit
Schuchat A, Perkins BA. 1999. Toxic shock population. Int J Infect Dis 8:292298.
syndrome in the United States: surveillance 44. Komisar JL, Weng CF, Oyejide A, Hunt RE,
update, 1979 1996. Emerg Infect Dis 5:807810. Briscoe C, Tseng J. 2001. Cellular and cytokine
33. Gaventa S, Reingold AL, Hightower AW, responses in the circulation and tissue reactions in
Broome CV, Schwartz B, Hoppe C, Harwell J, the lung of rhesus monkeys (Macaca mulatta)
Lefkowitz LK, Makintubee S, Cundiff DR, pretreated with cyclosporin A and challenged
et al. 1989. Active surveillance for toxic shock with staphylococcal enterotoxin B. Toxicol Pathol
syndromeintheUnitedStates,1986.RevInfectDis11 29:369378.
(Suppl1):S28S34. 45. Arad G, Levy R, Hillman D, Kaempfer R.
34. Andrews MM, Parent EM, Barry M, Parsonnet 2000. Superantigen antagonist protects against
J. 2001. Recurrent nonmenstrual toxic shock lethal shock and defines a new domain for T-cell
syndrome: clinical manifestations, diagnosis, and activation. Nat Med 6:414421.
treatment. Clin Infect Dis 32:14701479. 46. Visvanathan K, Charles A, Bannan J, Pugach P,
35. Stolz SJ, Davis JP, Vergeront JM, Crass BA, Kashfi K, Zabriskie JB. 2001. Inhibition of
Chesney PJ, Wand PJ, Bergdoll MS. 1985. bacterial superantigens by peptides and anti-
Development of serum antibody to toxic shock bodies. Infect Immun 69:875884.
toxin among individuals with toxic shock syn- 47. Rajagopalan G, Sen MM, David CS. 2004. In
drome in Wisconsin. J Infect Dis 151:883889. vitro and in vivo evaluation of staphylococcal
36. John CC, Niermann M, Sharon B, Peterson ML, superantigen peptide antagonists. Infect Immun
Kranz DM, Schlievert PM. 2009. Staphylococcal 72:67336737.
toxic shock syndrome erythroderma is associated 48. Wang S, Li Y, Xiong H, Cao J. 2008. A broad-
with superantigenicity and hypersensitivity. Clin spectrum inhibitory peptide against staphylococ-
Infect Dis 49:18931896. cal enterotoxin superantigen SEA, SEB and SEC.
37. Nissen D, Pedersen LJ, Skov PS, Vejlsgaard GL, Immunol Lett 121:167172.
Poulsen LK, Jarlov JO, Karlsmark T, Nolte H. 49. Buonpane RA, Churchill HR, Moza B, Sundberg
1997. IgE-binding components of staphylococcal EJ, Peterson ML, Schlievert PM, Kranz DM.
enterotoxins in patients with atopic dermatitis. 2007. Neutralization of staphylococcal enterotox-
Ann Allergy Asthma Immunol 79:403408. in B by soluble, high-affinity receptor antagonists.
38. Tada J, Toi Y, Akiyama H, Arata J, Kato H. 1996. Nat Med 13:725729.
Presence of specific IgE antibodies to staphylo- 50. Krakauer T, Buckley M. 2003. Doxycycline is
coccal enterotoxins in patients with atopic der- anti-inflammatory and inhibits staphylococcal
matitis. Eur J Dermatol 6:552554. exotoxin-induced cytokines and chemokines.
39. Savinko T, Lauerma A, Lehtimaki S, Gombert Antimicrob Agents Chemother 47:36303633.
M, Majuri ML, Fyhrquist-Vanni N, Dieu- 51. Krakauer T, Stiles BG. 1999. Pentoxifylline
Nosjean MC, Kemeny L, Wolff H, Homey B, inhibits superantigen-induced toxic shock and
Alenius H. 2005. Topical superantigen exposure cytokine release. Clin Diagn Lab Immunol 6:
induces epidermal accumulation of CD8+ T cells, a 594598.
mixed Th1/Th2-type dermatitis and vigorous 52. Krakauer T, Buckley M. 2006. Dexamethasone
production of IgE antibodies in the murine attenuates staphylococcal enterotoxin B-induced
model of atopic dermatitis. J Immunol 175:8320 hypothermic response and protects mice from
8326. superantigen-induced toxic shock. Antimicrob
40. Bozek A, Fisher A, Filipowska B, Mazur B, Agents Chemother 50:391395.
Jarzab J. 2012. Clinical features and immunolog- 53. Krakauer T, Buckley M, Issaq HJ, Fox SD. 2010.
ical markers of atopic dermatitis in elderly patients. Rapamycin protects mice from staphylococcal
Int Arch Allergy Immunol 157:372378. enterotoxin B-induced toxic shock and blocks
41. Kientz CE, Hulst AG, Wils ER. 1997. Determina- cytokine release in vitro and in vivo. Antimicrob
tion of staphylococcal enterotoxin B by on-line Agents Chemother 54:11251131.
(micro) liquid chromatography-electrospray mass 54. Casadevall A. 2006. The third age of antimicrobial
spectrometry. J Chromatogr A 757:5164. therapy. Clin Infect Dis 42:14141416.
318 FRIES AND VARSHNEY

55. Chow SK, Casadevall A. 2012. Monoclonal infections. J Infect Dis Aug 6. [Epub ahead of
antibodies and toxins-a perspective on function print]. doi:10.1093/infdis/jit421
and isotype. Toxins (Basel) 4:430454. 66. LeClaire RD, Hunt RE, Bavari S. 2002. Protec-
56. Muraille E, De Smedt T, Urbain J, Moser M, tion against bacterial superantigen staphylococcal
Leo O. 1995. B7.2 provides co-stimulatory func- enterotoxin B by passive vaccination. Infect
tions in vivo in response to staphylococcal entero- Immun 70:22782281.
toxin B. Eur J Immunol 25:21112114. 67. Tilahun ME, Rajagopalan G, Shah-Mahoney N,
57. LeClaire RD, Bavari S. 2001. Human antibodies Lawlor RG, Tilahun AY, Xie C, Natarajan K,
to bacterial superantigens and their ability to Margulies DH, Ratner DI, Osborne BA, Goldsby
inhibit T-cell activation and lethality. Antimicrob RA. 2010. Potent neutralization of staphylococcal
Agents Chemother 45:460463. enterotoxin B by synergistic action of chimeric
58. Tseng J, Komisar JL, Trout RN, Hunt RE, Chen antibodies. Infect Immun 78:28012811.
JY, Johnson AJ, Pitt L, Ruble DL. 1995. Humoral 68. Tilahun ME, Kwan A, Natarajan K, Quinn M,
immunity to aerosolized staphylococcal entero- Tilahun AY, Xie C, Margulies DH, Osborne
toxin B (SEB), a superantigen, in monkeys vacci- BA, Goldsby RA, Rajagopalan G. 2011. Chimeric
nated with SEB toxoid-containing microspheres. anti-staphylococcal enterotoxin B antibodies and
Infect Immun 63:28802885. lovastatin act synergistically to provide in vivo
59. Lowell GH, Kaminski RW, Grate S, Hunt RE, protection against lethal doses of SEB. PLoS One 6:
Charney C, Zimmer S, Colleton C. 1996. Intrana- e27203.
sal and intramuscular proteosome-staphylococcal 69. Larkin EA, Stiles BG, Ulrich RG. 2010. Inhibition
enterotoxin B (SEB) toxoid vaccines: immunoge- of toxic shock by human monoclonal antibodies
nicity and efficacy against lethal SEB intoxication against staphylococcal enterotoxin B. PLoS One
in mice. Infect Immun 64:17061713. 5:e13253.
60. Stiles BG, Garza AR, Ulrich RG, Boles JW. 2001. 70. Drozdowski B, Zhou Y, Kline B, Spidel J, Chan
Mucosal vaccination with recombinantly attenu- YY, Albone E, Turchin H, Chao Q, Henry M,
ated staphylococcal enterotoxin B and protec- Balogach J, Routhier E, Bavari S, Nicolaides NC,
tion in a murine model. Infect Immun 69:2031 Sass PM, Grasso L. 2010. Generation and char-
2036. acterization of high affinity human monoclonal
61. Woody MA, Krakauer T, Stiles BG. 1997. antibodies that neutralize staphylococcal entero-
Staphylococcal enterotoxin B mutants (N23K toxin B. J Immune Based Ther Vaccines 8:9.
and F44S): biological effects and vaccine potential 71. Karauzum H, Chen G, Abaandou L, Mahmoudieh
in a mouse model. Vaccine 15:133139. M, Boroun AR, Shulenin S, Devi VS, Stavale E,
62. Ulrich RG, Olson MA, Bavari S. 1998. Develop- Warfield KL, Zeitlin L, Roy CJ, Sidhu SS, Aman
ment of engineered vaccines effective against MJ. 2012. Synthetic human monoclonal antibodies
structurally related bacterial superantigens. toward staphylococcal enterotoxin B (SEB) protec-
Vaccine 16:18571864. tive against toxic shock syndrome. J Biol Chem 287:
63. Pang LT, Kum WW, Chow AW. 2000. Inhibition 2520325215.
of staphylococcal enterotoxin B-induced lympho- 72. Lin YS, Largen MT, Newcomb JR, Rogers TJ.
cyte proliferation and tumor necrosis factor alpha 1988. Production and characterisation of mono-
secretion by MAb5, an anti-toxic shock syndrome clonal antibodies specific for staphylococcal
toxin 1 monoclonal antibody. Infect Immun 68: enterotoxin B. J Med Microbiol 27:263270.
32613268. 73. Singh PK, Agrawal R, Kamboj DV, Gupta G,
64. Hamad AR, Herman A, Marrack P, Kappler JW. Boopathi M, Goel AK, Singh L. 2010. Construc-
1994. Monoclonal antibodies defining func- tion of a single-chain variable-fragment antibody
tional sites on the toxin superantigen staphylococ- against the superantigen staphylococcal entero-
cal enterotoxin B. J Exp Med 180:615621. toxin B. Appl Environ Microbiol 76:81848191.
65. Varshney AK, Wang X, Scharff MD, Macintyre 74. Liang B, Zhang Y, Liu A, Zhou Y, Chen F, Wang
J, Zollner RS, Kovalenko OV, Martinez LR, X. 2011. Production of a monoclonal antibody by
Byrne FR, Fries BC. 2013. Staphylococcal entero- simultaneous immunization of staphylococcal en-
toxin B specific monoclonal antibody 20B1 suc- terotoxin A and B. Appl Biochem Biotechnol 164:
cessfully treats diverse Staphylococcus aureus 831840.
TECHNICAL ADVANCES
Antibody Engineering

KIN-MING LO,1 OLIVIER LEGER,2 and BJRN HOCK3


19
Monoclonal antibodies are enjoying outstanding commercial success, with 4 of
the top 10 best-selling drugs. Although most of the preclinical and clinical
experiences with therapeutic antibodies have been gained from the treatment of
cancer and inflammatory diseases, effective therapy is not limited to these
indications. For example, palivizumab (Synagis) has been approved for the
prophylactic treatment for respiratory syncytial virus (RSV) infections and
remains the only therapeutic antibody for the treatment of infectious diseases
to date.
In this article, we review the latest advances in antibody-engineering
technologies to make an antibody clinically and commercially successful. We
discuss the rationale behind the choice of immunoglobulin isotype and the protein
engineering of the constant regions. This is followed by a review of the
humanization strategy to minimize immunogenicity and the recent approaches
to affinity mature the antigen binding of the antibody. Finally, we have an
overview of the next-generation antibodies, such as antibody-drug conjugates,
bispecific antibodies, and immunocytokines, which are being developed to meet
future challenges.

1
Department of Protein Engineering and Antibody Technologies, EMD Serono Research Institute, Billerica, MA
01821; 2Department of Protein Engineering and Antibody Technologies, Merck Serono S.A.Geneva, 1202
Geneva, Switzerland; 3Department of Protein Engineering and Antibody Technologies, Merck Serono, Merck
KGaA, D-64293 Darmstadt, Germany.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0007-2012

321
322 LO ET AL.

THE CONSTANT REGIONS: PROTEIN AND Therefore, a different strategy for engineering
GLYCOENGINEERING OF THE Fc antibodies against infectious agents may be
needed. Such a strategy should consider the
The Fc portion of the antibody, which deter- choice of Ig isotypes and subclasses best suited
mines its effector functions and is respons- for the disease, the glycosylation pattern to
ible for its pharmacokinetic properties, has facilitate the desired mode of action, and the
to be carefully chosen to suit its therapeutic optimal pharmacokinetic and pharmacody-
applications. To date, all of the approved namic properties. These considerations will
therapeutic antibodies are of the immunoglob- in turn determine the expression system, host
ulin G (IgG) isotype, because IgGs are the cell lines, and production method.
predominant serum immunoglobulins and are
readily manufacturable as biotherapeutics.
Choice of Ig Isotypes and Subclasses
Furthermore, IgG binds the Fc receptors
(FcRs) on immune cells to elicit various IgA as a Potential Isotype for Therapeutic
effector functions and is the only isotype that Antibodies Against Infectious Diseases
binds the protective neonatal Fc receptor The majority of the antibodies approved for
FcRn, which gives typical IgGs (IgG1, -2, and the treatment of cancer and autoimmune
-4) their long serum half-lives of about 21 days disorders are of the IgG1 subclass. Although
in humans. isotypes other than IgG have not been suc-
Most of the knowledge gained on antibody cessfully developed, IgA presents itself as a
therapy is from the treatment of cancer and logical candidate against infectious diseases
autoimmune disorders using IgG1, the pre- because IgAs in nature provide the principal
ferred IgG subclass because its potent effector defense against pathogens, in the first line, as
functions are expected to contribute to the IgA dimers against invasion by preventing the
mechanism of action of the antibody. For attachment of microbes or toxins to the
infectious diseases, the only therapeutic anti- mucosal lining, and, in the second line, as IgA
body approved to date is palivizumab, also monomers in circulation to neutralize patho-
an IgG1, for the prophylactic treatment for gens that have breached the mucosal barrier.
RSV. The potent antibody-dependent cel- Furthermore, IgA represents the most abun-
lular cytotoxicity (ADCC) and complement- dantly produced antibody and is the second
dependent cytotoxicity (CDC) of IgG1 that are most prevalent isotype in the blood behind
desirable for eradicating cancer or autoim- IgG.
mune disease-causing cells, however, may not IgA has two subclasses, IgA1 and IgA2. IgA1
be ideal for the treatment of certain infectious with an extended hinge region may offer
diseases. In cancer therapy, the major cell higher avidity antigen binding with distantly
types mediating ADCC are NK cells and spaced antigens, but it is more vulnerable to
macrophages, which also eradicate tumor proteolysis by enzymes from pathogenic bac-
cells by antibody-dependent cellular phagocy- teria (2). IgA, especially the IgA2 isoform, has
tosis (ADCP). To maximize cellular cytotoxi- been shown to be superior to IgG1 in recruiting
city, optimal engagement of the activating human PMN as effector cells to mediate ADCC
receptor FcRIII on NK cells and macrophage (3). In addition, ADCP by human peripheral
is important. On the other hand, polymor- blood neutrophils is more effectively mediated
phonuclear (PMN) cells, which are about by IgA than by IgG (4).
10 times more abundant than monocytes and The IgA dimer format may be preferable in
NK cells, constitute the first line of defense certain disease settings. However, the devel-
against bacteria, and FcRIIA, the most widely opment of an IgA dimer is more challenging
expressed FcR, is the central initiator of IgG- than that of a monomer. Toward that end, a
mediated phagocytosis by human PMN (1). recombinant dimeric IgA against epidermal
CHAPTER 19 Antibody Engineering 323

growth factor receptor (EGFR) was produced a yeast infection (Cryptococcus neoformans,
and shown to mediate effective tumor cell which causes meningitis in AIDS patients),
killing. Importantly, the dimeric IgA, but not IgG2 and IgG4, which have few ADCC/ADCP
the monomeric IgA or IgG, was directionally and CDC activities, respectively, protected the
transported by the polymeric Ig receptor mice, whereas for the effector functions com-
through an epithelial cell monolayer in vitro petent counterparts, IgG3 had no effect and
(5). This has important implications for the IgG1 actually potentiated the infection (8). This
IgA dimer as a potential anti-infectious agent unexpected result was explained by the obser-
at the mucosa. vation that phagocytosed yeast was not killed
to a significant extent, and the macrophages
can possibly act as a Trojan horse for its
IgG and its Subclasses
survival and dissemination (9). Interestingly,
IgG1 studies in complement-deficient mice sug-
The human IgG1 subclass was chosen almost a gested that complement may also play a role
priori for therapeutic antibodies. Besides being in the antibody-mediated enhancement of
the one that is most abundant in circulation, it cryptococcal infection (10).
has potent ADCC and CDC activities, and also
the most desirable chemical, manufacturing, IgG2
and control (CMC) properties as a biothera- Although IgG2 is often considered as the
peutic, e.g., it does not have the problems that effector-silent isotype in cancer treatment, it
IgG2 and IgG4 have (see below). Most of the should be noted that the principal effector cells
experience with therapeutic IgG1 antibodies in the human peripheral blood mononuclear
has been for the treatment of cancer, where cell used in the classical in vitro ADCC assay
the importance of CDC to the mechanism are NK cells, which express only FcRIII. In
of action is controversial. Based on studies this assay, IgG1 and IgG3 are the most potent,
in complement-deficient mice, it has been while IgG2 and IgG4 have no detectable
shown that the contribution of the comple- activity.
ment pathway to proinflammatory responses However, it has been reported that IgG2
initiated by IgG is minor (6). However, com- can trigger ADCC by neutrophils mediated
plement plays an important role in the phago- by FcRIIA (11). This finding may explain
cytosis of opsonized bacteria mediated by the the discrepancy between the lack of in vitro
complement receptors (7). Therefore, we need ADCC using NK cells, and yet readily detect-
to take a critical look to engineer the most able cytolytic activities of anti-CD8 IgG2 and
effective human isotype to provide protection IgG4 in mice (12), and, more importantly, the
against infectious agents because of the differ- cytolytic activity of alemtuzumab (Campath)
ent biology involved. IgG4 in humans (13, 14). In addition, although
IgG1 binds all three classes of FcR. Nor- complement plays a role in the phagocytosis of
mally, FcRI is saturated with serum IgG, opsonized bacteria mediated by the comple-
which explains why this high-affinity FcR ment receptors, IgG2 engagement of FcRIIA
may play only a minor role in IgG-mediated alone on phagocytic cells can lead to phagocy-
cellular responses such as ADCC and ADCP (6). tosis of opsonized bacteria (7).
The binding of IgG1 to the inhibitory FcRIIB IgG2 exhibits disulfide structural heteroge-
and the glycosylphosphatidylinositol-anchored neity, resulting in at least three isoforms which
FcRIIIB, which does not signal, may render differ by the disulfide connectivity at the hinge
IgG1 suboptimal in triggering effector functions region (15). This can be more than a CMC issue
by PMN (4). Surprisingly, in a mouse study on the homogeneity of the product, since some
designed to compare the efficacies of human of the isoforms have been shown to affect
IgG subclasses with identical V regions against binding affinity (16). IgG2 hinge region homo-
324 LO ET AL.

geneity with uniform disulfide bonds can be (24). Indeed, natalizumab showed significant
readily achieved by the use of a modified IgG1 levels of Fab-arm exchange in 15 of 16 patients,
hinge (17). as early as 1 h after infusion (23). There-
fore, therapeutic IgG4 antibodies should be
IgG3 designed to prevent the Fab-arm exchange by
IgG3 has an extended hinge region that introducing the S228P mutation to stabilize
imparts greater segmental flexibility and mo- the hinge region, and, more importantly, the
tion, resulting in a larger angle and longer R409 K mutation to stabilize the CH3 region
distance between the two binding sites, to (25). On the other hand, this Fab-arm ex-
attain bivalent binding (18). It is for this reason change property of IgG4 can be exploited to
that the IgG3 subclass was chosen for the generate recombinant bispecific therapeutic
development of a class of antibody cytokine antibody, such as the Duobody of Genmab
fusion proteins (19). This extra spacing and (Utrecht, The Netherlands).
flexibility provided by the IgG3 hinge may be
particularly relevant for antiviral antibodies,
Fc Glycosylation and Effector Functions
because viruses such as the human immuno-
deficiency virus (HIV) have envelope spikes The N-glycan in the Fc is essential for the
that are few and far between (20). Such spacing engagement of the FcRs (but not FcRn) and
between epitopes is a strategy HIV has evolved hence important for effector functions. There-
to avoid bivalent binding to thwart neutral- fore, antibodies whose effector functions are
ization by antibodies. For development as a important for their mechanisms of action have
biotherapeutic, IgG3 suffers from the fact that to be produced in mammalian cells, or, more
it has a relatively short serum half-life of about recently, in glycoengineered yeast (26). Im-
7 days in humans. portantly, the nature of the N-glycan chain,
such as the presence of the bisecting N-
IgG4 acetylglucosamine (GlcNAc), fucose, and ter-
The IgG4 subclass is usually chosen for its minal sialic acids, modulates the activation of
complete lack of CDC, low ADCC (despite its the FcRs and immune cells.
high binding affinity for FcRI [21]), and anti-
inflammatory activity (22). The hinge region of Bisecting GlcNAc and Fucosylation
the IgG4 H chain has two cysteine residues Impact ADCC
that can form either an intrachain disulfide It was recognized early on that the glycosylation
bond or two interchain disulfide bonds with its pattern and biological activity of alemtuzumab
partner H chain, and this accounts for the expressed in different cell lines varied. The
presence of about 50% of half-molecules (HL, alemtuzumab produced from rat myeloma
a light chain bound to one heavy chain) when cells had a bisecting GlcNAc and a low extent
IgG4 is analyzed by sodium dodecyl sulfate-gel of fucosylation, and it showed the highest
electrophoresis under non-reducing condi- ADCC (27). Although antibodies produced in
tions. This heterogeneity is more than a CMC (1,4)-N-acetylglucosaminyltransferase III
issue because recently it has been shown that (GnTIII) transfected Chinese hamster ovary
therapeutic IgG4 antibodies can engage in (CHO) cells had improved ADCC because of the
Fab-arm exchange with endogenous human addition of the bisecting GlcNAc (28), it was
IgG4 in vivo (23), resulting in the functional recognized later that the absence of fucose was
monovalency of IgG4, with the loss of binding dominant over bisecting GlcNAc in enhancing
avidity and cross-linking effects. It is notewor- the ADCC. Indeed, the enhancement effect
thy that of the two approved therapeutic IgG4 of bisecting GlcNAc was only observed in
antibodies, only gemtuzumab contains a hinge highly fucosylated IgG1, which has low ADCC
modification, whereas natalizumab does not activity (29).
CHAPTER 19 Antibody Engineering 325

Since ADCC was considered as an impor- possible that the higher-affinity binding of
tant mechanism of action for a number of antibodies with low fucose levels to FcRIIIB
therapeutic antibodies, and NK cells, the acts as a decoy mechanism, resulting in less
effector cells used in standard ADCC assays binding to FcRIIA, the receptor responsible for
in vitro, express only FcRIIIA, technology to activating the PMN-mediated ADCC (35). This
improve FcRIIIA binding of antibodies re- shows that the impact of Fc glycosylation on
ceived a lot of attention. Furthermore, in- ADCC is critically dependent on the recruited
creased FcRIIIA binding may also enhance effector cell type, an important point to consider
antigen uptake by FcRIIIA-expressing den- when designing antibodies for the treatment of
dritic cells and macrophages, thus potentiating infectious diseases.
the adaptive immune surveillance.
The importance of ADCC to antitumor Manipulating Effector Functions and
activity was supported by clinical data, which Pharmacokinetic Properties
showed that response to antibody therapy was In addition to the aforementioned GnTIII-
positively correlated to the binding affinity of transfected CHO cells by Roche Glycart (Schlie-
FcRIIIA to IgG1. For rituximab (Rituxan) in ren, Switzerland) to produce antibodies with
the treatment of non-Hodgkins lymphoma, bisecting GlcNAc, an -1,6-fucosyltransferase
Waldenstrms macroglobulinemia, and follic- (FUT8) knockout CHO cell line was devel-
ular lymphoma, FcRIIIA polymorphism was oped by Kyowa Hakko Kogyo (Tokyo, Japan)
found to be a good predictor of clinical to produce nonfucosylated antibodies (36). Fur-
outcome, with patients expressing the 158V thermore, glyco-optimized and fully human
variant, which has a higher binding affinity glycosylated antibodies can be produced
for IgG1, responding better than patients from glycoengineered mammalian cells (Gly-
expressing the 158F variant (30, 31). Similar cotope, Berlin, Germany) or yeast (GlycoFi
positive correlation was also reported for Merck, Lebanon, New Hampshire) (25).
trastuzumab (Herceptin) in the treatment Instead of glycoengineering, Fc mutations
of metastatic breast cancer (32), and for can be introduced to optimize binding to
Erbitux in the treatment of metastatic colo- specific FcRs, such as increased binding to
rectal cancer (33). There is also direct evidence activating FcRs and decreased binding to the
that increasing the binding affinity of an inhibitory FcRIIB, thereby improving the so-
antibody for FcRIIIA translates to improved called A/I ratio, to enhance ADCC and ADCP
efficacy in the clinic. Anti-RhD monoclonal (37, 38, 39). On the other hand, for therapeutic
antibodies (mAbs) produced in YB2/0 cells, applications where effector functions are
which have low fucose levels, demonstrated undesirable, mutations to abrogate ADCC
superior clearance of RhD-positive red blood and CDC are well described in the literature.
cells, relative to monoclonal antibodies pro- One common strategy is to incorporate natu-
duced in other producer lines (34). ral motifs from IgG2 and IgG4 to decrease
It should be noted that, while antibodies with binding to FcRs and complement, respec-
low fucose levels have enhanced ADCC mediated tively, while keeping potential immunogenic-
by NK cells, they were found to be less potent in ity of the changes to a minimum. For example,
inducing ADCC mediated by activated PMN, in Armour et al. (40) replaced residues 233 to 236
comparison with antibodies with high fucose in the lower hinge region of IgG1 responsible
levels. PMN express FcRIIA, and, instead of for FcR binding with the corresponding
FcRIIIA, the glycosylphosphatidylinositol an- residues from IgG2, and introduced IgG4
chored FcRIIIB, which does not have the residues at position 327, 330, and 331 to abolish
signaling ability to trigger effector functions by binding to C1q. Similarly, the effector silent
PMN. Since fucosylation levels on antibodies are IgG2/G4 composite antibody of Mueller et al
not known to affect binding to FcRIIA, it is (41) and the IgG2m4 of An et al. (42) were all
326 LO ET AL.

based on incorporating the nonbinding motifs endothelial and epithelial cells, but also found
of IgG2 and IgG4. However, as mentioned in human monocytes and dendritic cells. FcRn
earlier, incorporating natural motifs from on these immune cells can translocate to
IgG2 and IgG4 does not necessarily mean nascent phagosomes, thereby facilitating IgG-
complete elimination of binding. In fact, it was mediated bacterial phagocytosis (50).
recognized that IgG2 and IgG4 can lead to
unwanted side effects, presumably caused by Fc Sialylation
residual interactions with FcRIIA (H131) and High levels of sialylation of the Fc glycans in
FcRI (43). It should be pointed out that IgGs are associated with reduced binding to
complete removal of N-glycan greatly im- FcRIIIA on NK cells, resulting in lower
paired binding to cellular FcRs and C1q ADCC both in vitro (51) and in an in vivo
(44), without affecting the interaction with model of immune thrombocytopenia (52).
FcRn (37, 45). Therefore, an aglycosylated However, it is not clear whether this is entirely
form of IgG2 may offer the best approach to due to lower-affinity binding to FcRIIIA
effector silent antibodies (46). This is sup- specifically, or also partly due to lower binding
ported by the alemtuzumab study, which to cell surface antigens in general, because
showed that the IgG4 subclass, despite the many cell surface antigens and receptors have
lack of ADCC in vitro, had cytolytic activities terminal sialic acids (51). What is certain is
in humans, whereas the aglycosylated form that the effect of sialylation on FcR binding
had neither in vitro nor in vivo cytolytic ac- was independent of the 2,3- and 2,6-linkage of
tivities (13, 14). the sialic acid to the penultimate galactose on
In addition to optimizing the binding to the complex biantennary N-linked glycan (52).
FcRs selectively, Fc mutations were also The 2,6-linkage, however, is definitely
introduced to increase binding to FcRn at responsible for the anti-inflammatory activity
acidic pH, but not at neutral pH, thereby of intravenous immunoglobulin treatment,
enhancing the recycling of IgG from the which has been widely used to treat autoim-
endosomes back to the circulation. The affin- mune diseases such as immune thrombocyto-
ities of therapeutic IgGs and Fc-containing penia, rheumatoid arthritis, and systemic lupus
proteins for FcRn at pH 6.0 were shown to be erythematosus. Interestingly, the receptor re-
closely correlated with their serum half-lives sponsible for the binding of 2,6-sialylated IgG
from clinical studies (47). One well-character- is not one of the canonical FcRs, but a C-type
ized Fc mutant with enhanced binding to FcRn lectin receptor DC-SIGN on dendritic cells,
has the triple mutations YTE (M252Y/S254T/ resulting in the secretion of soluble anti-
T256E) introduced into the Fc portion of an inflammatory mediators that increase surface
anti-RSV monoclonal antibody, resulting in 10- expression of FcRIIB on macrophages found
fold increase in binding to human and monkey at the site of inflammation (53). Subsequent
FcRn at pH 6.0 but not pH 7.4. This translates cross-linking of FcRIIB and activating FcRs
to a fourfold improvement in serum half-life in on the macrophage by the autoantibody in the
monkeys (48). With human FcRn knock-in immunocomplex results in inhibition of mac-
mice being commercially available now, serum rophage activation, which accounts for the
half-life of therapeutic antibody candidates anti-inflammatory activity of intravenous im-
can be compared conveniently in this surro- munoglobulin. Therefore, enhancing the 2,6-
gate host (49). linkage sialylation of IgG may be an effective
It should be noted that FcRn, while pri- way of increasing anti-inflammatory activity of
marily known as a neonatal receptor and a therapeutic antibodies, e.g., in the treatment of
protective receptor for IgG, can also be infectious diseases where controlling inflam-
exploited to mediate immune functions against mation is of paramount importance. It is
infections. Its expression is not restricted to interesting to note that, despite that only 5%
CHAPTER 19 Antibody Engineering 327

of the total serum IgG is fully sialylated, innate nicity and increase their activity in the human
IgGs are generally anti-inflammatory in the immune system.
steady state; the extent of sialylation is reduced Antibody humanization comprises proven
in the course of an antigen-specific immune strategies for lessening the immunogenicity of
response so that the IgGs become more well-characterized mAbs from animal sources
proinflammatory (54). (commonly mice) and for ameliorating their
activation of the effector functions of the
Aglycosylated Fc human immune system, thus producing clini-
Production of full-length antibodies has been cal diagnostics and therapeutics. To overcome
done exclusively in eukaryotic cells because of the immunogenicity triggered by non-human
their complex tetrameric structure and the biologics, approaches such as the development
posttranslational glycosylation critical for ef- of hybridomas and B-cell cloning from anti-
fector functions. The demonstration that body-secreting cells from human peripheral
heavy and light chains can be efficiently blood and the production of human antibody
secreted and assembled in the Escherichia repertoires in transgenic mice or on the surface
coli periplasm paved the way to an inexpensive of different display systems have emerged as
method for the manufacture of therapeutic platforms for engineering fully human thera-
antibodies, especially for treatment in which peutic antibodies. However, because there are
effector functions are not desirable (55). neither royalties nor license fees attached to
Aglycosylated antibodies retain binding to the use of the hybridoma technology, the
FcRn (37) and have been shown to have development of animal (mainly mouse) mono-
normal pharmacokinetic profiles in the clinic clonal antibodies as therapeutics remains a
(56). If effector functions are desirable, point very attractive alternative providing that they
mutations can be readily introduced to restore are humanized to translate high-quality non-
FcR and complement binding, and, better human antibodies to the clinic.
still, selectively engage certain FcRs to
achieve a more favorable A/I ratio (56, 57,
CDR Grafting: Standard Technology
58), especially for the treatment of certain
infectious diseases in which enhancing phago- In the past 25 years, multiple strategies for the
cytosis is important. humanization of mAbs have been reported in
the literature. However, complementarity de-
termining region (CDR) grafting onto a ho-
HUMANIZATION mologous (high-sequence homology) human
antibody framework (acceptor) remains the
The key factor to reducing the inherent immu- most utilized technique for mAb humanization
nogenicity of a product candidate is to make (59). Strict CDR grafting usually results in
the therapeutic monoclonal antibody as simi- significant to complete loss of antigen recog-
lar as possible to naturally occurring human nition by the acceptor antibody. This problem
immunoglobulin sequences without losing the can be resolved by backmutating critical
changes that result in the desired functional framework sites on the acceptor to the corre-
properties. sponding amino acid residues in the donor
Humanized antibodies are antibodies from sequence. These critical framework residues
non-human species whose amino acid se- for back-mutation are selected from the wealth
quences have been changed to improve their of published structural data on antibodies and/
similarity to antibody variants produced natu- or from the homology-modeled structure of
rally in humans. Before rodent antibodies can the mouse Fv sequence that reveals the
be used in clinical application, it is essential to framework positions important for main-
humanize them to decrease their immunoge- taining the integrity of the Ig fold or for
328 LO ET AL.

supporting the conformation of the CDR resurfacing, is a technique whereby the ex-
(canonical residues). Figure 1 describes the posed framework residues are replaced by
main decision-making points during the CDR- amino acids found at corresponding positions
grafting process and their evolution over the in human antibody sequences. Although many
years. CDR grafting guided by molecular antibodies have been humanized by variable
modeling is a well-validated approach that domain resurfacing, to our knowledge, no
has been instrumental for the development of clinical studies with these antibodies have
several FDA-approved antibodies, such as been published until now.
Tysabri (60) and Actemra (61). For a detailed
description of the standard technology, see Grafting of Abbreviated CDRs
review by Leger and Saldanha (62). Containing SDRs
CDR-grafted humanized antibodies carry xe-
nogeneic CDRs which may evoke antivariable
Alternative Approaches
region, including anti-idiotypic, responses.
Veneering or Resurfacing Not all the CDR residues of a murine antibody
It was proposed by Padlan (63) that murine are essential for antigen binding; this rationale
variable domains could be veneered to reduce comes from a comprehensive analysis of the
their antigenicity. Veneering, also known as three-dimensional structures of the antibody

Non-human Ab sequence

Choice of region to graft

CDR HV loop SDR / MSD


Kabat structural specificity

Choice of human framework acceptor

Fixed Best fit Best fit Best fit


REI (VL) mature consensus
NEW, KOL (VH) sequences germ line

Back-mutations

Affinity Stability
1990s Today

more Non-human residues content less

CDR-grafted fully human like

FIGURE 1 Flow chart describing the main decision-making points and their evolution over the years, in a typical
CDR-grafting protocol. CDR, complementarity-determining region; HV, hypervariable loops; SDR, specicity-
determining regions; MSD, minimum specicity determinant. REI, NEW, and KOL are light or heavy variable
region sequences derived from human myeloma cell lines. doi:10.1128/microbiolspec.AID-0007-2012.f1
CHAPTER 19 Antibody Engineering 329

combining site where it was suggested that residues), and (ii) particular source framework
only 20 to 33% of the CDR residues are critical residues may, in some cases, actively partici-
in the antigen-antibody interaction (64). These pate in direct interactions with the antigen as
residues, located in the regions of high vari- has been observed in certain antigen/antibody
ability and most likely unique to each antibody, complexes.
are designated as specificity-determining res-
idues (SDRs). A new approach to the human- Human String Content Optimization
ization of antibodies, therefore, was based on Recently, Lazar et al. (67) have introduced a
grafting only the SDRs of a xenogeneic anti- new method of humanization based on a novel
body into the human antibody frameworks. and immunologically relevant metric of anti-
The immunogenicity of an antibody therefore body humanness, termed human string con-
could be reduced by transplanting only those tent, that quantifies a sequence at the level
parts of the CDRs that contain the SDRs. The of potential major histocompatibility complex/
abbreviated CDRs have been defined (65) as T-cell epitopes. Human identity is defined as
the boundaries of the potential SDRs in various the number of total human 9-mers, which is
antigen combining sites. an exact count of 9-mer stretches in the Fv that
perfectly match any one of the corresponding
Superhumanization stretches of nine amino acids in their set of
This humanization strategy is based on struc- functional human germ line sequences. Human
tural homologies between mouse and human 9-mer content is used as a measure of T-cell
CDRs and essentially ignores the frameworks. tolerance (i.e., if a stretch of nine consecutive
The first step of this CDR-grafting method is to amino acids matches one of the human germ
identify human germ line V genes that in line segments, it is more likely that T cells will
combination have the same canonical structure not recognize it because of developmental
class as the mouse antibody to be humanized. tolerance for the original human 9-mer). This
Within that matching subset, typically a half- approach utilizes the homology present in
dozen genes from 44 functional VH or 41 human germ line sequences to make murine-
functional VL genes in the human genome, H to-human substitutions that increase the
and L chain gene segments are picked whose human sequence content of the Fv.
CDRs have the best residue-to-residue homol-
ogy to the mouse antibody. In the selected Framework Shufing and Human
sequences, the remaining nonhomologous CDR Framework Adaptation
residues are simply converted to the mouse DallAcqua et al. (68) describes a new human-
antibody sequence. mAbs constructed by this ization approach called framework shuffling
strategy retain the ability to bind antigen, and, that does not require any rational design
because they are CDR grafted in a way that or structural information and for which
minimizes deviation from human sequences, there is no need to design backmutations. In
such Abs are called superhumanized (66). this method, a given non-human monoclonal
Because the choice of the human frame- antibody is humanized by synthesizing a
works is driven by the sequence and structure combinatorial library comprising its six
of the CDRs, this strategy has the potential to CDRs fused in frame to a pool of mixed and
generate humanized antibodies that retain matched human germ line frameworks. These
good binding to their cognate antigen. How- human frameworks encompassed all known
ever, the general applicability of such a heavy- and light-chain human germ line genes.
framework-ignoring strategy is uncertain Libraries were cloned into an M13-based
since (i) favorable conformation(s) of a given phage expression vector. The primary screen
CDR can be uniquely dependent on some consists of a single-point enzyme-linked im-
specific source framework residues (canonical munosorbent assay using periplasmic extracts
330 LO ET AL.

prepared from individual recombinant M13 Nishibori et al. (75) humanized a rabbit and
clones. chicken antibody, respectively.
Recently, an alternative to the FR shuffling,
human framework selection, was described
Concluding Remarks on Humanization
by Fransson et al. (69) as the first step of a new
humanization method called human frame- There is no simple correlation between se-
work adaptation. In this method, the human quence humanness and immune response, as
germ line genes are selected based on sequence illustrated by the virtual absence of immuno-
and structural considerations. This is followed genicity for the CDR-grafted humanized anti-
by a second step of SDR optimization. body trastuzumab and the 5 to 89% neutralizing
response rate in a subset of patients, which
Combinatorial Library Approaches varies depending on the disease and the
therapy, to the fully human mAb adalimumab
Guided selection (Humira) (76). Many factors in addition to
An alternative method, guided selection, has protein sequence can affect immunogenicity,
been developed to convert murine antibodies such as dose and administration parameters
into completely human antibodies with similar (route, duration, and frequencies, as well as
binding characteristics. This methodology was target-specific effects), but one of the most
originally developed by Jespers et al. (70). In critical factors is the biophysical properties of
this method, mouse VH and VL domains are the final molecule (77). These properties should
sequentially or in parallel replaced by human be carefully monitored during the humaniza-
VH and VL domains, respectively, using phage tion process.
selection to derive human antibodies with best To conclude, we propose that a mixing and
affinity. A potential disadvantage of the guided matching of techniques could be used in future
selection approach is that shuffling one or both protocols to take into consideration all the
antibody chains can result in epitope drift (71). different aspects of humanization that we
This can however be seen as an advantage for considered above with the ultimate goal of
introducing minor but essential changes in engineering fully human monoclonal anti-
specificity such as removing cross-reactivities bodies. Such an attempt has recently been
to unwanted antigens (72). To maintain the reported by Bernett et al. (78). Starting from
epitope recognized by the source non-human the source murine variable regions of three
antibody, CDRs can be conserved. In this currently marketed mAbs targeting CD25
alternative method, one or both non-human (daclizumab), vascular endothelial growth
CDR3s are commonly retained since they factor (bevacizumab), and tumor necrosis
usually play a critical role in the recognition factor alpha (infliximab), they generated
of the antigen. fully human antibodies. All three engineered
mAbs had levels of sequence humanness
Combinatorial Library Strategy comparable to other fully human mAbs and
Rosok et al. (73), using a combinatorial ap- yet maintained antigen binding and in vitro
proach and colony lift as selection method, activity comparable to the three marketed
have described a general antibody-engineering drugs.
strategy that combines elements of structure-
based approaches with in vitro evolution
strategies to address the difficulty of main- AFFINITY MATURATION
taining antibody binding activity following
humanization. More recently, also using the The antibody leads resulting from the discovery
combinatorial approach but phage display as efforts meet established criteria regarding
selection method, Rader et al. (74) and affinity and specificity. Affinity to the target
CHAPTER 19 Antibody Engineering 331

molecule defines the future monoclonal anti- than 100 pM cannot be obtained; rather, there
body drugs efficacy and influences the drug is unlikely to be actual selection in favor of
dose required to obtain the desired effective- mutations that take affinities beyond this
ness. A key requirement during antibody value. This prediction of an in vivo affinity
development is therefore to engineer a mole- ceiling has been frequently challenged. In-
cule with sufficiently high affinity to guar- deed, with the use of transgenic mice produc-
antee effective occupancy over prolonged ing fully human antibodies, the generation of
periods. On one hand, for neutralization of antibodies with subnanomolar affinities (less
soluble antigens, like cytokines, or viruses, than 10 pM) has frequently been reported (83).
the neutralization efficiency seems to be well The existence of an in vivo generated antibody
correlated with the binding affinity. On the to interleukin-8 with a subpicomolar affinity
other hand, the assessment of affinity goals (610 fM) has also been reported (84). This
for antibodies against membrane receptors is confirms that, by using efficient antibody
often challenging. generation procedures to screen a larger pro-
portion of the available immune repertoire in
hyperimmune animals, it is possible to isolate
In Vivo Afnity Maturation
antibodies with exceptionally high affinities
The affinities of antibodies obtained in vivo well beyond the proposed affinity ceiling.
tend to fall within characteristic ranges con- Generation of high-affinity antibody against
strained by biological requirements imposed T cell-dependent antigen has also been de-
during the ontogeny of B cells. Specific antigen scribed in germinal center-associated DNA
recognition by a surface antibody on a B cell, primase gene-transgenic C57BL/6 mice
followed by endocytosis, leads to efficient (GanpTg). Recent data suggest that germinal
antigen presentation (79), initiating cytokine center-associated DNA primase may serve as
release, B- and T-cell proliferation and differ- an essential link required to transport activa-
entiation, somatic hypermutation, and so on. tion-induced cytidine deaminase (AID), whose
Foote and Eisen (80, 81) argued that the action during transcription is necessary for
residence time of an antigen complexed to a somatic hypermutation (SHM) (85), to actively
B-cell surface antibody would constrain the transcribed immunoglobulin variable regions
dissociation rate constant (koff) selectable in (86). These GanpTg mice generate exception-
vivo. The essence of their proposal was that ally high-affinity monoclonal antibodies
antibody-antigen complexes with lifetimes against nitrophenyl-hapten, HIV-1 (V3 pep-
much longer than the time necessary for tide)-epitope (87) and SARS-CoV-epitope (Kd
uptake would all be processed equally well, > 1011 M) (86). These mice develop Hodgkin-
hence, could not be distinguished. This idea of like lymphomas of non-T and non-B surface
an intrinsic affinity ceiling for the selection of phenotype with multiple mutations at immu-
antibodies generated in vivo was supported by noglobulin variable heavy chain region genes
the result of a study by Batista and Neuberger (88). Custom production of antibodies using
(82). To study how signaling and antigen these mice is offered by Trans Genic Inc.
presentation through B-cell receptors depend
on antigen/B-cell receptor affinity, lysozyme-
In Vitro Mutagenesis, Display,
specific B-cell transfectants were challenged
and Selection
with mutated lysozymes differing in their
binding kinetics. The results of their study In vitro mutagenesis remains one of the most
confirmed the predicted ceiling value for Ka in commonly used and widely accepted stra-
the order of 1010 M1 (Kd of 100 pM) proposed tegies for affinity maturating antibodies to the
by Foote and Eisen. This, of course, does not nanomolar (89), picomolar (90), or even fem-
imply that antibodies with affinities greater tomolar (91) range. In vitro antibody display
332 LO ET AL.

technologies, such as phage, yeast, and ribo- able to affinity mature an antibody to human
some display, have been widely utilized to -nerve growth factor, as full-length, glyco-
increase antibody affinity. There are many sylated IgGs, to low pM Kd.
options for diversifying V genes for affinity
maturation, including the random introduc- Afnity Maturation of Antibody by
tion of mutations, for example, by error-prone Mammalian display
PCR, CDR mutagenesis using spiked oligo- Problems with protein folding, posttranslation-
nucleotides and chain shuffling. These differ- al modification, and codon usage still limit the
ent strategies have been reviewed extensively number of improved antibodies that can be
(92, 93). Here we are going to describe some obtained by display of antibodies on the surface
alternative and new developments for in vitro of microorganisms (phage, bacteria, and yeast).
affinity maturation of antibodies. An ideal system would select and improve
antibodies in a mammalian cell environment
In Vitro Afnity Maturation by where they are naturally made. Ho et al. have
Somatic Hypermutation described a system to display single-chain anti-
The genetic diversity created by the immune CD22 Fv fragments on human embryonic
system is so large that, for nearly every foreign kidney 293T cells (101). In a proof-of-concept
antigen, an antibody can be generated to bind it. experiment, cells expressing a rare mutant
After combinatorial rearrangement of gene antibody with higher affinity were enriched
segments, the antibody variable region is 240-fold by a single-pass cell sorting from a
mutated at a rate orders of magnitude greater large excess of cells expressing wild-type
than the spontaneous mutation rate. This antibody with a slightly lower affinity. Fur-
occurs when activated B cells encounter anti- thermore, they successfully obtained a highly
gen and is termed immunoglobulin (Ig) SHM. enriched mutant with increased binding affin-
AID is essential for the initiation of SHM in B ity for CD22 after a single selection of a
cells by the deamination of cytidine residues combinatory library randomizing an intrinsic
directly in Ig genes (94). To achieve this, AID is antibody hotspot. Standard transfection or
targeted to V-region DNA sequences, termed electroporation methods, however, will typi-
hotspots (e.g., WRCH) that result in mutations cally introduce multiple copies of vectors into
and amino acid substitutions, which are fre- each cell. It is difficult to control the number of
quently in positions biased to modulate antigen vectors introduced into each cell and, more
binding (95). Expression of AID alone has been importantly, the location of the integration into
shown to be sufficient to reproduce the salient the host cell genome. Multiple copies of vectors
features of SHM in both B cells (96, 97) and and multiple integration sites in the host cell
other mammalian cells (98). However, these create a time-consuming process for screening
cell lines are difficult to transfect at efficiencies and characterization. It also becomes extreme-
suitable for use with diverse libraries. Never- ly difficult to compare precisely the affinity and
theless, the potential of using SHM in vitro has expression on the mammalian cell surface and
been apparent since Cumbers et al. (99) were isolate the desired cell clones efficiently. Zhou
able to evolve the endogenous IgM in Ramos et al. described a novel mammalian full-length
cells to recognize streptavidin with an apparent IgG display system successful in solving the
affinity of 11 nM after 19 rounds of fluores- critical issues of single specificity and relatively
cence-activated cell sorting; demonstrating equal expression through the recombinase Flp-
that hypermutating cell lines can be used for mediated single-copy integration. They dem-
iterative maturation of antibodies in vitro (99). onstrated the application of their system in
More recently, Bowers et al. (100) have devel- affinity maturation, identifying anti-OX40 li-
oped a novel approach where, by coupling in gand mutants of parental mAb40-L11 antibody
vitro SHM with mammalian display, they were with significantly improved neutralizing activ-
CHAPTER 19 Antibody Engineering 333

ity from a randomly mutagenized library (102). well implemented, very complex libraries can
More recently, two other mammalian cell- be screened relatively easily by panning or cell
displayed full-length antibody display systems sorting, respectively. However, the single-
that could be used for affinity maturation have pot phage display panning approach of large
been described (103, 104). libraries has well-documented drawbacks
with respect to selection bias toward variants
Afnity Maturation by Somatic Mutations that are better displayed or that allow faster
and Computational Design growth of the expressing host cell (112, 113).
Recent advances in computational technolo- The effects are magnified over the multiple
gies combined with dramatic advances in high- rounds of selection and amplification, so that
throughput technology have the potential to useful candidates may be lost, if they do not
make important contributions to the develop- also compete well in these therapeutically
ment of biological therapeutic antibodies irrelevant selection criteria. In addition, it is
(105). In the ideal case where high-resolution impractical to generate and screen by display
antibody structures or, preferably, antibody- methods many dozens of mutagenic libraries
antigen complex structures are available, simultaneously but individually, an approach
determination of contact residues is straight- that proved important in the present work.
forward, and this information can be applied to Finally, display methods generally have an
guide the maturation process (106). If exper- insufficient dynamic range to allow reliable
imentally determined structures are not avail- isolation of the tightest protein target binders
able, but the paratope has been reliably when starting with an already tightly binding
mapped, a three-dimensional model of the antibody. As an alternative to display methods,
variable domains can be constructed (107) and antibody variants can be tested in a clonal,
the residues affecting affinity can be projected well-based, high-throughput screening pro-
onto it (108), thereby facilitating the selection cess with quantitative determination of prop-
of candidate positions for maturation. For erties to ensure selection of candidates with
review of recent progress in the field of the best mix of desired characteristics.
computer-aided antibody development mainly Votsmeier et al. (91) describe an elegant and
focusing on antibody modeling, see Kuroda generic method to improve antibody affinities
et al. (109). Improvements are still needed in without phage or cell surface display. In their
all of the computational approaches; for approach, they used clonal, quantitative, well-
example, understanding loop flexibility is based high-throughput screening for selection
particularly important in antibody design be- of soluble Fab (fragment antigen binding)
cause large conformational changes can occur expression of the library variants and limited
upon antigen binding, and affinity maturation library diversification to CDRs only. In each
can modulate both the conformation and library, two neighboring residues were fully
dynamics of the CDR loops. diversified. The mutation pairs in each screen-
In terms of improvements of the prop- ing hit were deconvoluted to identify contri-
erties in antibodies, such as immunogenicity butions from each individual residue change.
and solubility, sequence-based methods have In total, 33 double-NNK libraries were gener-
shown promise in antibody engineering (110, ated, encompassing all 62 CDR positions of
111). adalimumab (114). Their multistep optimiza-
tion approach includes a first mutation
gathering round of library generation and
In Vitro Afnity Maturation
screening, followed by rounds in which
without Display
the gathered mutations are recombined.
The widespread use of phage and yeast display They applied their approach to the affinity
can be attributed largely to the fact that, when maturation of antitumor necrosis factor (TNF)
334 LO ET AL.

antibody adalimumab, which binds TNF with Several Next-Generation Antibody tech-
100 pM affinity, and achieved 500-fold nologies have been developed to overcome one
affinity improvement, resulting in femtomolar or more of the liabilities mentioned above. The
binding. With similar reasoning, adalimumab smaller size and less complex structure of
was previously subjected to affinity matura- antibody fragments and alternative scaffold
tion by using a single-chain variable fragment binders are cheaper to produce and may have
(scFv) yeast display approach including diver- better tumor penetration (118). Antibody drug
sity generation at every CDR residue posi- conjugates (ADCs), bispecific antibodies, and
tion and recombination of the beneficial the glycoengineering of the Fc glycan men-
mutations (115). In comparison with Rajpal et tioned above may result in antibodies with
al., the Votsmeier et al. process resulted in improved efficacy (119). However, all these
fewer mutations for a given affinity increase. technologies have one thing in common: they
Given the large flexibility in assay design of are still nascent and it remains to be shown
well-based screening, the approach described that the innovation translates into clinical and
by Votsmeier et al. has clear applications to commercial success.
other antibody properties, such as stability,
solubility, selectivity, species cross-reactivity,
Antibody Drug Conjugates
expression yield, receptor modulation, or
cellular responses. One approach to increase the efficacy of mAbs
in the treatment of cancer and virally infected
cells is the antibody drug conjugates (ADC)
THE NEXT-GENERATION ANTIBODIES TO concept (120, 121). Because small-molecule
MEET FUTURE CHALLENGES cancer drugs often have issues with dose-
limiting systemic toxicities, the concept of
Despite mAbs having outstanding commercial ADC is to use mAbs to target those compounds
success with 4 of the top 10 best-selling drugs in directly to the tumor. To this end, conjugated
2009 (116), they suffer from several agents may include small-molecule drugs,
shortcomings. First, the addressable target toxins, radionuclides, proteins, peptides, and
space appears to be limited, causing fierce other classes (122). Although two radionuclide
competition. In oncology, for example, nine conjugates, Zevalin and Bexxar (123), have
marketed products correspond to only six been marketed, neither of those molecules is a
different targets (vascular endothelial growth particular commercial success, and it appears
factor, EGFR, Her2, CD20, CD33, and CD52). that industry nowadays has regained interest
For autoimmune and inflammatory diseases, for conjugation of small-molecule drugs to
four TNF-neutralizing mAbs have been antibodies (124). The idea of using an antibody
marketed (infliximab, adalimumab, golimumab, chemically linked to a cytotoxic small mole-
and certolizumab PEGol) (117). The number of cule to target a tumor is more than 25 years old,
mAb launches validating new targets is limited but the therapeutic index appeared to be an
to only a few per year, demonstrating that new issue. Gemtuzumab ozogamicin (125), a con-
therapeutic target discovery is an elusive and jugate of calicheamicin to a humanized anti-
costly business. Another major drawback is that CD33 mAb, was released in 2000 but subse-
mAbs have relatively low clinical efficacy and quently withdrawn because of production and
are usually not effective as monotherapy in safety concerns (126). Only recently, another
cancer. As a consequence of their large molec- ADC was launched, and some 20 are in the
ular weight and complex structure, mAbs do clinical pipeline (124). Brentuximab vedotin, a
not penetrate tissues very well and need to be conjugate of an auristatin derivative to an anti-
administered parenterally. In addition, manu- CD30 antibody (127), was developed by Seattle
facturing of mAbs is complex and costly. Genetics in collaboration with Millenium, and
CHAPTER 19 Antibody Engineering 335

approved for the treatment of relapsed and these medicines come along with a steep price
refractory Hodgkins lymphoma. Another ad- tag. In 2011, Seattle Genetics announced that
vanced clinical candidate, T-DM1, which is a the treatment with brentuximab vedotin will
maytansinoid fusion with Roche Genentechs cost $13,500 per dose with seven to nine doses
Trastuzumab, is currently in Phase 3 for Her2+ per trial treatment (124).
metastatic breast cancer (128). T-DM1 exploits Immunocytokine (ICK) can be considered
the ADC technology developed by Immuno- another form of ADC with cytokine as a
gen, Seattle Genetics major competitor. Both payload (131). As a recombinant antibody-
types of payloads, Seattle Genetics auristatins cytokine fusion protein instead of a chemical
and Immunogens maytansinoids, are highly conjugate, ICK does not have the production
potent antimitotic agents that show intrinsic issues of a typical ADC. Also, its cytokine is not
activity in proliferating cells. packaged in the ICK as an inactive pro-drug,
To achieve an optimal ADC, several and hence it exerts its biological activity once
parameters need to be considered. In general, in circulation. Therefore, the concept of ICK
it is important to utilize a highly selective relies on the specific targeting by the antibody
antibody directed against a cell surface recep- moiety to bring the potent cytokine moiety to
tor that is overexpressed on tumor cells the tumor site to activate the tumor-infiltrating
relative to normal tissues to avoid unwanted immune cells, resulting in the induction of a
side effects. If the toxin is active inside the cell, long-term protective immune response. Simi-
such as the tubulin blockers or DNA-methyl- lar to ADC, the specificity of the antibody is of
ating agents, the ADC needs to be internalized utmost importance to minimize its side effects.
by the target cell after ligand binding and the For treatment of infectious diseases, the
toxin released to initiate the cytotoxic activity. cytokine can be chosen for its proinflammato-
The chemical linker connecting the small- ry/anti-inflammatory profile to suit the appli-
molecule toxin and the antibody plays a critical cation. Furthermore, the cytokine can be
role for every ADC concept, and cleavable and further engineered to decrease systemic tox-
noncleavable linkers have been described. icity and increase the therapeutic index, as
While noncleavable linkers release the toxin demonstrated by a low-toxicity IL-2 ICK (132).
by degradation of the antibody in the lysosomal
pathway, release of the payload from cleavable
Bispecic Antibodies
linkers is either driven by enzymatic activity or
by acidic pH in the hypoxic environment Combining mAbs with different mechanisms
inside a tumor (129). of action (MOAs) is another approach for
On the CMC side, ADC appears to be improving the efficacy of mAbs, and is cur-
particularly challenging (130). Chemical con- rently being explored in a number of clinical
jugation of drug-linker complexes to mAbs and preclinical programs. For example, differ-
needs to be carefully controlled to guarantee ent targets such as EGFR and insulin growth
product homogeneity. Depending on the factor receptor can be combined to explore
chemistry utilized, several conjugations can synergistic MOAs (133) or to suppress resis-
take place at multiple sites of a single mAb tance mechanisms as described for EGFR and
molecule, and the number of toxins added cMET (134).
needs to be optimized and controlled. In Monoclonal antibody combinations are
addition, conjugation of a drug-linker complex challenging from a development and marketing
may change the CMC properties of the ADC perspective, since each single antibody as well
relative to the parental antibody requiring as the combination will likely require regula-
more complex production processes and ana- tory testing and approval from authorities such
lytics. In summary, developing an ADC is as the FDA and European Medicines Evalua-
challenging, and, thus, it is no surprise that tion Agency, leading to delays and a significant
336 LO ET AL.

increase in development costs. In addition, the heterodimerization could also be achieved by


interdependency of efficacy and pharmacoki- electrostatic steering (145). Recently, Davis
netics needs to be carefully studied. Bispecific et al. (146) designed strand-exchange engi-
antibody (bsAb) technologies appear to be neered domains (SEED), a novel bsAb platform
an attractive alternative, because they offer based on the engineering of the CH3 domain.
a solution to combine synergistic MOAs in a SEED is based on a rational design strategy
single entity. In addition, bsAbs open new that combines structurally related and con-
MOAs as demonstrated by Micromets BiTE served sequences of human IgG and IgA CH3
(135), which physically links the effector cell to domains. By alternating sequences derived
the target cell through a bridging mechanism from both CH3 domains, two distinct SEED
that cannot be achieved by a combination of the protein chains (designated AG and GA) were
two separate antibodies. generated, creating asymmetric interfaces with
Although the bsAb concept provides a modified -sheet structures that preferentially
striking solution for the combination of differ- heterodimerize. Because CH3 domains drive
ent MOAs, production of these molecules the heterodimerization of immunoglobulin
turned out to be very challenging. Shortly heavy chains, AG and GA chains expressed in
after the invention of the mAb technology, the context of antibody heavy chains can be
Milstein and Cuello (136, 137) described the utilized to produce monovalent and monospe-
Quadroma technology for the production of cific or bivalent and bispecific antibody-like
bsAb which was essentially a hybrid of two molecules.
hybridomas. Although scientifically very ele- Since light chain pairing to the heavy chain
gant, it became apparent that production in is random during the assembly of a bispecific
industrial scale was not feasible because of the antibody, there is a need for a technology
complexity of the molecular species produced that can direct the specific pairing of the
(138). Other early approaches included chem- free light chain to its cognate heavy chain.
ical cross-linking (139, 140), tandem fusions of The Crossmab approach uses immunoglobulin
ScFv or V domains (141, 142), or their fusion to domain crossover (147) to get around this,
the C terminus of heavy chains. Up to now, while some technologies screen for a common
only one bsAb, the anti-EpCAM x anti-CD3 light chain for the two different Fabs (148), or
catumaxomab (143), has been approved in use single variable domains to avoid the use of
Europe for the treatment of malignant ascites. the light chain altogether (149). One approach
Catumaxomab is a rat/mouse chimeric IgG2a succeeded to provide a completely natural
that is very immunogenic. Other bsAbs in ad antibody with bispecific targeting by screening
vanced clinical development, such as the anti- for a single Fv that can bind two different
CD19 x anti-CD3 blinatumomab (Micromet), antigens. This two-in-one antibody uses
are based on tandem fusions of scFvs and an Fv variant of the antibody trastuzumab
therefore do not display mAb effector functions that interacts with both Her2 and vascular
or favorable pharmacokinetic properties such endothelial growth factor (150). On the other
as a long serum half-life in humans. hand, the dual variable domains-Ig approach
A heterodimeric human Fc fusion retaining covalently links the two VLs of different spe-
a long serum half-life and mAb-like effector cificities, which then pair up with the two
functions is the ideal vehicle for engineer- cognate VHs also similarly linked, thus creat-
ing therapeutic bsAbs. Genentechs knobs- ing the two variable domains of different
into-holes technology (144) offered a first specificities in tandem (151).
technical solution by introducing different In the development and design of the
complementary mutations in the two CH3 heterodimeric molecules, special care has to
domains that favor heterodimerization. In- be taken so that the resulting bispecific
stead of designs based on steric considerations, molecule bears the same biological properties
CHAPTER 19 Antibody Engineering 337

as the parental IgG molecule. This includes 8. Beenhouwer DO, Yoo EM, Lai CW, Rocha MA,
long serum half-life, ADCC and CDC, as well as Morrison SL. 2007. Human immunoglobulin G2
(IgG2) and IgG4, but not IgG1 or IgG3, protect
excellent biophysical properties such as high mice against Cryptococcus neoformans infection.
protein homogeneity, protein A binding, and Infect Immun 75:14241435.
thermal stability. In addition, it is important to 9. Tucker SC, Casadevall A. 2002. Replication of
evaluate the manufacturability of this new Cryptococcus neoformans in macrophages is ac-
molecule, including expression levels, ease of companied by phagosomal permeabilization and
accumulation of vesicles containing polysaccha-
purification, thermal stability, and long-term ride in the cytoplasm. Proc Natl Acad Sci USA
storage potential. Only a molecule that has 99:31653170.
passed all of these tests would be ready for 10. Zebedee SL, Koduri RK, Mukherjee J, Mukherjee
incorporation into the therapeutic pipeline. S, Lee S, Sauer DF, Scharff MD, Casadevall A.
1994. Mouse-human immunoglobulin G1 chi-
meric antibodies with activities against Crypto-
ACKNOWLEDGMENT coccus neoformans. Antimicrob Agents Chemother
38:15071514.
Conflicts of interest: We disclose no conflicts. 11. Schneider-Merck T, Lammerts van Bueren JJ,
Berger S, Rossen K, van Berkel PH, Derer S,
Beyer T, Lohse S, Bleeker WK, Peipp M, Parren
CITATION PW, van de Winkel JG, Valerius T, Dechant M.
2010. Human IgG2 antibodies against epidermal
Lo K-M, Leger O, Hock B. 2014. Antibody
growth factor receptor effectively trigger anti-
engineering. Microbiol Spectrum 2(1):AID- body-dependent cellular cytotoxicity but, in con-
0007-2012 trast to IgG1, only by cells of myeloid lineage.
J Immunol 184:512520.
12. Isaacs JD, Clark MR, Greenwood J, Waldmann
REFERENCES H. 1992. Therapy with monoclonal antibodies. An
1. Rodriguez ME, van der Pol WL, Sanders LA, in vivo model for the assessment of therapeutic
van de Winkel JG. 1999. Crucial role of potential. J Immunol 148:30623071.
FcgammaRIIa (CD32) in assessment of functional 13. Isaacs JD, Wing MG, Greenwood JD, Hazleman
anti-Streptococcus pneumoniae antibody activity BL, Hale G, Waldmann H. 1996. A therapeutic
in human sera. J Infect Dis 179:423433. human IgG4 monoclonal antibody that depletes
2. Woof JM, Kerr MA. 2004. IgA function target cells in humans. Clin ExpImmunol 106:427
variations on a theme. Immunology 113:175177. 433.
3. Dechant M, Beyer T, Schneider-Merck T, 14. Isaacs JD. 2001. From bench to bedside: discov-
Weisner W, Peipp M, van de Winkel JG, ering rules for antibody design, and improving
Valerius T. 2007. Effector mechanisms of recom- serotherapy with monoclonal antibodies. Rheu-
binant IgA antibodies against epidermal growth matology 40:724738.
factor receptor. J Immunol 179:29362943. 15. Wypych J, Li M, Guo A, Zhang Z, Martinez T,
4. Weisbart RH, Kacena A, Schuh A, Golde DW. Allen MJ, Fodor S, Kelner DN, Flynn GC, Liu
1988. GM-CSF induces human neutrophil IgA- YD, Bondarenko PV, Ricci MS, Dillon TM,
mediated phagocytosis by an IgA Fc receptor Balland A. 2008. Human IgG2 antibodies display
activation mechanism. Nature 332:647648. disulfide-mediated structural isoforms. J Biol
5. Lohse S, Derer S, Beyer T, Klausz K, Peipp M, Chem 283:1619416205.
Leusen JH, van de Winkel JG, Dechant M, 16. Dillon TM, Ricci MS, Vezina C, Flynn GC, Liu
Valerius T. 2011. Recombinant dimeric IgA YD, Rehder DS, Plant M, Henkle B, Li Y,
antibodies against the epidermal growth factor Deechongkit S, Varnum B, Wypych J, Balland
receptor mediate effective tumor cell killing. A, Bondarenko PV. 2008. Structural and func-
J Immunol 186:37703778. tional characterization of disulfide isoforms of the
6. Nimmerjahn F, Ravetch JV. 2010. Antibody- human IgG2 subclass. J Biol Chem 283:16206
mediated modulation of immune responses. 16215.
Immunol Rev 236:265275. 17. Lo KM, Zhang J, Sun Y, Morelli B, Lan Y,
7. Noya FJ, Baker CJ, Edwards MS. 1993. Neutro- Lauder S, Brunkhorst B, Webster G, Hallakou-
phil Fc receptor participation in phagocytosis of Bozec S, Doar L, Gillies SD. 2005. Engineering a
type III group B streptococci. Infect Immun pharmacologically superior form of leptin for the
61:14151420. treatment of obesity. Protein Eng Des Sel 18:110.
338 LO ET AL.

18. Tan LK, Shopes RJ, Oi VT, Morrison SL. 1990. antineuroblastoma IgG1 with optimized antibody-
Influence of the hinge region on complement dependent cellular cytotoxic activity. Nat Bio-
activation, C1q binding, and segmental flexibility technol 17:176180.
in chimeric human immunoglobulins. Proc Natl 29. Shinkawa T, Nakamura K, Yamane N, Shoji-
Acad Sci USA 87:162166. Hosaka E, Kanda Y, Sakurada M, Uchida K,
19. Dela Cruz JS, Huang TH, Penichet ML, Anazawa H, Satoh M, Yamasaki M, Hanai
Morrison SL. 2004. Antibody-cytokine fusion N, Shitara K. 2003. The absence of fucose
proteins: innovative weapons in the war against but not the presence of galactose or bisecting
cancer. Clin Exp Med 4:5764. N-acetylglucosamine of human IgG1 complex-
20. Klein JS, Bjorkman PJ. 2010. Few and far type oligosaccharides shows the critical role of
between. How HIV may be evading antibody enhancing antibody-dependent cellular cytoto-
avidity. PLoS Pathogens 6:16. xicity. J Biol Chem 278:34663473.
21. Bruhns P. 2012. Properties of mouse and human 30. Cartron G, Dacheux L, Salles G, Solal-Celigny P,
IgG receptors and their contribution to disease Bardos P, Colombat P, Watier H. 2002. Thera-
models. Blood 119:56405649. peutic activity of humanized anti-CD20 monoclo-
22. van der Neut Kolfschoten M, Schuurman J, nal antibody and polymorphism in IgG Fc
Losen M, Bleeker WK, Martnez-Martnez P, receptor FcRIIIa gene. Blood 99:754758.
Vermeulen E, den Bleker TH, Wiegman L, Vink 31. Treon SP, Hansen M, Branagan AR, Verselis S,
T, Aarden LA, De Baets MH, van de Winkel JG, Emmanouilides C, Kimby E, Frankel SR,
Aalberse RC, Parren PW. 2007. Anti-inflammatory Touroutoglou N, Turnbull B, Anderson KC,
activity of human IgG4 antibodies by dynamic Fab Maloney DG, Fox EA. 2005. Polymorphisms in
arm exchange. Science 317:15541557. FcRIIIA (CD16) receptor expression are associ-
23. Labrijn AF, Buijsse AO, van den Bremer ET, ated with clinical response to rituximab in
Verwilligen AY, Bleeker WK, Thorpe SJ, Waldenstrms macroglobulinemia. J Clin Oncol
Killestein J, Polman CH, Aalberse RC, 23:474481.
Schuurman J, van de Winkel JG, Parren PW. 32. Musolino A, Naldi N, Bortesi B, Pezzuolo D,
2009. Therapeutic IgG4 antibodies engage in Capelletti M, Missale G, Laccabue D, Zerbini A,
Fab-arm exchange with endogenous human Camisa R, Bisagni G, Neri TM, Ardizzoni A.
IgG4 in vivo. Nat Biotechnol 27:767771. 2008. Immunoglobulin G fragment C receptor
24. Shapiro RI, Plavina T, Schlain BR, Pepinsky RB, polymorphisms and clinical efficacy of trastuzumab-
Garber EA, Jarpe M, Hochman PS, Wehner NG, based therapy in patients with HER-2/neu-
Bard F, Motter R, Yednock TA, Taylor FR. 2011. positive metastatic breast cancer. J Clin Oncol
Development and validation of immunoassays 26:17891796.
to quantify the half-antibody exchange of an IgG4 33. Bibeau F, Lopez-Crapez E, Di Fiore F, Thezenas
antibody, natalizumab (Tysabri) with endogenous S, Ychou M, Blanchard F, Lamy A, Penault-
IgG4. J Pharm Biomed Anal 55:168175. Llorca F, Frbourg T, Michel P, Sabourin JC,
25. Rispens T, Meesters J, den Bleker TH, Boissire-Michot F. 2009. Impact of FcRIIa-
Ooijevaar-De Heer P, Schuurman J, Parren FcRIIIa polymorphisms and KRAS mutations on
PW, Labrijn A, Aalberse RC. 2013. Fc-Fc inter- the clinical outcome of patients with metastatic
actions of human IgG4 require dissociation of colorectal cancer treated with cetuximab plus
heavy chains and are formed predominantly by the irinotecan. J Clin Oncol 27:11221129.
intra-chain hinge isomer. Mol Immunol 53:3542. 34. Kumpel BM. 2007. Efficacy of RhD monoclo-
26. Li H, Sethuraman N, Stadheim TA, Zha D, Prinz nal antibodies in clinical trials as replacement
B, Ballew N, Bobrowicz P, Choi BK, Cook WJ, therapy for prophylactic anti-D immunoglob-
Cukan M, Houston-Cummings NR, Davidson R, ulin: more questions than answers.Vox Sang
Gong B, Hamilton SR, Hoopes JP, Jiang Y, Kim 93:99111.
N, Mansfield R, Nett JH, Rios S, Strawbridge 35. Peipp M, Lammerts van Bueren JJ, Schneider-
R, Wildt S, Gerngross TU. 2006. Optimization Merck T, Bleeker WW, Dechant M, Beyer T,
of humanized IgGs in glycoengineered Pichia Repp R, van Berkel PH, Vink T, van de Winkel
pastoris. Nat Biotechnol 24:210215. JG, Parren PW, Valerius T. 2008. Antibody
27. Lifely MR, Hale C, Boyce S, Keen MJ, Phillips J. fucosylation differentially impacts cytotoxicity
1995. Glycosylation and biological activity of mediated by NK and PMN effector cells. Blood
CAMPATH-1H expressed in different cell lines 112:23902399.
and grown under different culture conditions. 36. Mori K, Iida S, Yamane-Ohnuki N, Kanda Y,
Glycobiology 5:813822. Kuni-Kamochi R, Nakano R, Imai-Nishiya H,
28. Umaa P, Jean-Mairet J, Moudry R, Amstutz H, Okazaki A, Shinkawa T, Natsume A, Niwa R,
Bailey JE. 1999. Engineered glycoforms of an Shitara K, Satoh M. 2007. Non-fucosylated
CHAPTER 19 Antibody Engineering 339

therapeutic antibodies: the next generation of 48. DallAcqua WF, Kiener PA, Wu H. 2006.
therapeutic antibodies. Cytotechnology 55:109 Properties of human IgG1s engineered for en-
114. hanced binding to the neonatal Fc receptor
37. Shields RL, Namenuk AK, Hong K, Meng YG, (FcRn). J Biol Chem 281:2351423524.
Rae J, Briggs J, Xie D, Lai J, Stadlen A, Li B, Fox 49. Petkova SB, Akilesh S, Sproule TJ, Christianson
JA, Presta LG. 2001. High resolution mapping of GJ, Al Khabbaz H, Brown AC, Presta LG, Meng
the binding site on human IgG1 for FcRI, FcRII, YG, Roopenian DC. 2006. Enhanced half-life of
FcRIII, and FcRn and design of IgG1 variants genetically engineered human IgG1 antibodies in a
with improved binding to the FcR. J Biol Chem humanized FcRn mouse model: potential applica-
276:65916604. tion in humorally mediated autoimmune disease.
38. Nimmerjahn F, Ravetch JV. 2005. Divergent Int Immunol 18:17591769.
immunoglobulin G subclass activity through se- 50. Vidarsson G, Stemerding AM, Stapleton NM,
lective Fc receptor binding. Science 310:15101512. Spliethoff SE, Janssen H, Rebers FE, de Haas M,
39. Lazar GA, Dang W, Karki S, Vafa O, Peng JS, van de Winkel JG. 2006. FcRn: an IgG receptor
Hyun L, Chan C, Chung HS, Eivazi A, Yoder SC, on phagocytes with a novel role in phagocytosis.
Vielmetter J, Carmichael DF, Hayes RJ, Blood 108:35733579.
Dahiyat BI. 2006. Engineered antibody Fc vari- 51. Scallon BJ, Tam SH, McCarthy SG, Cai AN, Raju
ants with enhanced effector function. Proc Natl TS. 2007. Higher levels of sialylated Fc glycans
Acad Sci USA 103:40054010. in immunoglobulin G molecules can adversely
40. Armour KL, Clark MR, Hadley AG, Williamson impact functionality. Mol Immunol 44:15241534.
LM. 1999. Recombinant human IgG molecules 52. Anthony RM, Nimmerjahn F, Ashline DJ,
lacking Fc receptor I binding and monocyte Reinhold VN, Paulson JC, Ravetch JV. 2008.
triggering activities. Eur J Immunol 29:26132624. Recapitulation of IVIG anti-inflammatory activity
41. Mueller JP, Giannoni MA, Hartman SL, Elliott with a recombinant IgG Fc. Science 320:373376.
EA, Squinto SP, Mathis LA, Evans MJ. 1997. 53. Anthony RM, Wermeling F, Karlsson MC,
Humanized porcine VCAM-specific monoclonal Ravetch JV. 2008. Identification of a receptor
antibodies with chimeric IgG2/G4 constant re- required for the anti-inflammatory activity of
gions block human leukocyte binding to porcine IVIG. Proc Natl Acad Sci USA 105:1957119578.
endothelial cells. Mol Immunol 34:441452. 54. Kaneko Y, Nimmerjahn F, Ravetch JV. 2006.
42. An Z, Forrest G, Moore R, Cukan M, Haytko P, Anti-inflammatory activity of immunoglobulin
Huang L, Vitelli S, Zhao JZ, Lu P, Hua J, Gibson G resulting from Fc sialylation. Science 313:670
CR, Harvey BR, Montgomery D, Zaller D, Wang 673.
F, Strohl W. 2009. IgG2m4, an engineered anti- 55. Simmons LC, Reilly D, Klimowski L, Raju TS,
body isotype with reduced Fc function. MAbs Meng G, Sims P, Hong K, Shields RL, Damico
1:572579. LA, Rancatore P, Yansura DG. 2002. Expression
43. Labrijn AF, Aalberse RC, Schuurman J. 2008. of full-length immunoglobulins in Escherichia coli:
When binding is enough: nonactivating antibody rapid and efficient production of aglycosylated
formats. Curr Opin Immunol 20:479485. antibodies. J Immunol Methods 263:133147.
44. Tao MH, Morrison SL. 1989. Studies of aglyco- 56. Jung ST, Kang TH, Kelton W, Georgiou G. 2011.
sylated chimeric mouse-human IgG. Role of Bypassing glycosylation: engineering aglycosylated
carbohydrate in the structure and effector func- full-length IgG antibodies for human therapy. Curr
tions mediated by the human IgG constant region. Opin Biotechnol 22:858867.
J Immunol 143:25952601. 57. Sazinsky SL, Ott RG, Silver NW, Tidor B,
45. Nimmerjahn F, Ravetch JV. 2008. Fc recep- Ravetch JV, Wittrup KD. 2008. Aglycosylated
tors as regulators of immune responses. Nat Rev immunoglobulin G1 variants productively engage
Immunol 8:3447. activating Fc receptors. Proc Natl Acad Sci USA
46. Jefferis R. 2007. Antibody therapeutics: isotype 105:2016720172.
and glycoform selection. Expert Opin Biol Ther 58. Jung ST, Reddy ST, Kang TH, Borrok MJ,
7:14011413. Sandlie I, Tucker PW, Georgiou G. 2010.
47. Suzuki T, Ishii-Watabe A, Tada M, Kobayashi T, Aglycosylated IgG variants expressed in bacteria
Kanayasu-Toyoda T, Kawanishi T, Yamaguchi that selectively bind FcRI potentiate tumor cell
T. 2010. Importance of neonatal FcR in regulating killing by monocyte-dendritic cells. Proc Natl
the serum half-life of therapeutic proteins con- Acad Sci USA 107:604609.
taining the Fc domain of human IgG1: a compar- 59. Jones TP, Dear PH, Foote J, Neuberger MS,
ative study of the affinity of monoclonal antibodies Winter G. 1986. Replacing the complementarity-
and Fc-fusion proteins to human neonatal FcR. determining regions in a human antibody with
J Immunol 184:19681976. those from a mouse. Nature 321:522525.
340 LO ET AL.

60. Lger OJ, Yednock TA, Tanner L, Horner HC, 72. Christensen PA, Danielczyk A, Ravn P, Larsen
Hines DK, Keen S, Saldanha J, Jones ST, Fritz M, Stahn R, Karsten U, Goletz S. 2009. Modify-
LC, Bendig MM. 1997. Humanization of a mouse ing antibody specificity by chain shuffling of V/V
antibody against human alpha-4 integrin: a po- between antibodies with related specificities.
tential therapeutic for the treatment of multiple Scand J Immunol 69:110.
sclerosis. Hum Antibodies 8:316. 73. Rosok MJ, Yelton DE, Harris LJ, Bajorath J,
61. Sato K, Tsuchiya M, Saldanha J, Koishihara Y, Hellstrm KE, Hellstrm I, Cruz GA, Kristensson
Ohsugi Y, Kishimoto T, Bendig MM. 1994. K, Lin H, Huse WD, Glaser SM. 1996. A com-
Humanization of a mouse anti-human interleu- binatorial library strategy for the rapid human-
kin-6 receptor antibody comparing two methods ization of anticarcinoma BR96 Fab. J Biol Chem
for selecting human framework regions. Mol 271:2261122618.
Immunol 31:371381. 74. Rader C, Ritter G, Nathan S, Elia M, Gout I,
62. Leger O, Saldanha WJ. 2012. Humanization of Jungbluth AA, Choen LS, Welt S, Old LJ,
antibodies, p 123. In Wood CR (ed), Molecular Barbas CF 3rd. 2000. The rabbit antibody re-
Medicine and Medicinal Chemistry, vol 4. Anti- pertoire as a novel source for the generation of
body Drug Discovery. Imperial College Press, therapeutic human antibodies. J Biol Chem 275:
London, United Kingdom. 1366813676.
63. Padlan EA. 1991. A possible procedure for reduc- 75. Nishibori N, Horiuchi H, Furusawa S, Matsuda
ing the immunogenicity of antibody variable H. 2006. Humanization of chicken monoclonal
domains while preserving their ligand-binding antibody using phage-display system. Mol Immunol
properties. Mol Immunol 28:489498. 43:634642.
64. Padlan EA. 1994. Anatomy of the antibody 76. Radstake TR, Svenson M, Eijsbouts AM, van
molecule. Mol Immunol 31:169217. den Hoogen FH, Enevold C, van Riel PL,
65. Padlan EA, Abergel C, Tipper JP. 1995. Identi- Bendtzen K. 2009. Formation of antibodies
fication of specificity-determining residues in against infliximab and adalimumab strongly
antibodies. FASEB J 9:133139. correlates with functional drug levels and clinical
66. Tan P, Mitchell DA, Buss TN, Holmes MA, responses in rheumatoid arthritis. Ann Rheum
Anasetti C, Foote J. 2002. Superhumanized Dis 68:17391745.
antibodies: reduction of immunogenic potential 77. De Groot AS, Scott DW. 2007. Immunogeni-
by complementarity-determining region grafting city of protein therapeutics. Trends Immunol
with human germline sequences: application to an 28:482490.
anti-CD28. J Immunol 169:11191125. 78. Bernett MJ, Karki S, Moore GL, Leung IW,
67. Lazar GA, Desjarlais JR, Jacinto J, Karki S, Chen H, Pong E, Nguyen DH, Jacinto J,
Hammond PW. 2007. A molecular immuno- Zalevsky J, Muchhal US, Desjarlais JR, Lazar
logy approach to antibody humanization and GA. 2010. Engineering fully human monoclonal
functional optimization. Mol Immunol 44:1986 antibodies from murine variable regions. J Mol
1998. Biol 396:14741490.
68. DallAcqua WF, Damschroder MM, Zhang J, 79. Lanzavecchia A. 1985. Antigen-specific inter-
Woods RM, Widjaja L, Yu J, Wu H. 2005. action between T and B cells. Nature 314:537
Antibody humanization by framework shuffling. 539.
Methods 36:4360. 80. Foote J, Eisen HN. 1995. Kinetic and affinity
69. Fransson J, Teplyakov A, Raghunathan G, Chi limits on antibodies produced during immune
E, Cordier W, Dinh T, Feng Y, Giles-Komar J, responses. Proc Natl Acad Sci USA 92:10541056.
Gilliland G, Lollo B, Malia TJ, Nishioka W, 81. Foote J, Eisen HN. 2000. Breaking the affinity
Obmolova G, Zhao S, Zhao Y, Swanson RV, ceiling for antibodies and T cell receptors. Proc
Almagro JC. 2010. Human framework adaptation Natl Acad Sci USA 97:1067910681.
of a mouse anti-human IL-13 antibody. J Mol Biol 82. Batista FD, Neuberger MS. 1998. Affinity depen-
398:214231. dence of the B cell response to antigen: a
70. Jespers LS, Roberts A, Mahler SM, Winter G, threshold, a ceiling, and the importance of off-
Hoogenboom HR. 1994. Guiding the selection of rate. Immunity 8:751759.
human antibodies from phage display repertoires 83. Drake AW, Myszka DG, Klakamp SL. 2004.
to a single epitope of an antigen. Biotechnology Characterizing high-affinity antigen/antibody
(NY) 12:899903. complexes by kinetic- and equilibrium-based
71. Kang AS, Jones TM, Burton DR. 1991. Antibody methods. Anal Biochem 328:3543.
redesign by chain shuffling from random combi- 84. Rathanaswami P, Roalstad S, Roskos L, Su QJ,
natorial immunoglobulin libraries. Proc Natl Acad Lackie S, Babcook J. 2005. Demonstration of an
Sci USA 88:1112011123. in vivo generated sub-picomolar affinity fully
CHAPTER 19 Antibody Engineering 341

human monoclonal antibody to interleukin-8. 95. Goodman MF, Scharff MD, Romesberg FE.
Biochem Biophys Res Commun 334:10041013. 2007. AID-initiated purposeful mutations in im-
85. Liu M, Duke JL, Richter DJ, Vinuesa CG, munoglobulin genes. Adv Immunol 94:127155.
Goodnow CC, Kleinstein SH, Schatz DG. 2008. 96. Delker RK, Fugmann SD, Papavasiliou FN.
Two levels of protection for the B cell genome 2009. A coming-of-age story: activation-induced
during somatic hypermutation. Nature 451:841 cytidine deaminase turns 10. Nat Immunol 10:
845. 11471153.
86. Maeda K, Singh SK, Eda K, Kitabatake M, 97. Kajita M, Okazawa T, Ikeda M, Todo K, Magari
Pham P, Goodman MF, Sakaguchi N. 2010. M, Kanayama N, Ohmori H. 2010. Efficient
GANP-mediated recruitment of activation- affinity maturation of antibodies in an engineered
induced cytidine deaminase to cell nuclei and to chicken B cell line DT40-SW by increasing point
immunoglobulin variable region DNA. J Biol Chem mutation. J Biosci Bioeng 110:351358.
285:2394523953. 98. Martin A, Scharff MD. 2002. Somatic
87. Sakaguchi N, Kimura T, Matsushita S, Fujimura hypermutation of the AID transgene in B and
S, Shibata J, Araki M, Sakamoto T, Minoda C, non-B cells. Proc Natl Acad Sci USA 99:12304
Kuwahara K. 2005. Generation of high-affinity 12308.
antibody against T cell-dependent antigen in the 99. Cumbers SJ, Williams GT, Davies SL, Grenfell
Ganp gene-transgenic mouse. J Immunol 174: RL, Takeda S, Batista FD, Sale JE, Neuberger
44854494. MS. 2002. Generation and iterative affinity mat-
88. Fujimura S, Xing Y, Takeya M, Yamashita uration of antibodies in vitro using hypermutating
Y, Ohshima K, Kuwahara K, Sakaguchi N. B-cell lines. Nat Biotechnol 20:11291134.
2005. Increased expression of germinal center- 100. Bowers PM, Horlick RA, Neben TY, Toobian
associated nuclear protein RNA-primase is asso- RM, Tomlinson GL, Dalton JL, Jones HA, Chen
ciated with lymphomagenesis. Cancer Res 65: A, Altobell L 3rd, Zhang X, Macomber JL, Krapf
59255934. IP, Wu BF, McConnell A, Chau B, Holland T,
89. de Haard HJ, van Neer N, Reurs A, Hufton Berkebile AD, Neben SS, Boyle WJ, King DJ.
SE, Roovers RC, Henderikx P, de Brune AP, 2011. Coupling mammalian cell surface display
Arends JW, Hoogenboom HR. 1999. A large with somatic hypermutation for the discovery and
non-immunized human Fab fragment phage maturation of human antibodies. Proc Natl Acad
library that permits rapid isolation and kinetic Sci USA 108:2045520460.
analysis of high affinity antibodies. J Biol Chem 101. Ho M, Nagata S, Pastan I. 2006. Isolation of
274:1821818230. anti-CD22 Fv with high affinity by Fv display
90. Schier R, McCall A, Adams GP, Marshall KW, on human cells. Proc Natl Acad Sci USA
Merritt H, Yim M, Crawford RS, Weiner LM, 103:96379642.
Marks C, Marks JD. 1996. Isolation of picomolar 102. Zhou C, Jacobsen FW, Cai L, Chen Q, Shen WD.
affinity anti-c-erbB-2 single-chain Fv by molecu- 2010. Development of a novel mammalian cell
lar evolution of the complementarity determining surface antibody display platform. MAbs 2:508
regions in the center of the antibody binding site. J 518.
Mol Biol 263:551567. 103. Li CZ, Liang ZK, Chen ZR, Lou HB, Zhou Y,
91. Votsmeier C, Plittersdorf H, Hesse O, Scheidig Zhang ZH, Yu F, Liu S, Zhou Y, Wu S, Zheng W,
A, Strerath M, Gritzan U, Pellengahr K, Scholz P, Tan W, Jiang S, Zhou C. 2012. Identification of
Eicker A, Myszka D, Coco WM, Haupts U. 2012. HBsAg-specific antibodies from a mammalian cell
Femtomolar Fab binding affinities to a protein displayed full-length human antibody library of
target by alternative CDR residue co-optimization healthy immunized donor. Cell Mol Immunol
strategies without phage or cell surface display. 9:184190.
MAbs 4:341348. 104. Li F, Liu YH, Li YW, Ju Q, Chen L, Xie PL, Li YH,
92. Wark KL, Hudson PJ. 2006. Latest technologies Li GC. 2012. Human anti-EGFL7 recombinant
for the enhancement of antibody affinity. Adv full-length antibodies selected from a mammalian
Drug Deliv Rev 58:657670. cell-based antibody display library. Mol Cell
93. Lowe D, Wilkinson T, Vaughan TJ. 2012. Affinity Biochem 365:7784.
maturation approaches for antibody lead optimi- 105. Reddy ST, Georgiou G. 2011. Systems analysis
zation, p 85119. In Wood CR (ed), Molecular of adaptive immunity by utilization of high-
Medicine and Medicinal Chemistry, vol 4. Anti- throughput technologies. Curr Opin Biotechnol
body Drug Discovery. Imperial College Press, 22:584589.
London, United Kingdom. 106. Clark LA, Boriack-Sjodin PA, Eldredge J, Fitch
94. Maul RW, Gearhart PJ. 2010. AID and somatic C, Friedman B, Hanf KJ, Jarpe M, Liparoto SF,
hypermutation. Adv Immunol 105:159191. Li Y, Lugovskoy A, Miller S, Rushe M, Sherman
342 LO ET AL.

W, Simon K, Van Vlijmen H. 2006. Affinity of theraputic antibodies. Nat Rev Immunol 10:
enhancement of an in vivo matured therapeutic 345352.
antibody using structure-based computational 120. Teicher BVA, Chati RVJ. 2011. Antibody conju-
design. Protein Sci 15:949960. gate therapeutics: challenges and potential. Clin
107. Fontayne A, De Maeyer B, De Maeyer M, Cancer Res 17:63896397.
Yamashita M, Matsushita T, Deckmyn H. 121. Mayes S, Brown N, Illidge TM. 2011. New
2007. Paratope and epitope mapping of the antibody drug treatments for lymphoma. Expert
antithrombotic antibody 6B4 in complex with Opin Biol Ther 11:623640.
platelet glycoprotein Ib. J Biol Chem 282:23517 122. Visiongain. 2012. Next generation antibody thera-
23524. pies: pipeline and market 20112012. Visiongain
108. Fontayne A, Vanhoorelbeke K, Pareyn I, Van Ltd., London, United Kingdom.
Rompaey I, Meiring M, Lamprecht S, Roodt J, 123. Stasi R. 2008. Gemtuzuman Ozogamicin: An anti-
Desmet J, Deckmyn H. 2006. Rational human- CD33 immunoconjugate for the treatment of acute
ization of the powerful antithrombotic anti-GPIb myeloid leukemia. Expert Opin Biol Ther 8:527
antibody: 6B4. Thromb Haemost 96:671684. 540.
109. Kuroda D, Shirai H, Jacobson MP, Nakamura H. 124. Gualberto A. 2012. Brentuximab Vedotin (SGN-
2012. Computer-aided antibody design. Protein 35), an antibody-drug conjugate for the treatment
Eng Des Sel 25:507522. of CD30-positive malignancies. Expert Opin
110. Abhinandan KR, Martin AC. 2007. Analyzing the Investig Drugs 21:205216.
degree of humanness of antibody sequences. 125. Burris HA 3rd, Tibbitts J, Holden SN, Sliwkowski
J Mol Biol 369:852862. MX, Lewis Phillips GD. 2011. Trastuzumab
111. David MP, Concepcion GP, Padlan EA. 2010. emtansine (T-DM1): a novel agent for target-
Using simple artificial intelligence methods for ing HER2+ breast cancer. Clin Breast Cancer
predicting amyloidogenesis in antibodies. BMC 11:275282.
Bioinformatics 11:79. 126. Alley SC, Okeley NM, Senter PD. 2010. Anti-
112. Clackson T, Wells JA. 1994. In vitro selection body-drug conjugates: targeted drug delivery for
from protein and peptide libraries. Trends Bio- cancer. Curr Opin Chem Biol 14:529537.
technol 12:173184. 127. Hughes B. 2010. Antibody-drug conjugates for
113. Derda R, Tang SK, Li SC, Ng S, Matochko W, cancer: poised to deliver? Nat Rev Drug Discov
Jafari MR. 2011. Diversity of phage-displayed 9:665667.
libraries of peptides during panning and amplifi- 128. Lwenberg B, Beck J, Graux C, van Putten W,
cation. Molecules 16:17761803. Schouten HC, Verdonck LF, Ferrant A, Sonneveld
114. Salfeld JG, Allen DJ, Hoogenboom HR, P, Jongen-Lavrencic M, von Lilienfeld-Toal M,
Kaymakcalan Z, Labkovsky B, Mankovich JA, Biemond BJ, Vellenga E, Breems D, de Muijnck
McGuinness BT, Roberts AJ, Sakorafas P, H, Schaafsma R, Verhoef G, Dhner H, Gratwohl
Schoenhaut D, Vaughan TJ, White M, Wilton A, Pabst T, Ossenkoppele GJ, Maertens J;
AJ. 2003. Human antibodies that bind human Dutch-Belgian Hemato-Oncology Cooperative
TNFa. US Patent 6090382. Group (HOVON); German Austrian AML Study
115. Rajpal A, Beyaz N, Haber L, Cappuccilli G, Group (AMLSG); Swiss Group for Clinical
Yee H, Bhatt RR, Takeuchi T, Lerner RA, Crea Cancer Research Collaborative Group (SAKK).
R. 2005. A general method for greatly improv- 2010. Gemtuzumab ozogamicin as postremission
ing the affinity of antibodies by using combina- treatment in AML at 60 years of age or more:
torial libraries. Proc Natl Acad Sci USA 102:8466 results of a multicenter phase 3 study. Blood 115:
8471. 25862591.
116. Via MC. 2009. Monoclonal Antibodies: Pipeline 129. Ducry L, Stump B. 2010. Antibody-drug con-
Analysis and Competitive Assessment. Cambridge jugates: linking cytotoxic payloads to monoclonal
Healthtech Institute, Needham, MA. antibodies. Bioconjugate Chem 21:513.
117. Licastro F, Chiappelli M, Ianni M, Porcellini E. 130. Lin K, Tibbitts J. 2012. Pharmacokinetic consi-
2009. Tumor necrosis factor-alpha antagonists: derations for antibody drug conjugates. Pharm Res
differential clinical effects by different biotechno- 29:23542366.
logical molecules. Int J Immunopathol Pharmacol 131. Gillies SD, Lan Y, Wesolowski JS, Qian X,
22:567572. Reisfeld RA, Holden S, Super M, Lo K-M. 1998.
118. Gebauer M, Skerra A. 2009. Engineered thera- Antibody-IL-12 fusion proteins are effective in
peutic scaffolds as next generation antibody SCID mouse models of prostate and colon carci-
therapeutics. Curr Opin Chem Biol 13:245255. noma metastases. J Immunol 160:61956203.
119. Beck A, Wurch T, Bailly C, Corvaia N. 2010. 132. Gillies SD, Lan Y, Hettmann T, Brunkhorst B,
Strategies and challenges for the next generation Sun Y, Mueller SO, Lo K-M. 2011. A low-toxicity
CHAPTER 19 Antibody Engineering 343

IL-2 based immunocytokine retains anti-tumor 144. Ridgway JB, Presta LG, Carter P. 1996. Knobs-
activity despite its high degree of IL-2 receptor into-holes engineering of antibody CH3 domains
selectivity. Clin Cancer Res 17:36733685. for heavy chain heterodimerization. Protein Eng
133. Lu D, Zhang H, Ludwig D, Persaud A, Jimenez 9:617621.
X, Burtrum D, Balderes P, Liu M, Bohlen P, 145. Gunasekaran K, Pentony M, Shen M, Garrett L,
Witte L, Zhu Z. 2004. Simultaneous blockade of Forte C, Woodward A, Ng SB, Born T, Retter M,
both the epidermal growth factor receptor and Manchulenko K, Sweet H, Foltz IN, Wittekind
the insulin-like growth factor receptor signaling M, Yan W. 2010. Enhancing antibody Fc hetero-
pathways in cancer cells with a fully human recom- dimer formation through electrostatic steering
binant bispecific antibody. J Biol Chem 279:28562865. effects: applications to bispecific molecules and
134. Stommel JM, Kimmelman AC, Ying H, monovalent IgG. J Biol Chem 285:1963719646.
Nabioullin R, Ponugoti AH, Wiedemeyer R, 146. Davis JH, Aperlo C, Li Y, Kurosawa E, Lan Y, Lo
Stegh AH, Bradner JE, Ligon KL, Brennan C, K-M, Huston JS. 2010. SEEDbodies: fusion
Chin L, DePinho RA. 2007. Coactivation of proteins based on strand-exchange engineered
receptor tyrosine kinases affects the response of domain (SEED) CH3 heterodimers in an Fc
tumor cells to targeted therapies. Science 318:287 analogue platform for asymmetric binders or
290. immunofusions and bispecific antibodies. Protein
135. Baeuerle PA, Reinhardt C. 2009. Bispecific T-cell Eng Des Sel 23:195202.
engaging antibodies for cancer therapy. Cancer 147. Schaefer W, Regula JT, Bhner M, Schanzer J,
Res 69:49414944. Croasdale R, Drr H, Gassner C, Georges G,
136. Milstein C, Cuello AC. 1983. Hybrid hybridomas Kettenberger H, Imhof-Jung S, Schwaiger M,
and their use in immunohistochemistry. Nature Stubenrauch KG, Sustmann C, Thomas M,
305:537540. Scheuer W, Klein C. 2011. Immunoglobulin
137. Milstein C, Cuello AC. 1984. Hybrid hybridomas domain crossover as a generic approach for the
and the production of bi-specific monoclonal production of bispecific IgG antibodies. Proc Natl
antibodies. Immunology Today 5:299304. Acad Sci USA 108:1118711192.
138. Carter P. 2001. Bispecific human IgG by design. 148. Merchant AM, Zhu Z, Yuan JQ, Goddard A,
J Immunol Methods 248:715. Adams CW, Presta LG, Carter P. 1998. An
139. Brennan M, Davison PF, Paulus H. 1985. Prep- efficient route to human bispecific IgG. Nat Bio-
aration of bispecific antibodies by chemical re- technol 16:677681.
combination of monoclonal immunoglobulin G1 149. Shen J, Vil MD, Jimenez X, Zhang H, Iacolina
fragments. Science 229:8183. M, Mangalampalli V, Balderes P, Ludwig DL,
140. Glennie MJ, McBride HM, Worth AT, Zhu Z. 2007. Single variable domain antibody as
Stevenson GT. 1987. Preparation and perfor- a versatile building block for the construction
mance of bispecific F(ab gamma)2 antibody of IgG-like bispecific antibodies. J Immunol
containing thioether-linked Fab gamma frag- Methods 318:6574.
ments. J Immunol 139:23672375. 150. Bostrom J, Yu SF, Kan D, Appleton BA, Lee
141. Atwell JL, Pearce LA, Lah M, Gruen LC, Kortt CV, Billeci K, Man W, Peale F, Ross S,
AA, Hudson PJ. 1996. Design and expression of a Wiesmann C, Fuh G. 2009. Variants of the
stable bispecific scFv dimer with affinity for both antibody herceptin that interact with HER2
glycophorin and N9 neuraminidase. Mol Immunol and VEGF at the antigen binding site. Science
33:13011312. 323:16101614.
142. Huston JS, George AJ. 2001. Engineered anti- 151. Wu C, Ying H, Grinnell C, Bryant S, Miller R,
bodies take center stage. Hum Antibodies 10:127 Clabbers A, Bose S, McCarthy D, Zhu RR,
142. Santora L, Davis-Taber R, Kunes Y, Fung E,
143. Seimetz D, Lindhofer H, Bokemeyer C. 2010. Schwartz A, Sakorafas P, Gu J, Tarcsa E,
Development and approval of the trifunctional Murtaza A, Ghayur T. 2007. Simultaneous
antibody catumaxomab (anti-EpCAM x anti-CD3) targeting of multiple disease mediators by a dual-
as a targeted cancer immunotherapy. Cancer Treat variable-domain immunoglobulin. Nat Biotechnol
Rev 36:458467. 25:12901297.
High-Throughput DNA Sequencing
Analysis of Antibody Repertoires

SCOTT D. BOYD1 and SHILPA A. JOSHI1


20
INTRODUCTION

New high-throughput DNA sequencing (HTS) technologies developed in


the past decade have rapidly increased the scale of data collection for all
aspects of human genetics (1, 2). The complex somatic gene rearrangements
of immunoglobulin (Ig) and T-cell antigen receptors (TCRs) in the adaptive
immune system are particularly appropriate targets for investigation using
these new technologies. The antigen specificity of adaptive human immune
responses and the storage of specific immunological memory depend on the
sequences of the Ig and TCR gene rearrangements expressed by B cells and
T cells. Until recently, the difficulty and cost of obtaining sequence data
limited the kinds of immunological research questions that could be studied.
Pioneering work examining dozens to hundreds of Ig rearrangements with
Sanger sequencing has revealed some overall features of the repertoires of
these receptors, while physical selection and sorting of B-cell populations
of interest has led to the identification of antibodies specific for a variety
of infectious agents and vaccine components. However, given that a single
human body contains an estimated 1011 B cells representing, at a minimum,
millions of distinct clonal populations, experiments using Sanger sequenc-
ing were underpowered to evaluate the full scale of antibody repertoires.

1
Department of Pathology, Stanford University, Stanford, CA 94305.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0017-2014

345
346 BOYD AND JOSHI

This chapter first reviews genetic features of elsewhere (3). The region of the antibody
Ig loci and the HTS technologies that have heavy or light chain structure encoded by
been applied to human repertoire studies, then the fusion of V, (D), and J segments forms a
discusses experimental design, data analysis loop that is the most diverse region of the
choices in these experiments, and insights antibody and is termed the complementar-
gained in immunological and infectious dis- ity-determining region 3 (CDR3). CDR3 is
ease studies using these approaches. often the most important portion of the
antibody for binding to antigen, but two
other loops (CDR1 and CDR2) encoded by
ANTIBODY GENE REARRANGEMENTS the variable gene segment can also contrib-
ute significantly to antigen specificity.
Antibodies in humans are protein com- The genetic complexity of the antibody
plexes whose basic unit is a disulfide-linked repertoire in an individuals B cells prior
pair of heavy chain proteins, each with an to exposure to antigen derives from the
associated light chain (Fig. 1). The N- combinatorial assembly of V, (D), and J
terminal regions of heavy and light chains segments, the diversification at the segment
are highly variable in their sequences, and junctions encoding the CDR3 loops of the
are the antigen-binding portions of the antibody heavy or light chain, and the
antibody. The C-terminal regions of the pairwise combination of heavy and light
proteins are termed the constant regions. chains. Earlier rough estimates of the
Light chains are of two types, kappa (IgK) potential number of different antibodies
or lambda (IgL), while heavy chains (IgH) that can be generated by these mechanisms
are of five major isotypes (IgM, IgD, IgG, were greater than 1011 (4). Such figures are
IgA, and IgE), with four subtypes of IgG and undoubtedly underestimates, because the
two subtypes of IgA. Developing B cells theoretical number of unique heavy or
assemble the genes encoding the antigen- light chains is limited only by the maximal
binding regions of their immunoglobulin length of the nontemplated junctional base
heavy chains from germ line arrays of sequences considered in the calculation.
variable (V), diversity (D), and joining (J) Antigen-stimulated B cells activate an addi-
gene segments, by first joining a D and J tional program of antibody sequence diver-
segment together, and then selecting a V sification called somatic hypermutation that
segment to join to the newly generated D-J generates new point mutations throughout
product. During joining, the ends of the the rearranged V(D)J sequence, creating
gene segments are subject to exonuclease clonal offspring whose antibodies may pos-
digestion, and nontemplated randomized sess increased binding affinity for the
bases (N bases) are added at the segment antigen. Rarer processes such as receptor
junctions. In the developing B cell, an initial editing contribute further mechanisms for
attempt to rearrange the immunoglobulin antibody repertoire generation by enabling
heavy chain locus on one copy of chromo- secondary rearrangements and replacement
some 14 can potentially give rise to an out- of V gene segments. At the level of human
of-frame product; if this happens, then the populations, additional genetic diversity of
B cell attempts to rearrange the other copy antibody repertoires results from allelic
of the locus. After a productive IgH is variants for immunoglobulin gene segments
expressed, an analogous rearrangement of and structural variants within the immuno-
V and J segments at the kappa or lambda globulin loci that delete or increase the copy
immunoglobulin light chain locus takes number of particular gene segments.
place. The mechanistic details and regula- Current understanding of the human germ
tion of these events have been reviewed line DNA sequence for the immunoglobulin
CHAPTER 20 HTS Analysis of Antibody Repertoires 347

FIGURE 1 Antibody structure and genetic encoding. The germ line (unrearranged) genomic DNA
conguration of the immunoglobulin heavy chain locus is depicted at the top of the gure, showing the
tandem arrays of V, D, and J gene segments (not to scale). A germ line kappa or lambda light chain locus
is depicted on the left-hand side, with unrearranged V and J segments. Stepwise rearrangement of the
germ line DNA results in the joining of a heavy chain D and J gene segment, followed by joining of a V
segment to the D-J product, to generate the DNA encoding the heavy chain variable region. In the
process of rearrangement, the ends of the gene segments are subject to variable amounts of
exonuclease digestion, and randomized nontemplated bases are added at the segment ends, to
produce additional sequence diversity at the VDJ junctional region that encodes the complementarity-
determining region 3 (CDR3) loop, which is often the region of the antibody heavy chain that has
the greatest impact on antigen specicity. A similar process of V and J gene rearrangement with
diversication of the VJ junction occurs in the light chain locus, to produce the rearranged light chain
gene. The constant regions of the heavy and light chains (domains CH1, CH2, and CH3 for the heavy
chain, and CL for the light chain) are encoded by downstream exons that are joined to the rearranged V
(D)J gene by mRNA splicing. Disulde bridges joining protein chains in the full antibody structure are
shown with black line segments. doi:10.1128/microbiolspec.AID-0017-2014.f1

heavy chain locus, and the number of V, D, and sequencing efforts have often been less helpful
J segments it contains, remains heavily indebt- for evaluating immunoglobulin loci, in part,
ed to the initial sequence generated by Matsuda because of the widespread use of oligoclonal
et al. in 1998 (5). Recent human genome- Epstein-Barr virus (EBV)-transformed B-cell
348 BOYD AND JOSHI

lines with rearranged Ig genes as the source regions. The process of rearranging the
of DNA for sequencing, resulting in the loss genomic DNA at heavy and light chain loci
of information about gene segments deleted to generate in-frame heavy and light chains
during rearrangement, as well as the relatively results in a compact V(D)J gene that is
shallow depth of sequencing performed in approximately 400 bases in length.
population-level studies (6). In contrast, Somatic hypermutation of human anti-
HTS has very recently been used to study the bodies occurs primarily in the specialized
germ line locus in particularly tractable hu- microenvironments of secondary lymphoid
man samples, such as cells from a haploid tissues, where B cells have access to anti-
hydatidiform mole, and to survey the haplo- gen, specialized dendritic and stromal cells,
types in this locus in different human popula- and T cells that stimulate B cells via soluble
tion groups (7). The curated sequences in mediators and cell-cell contact (13). Human
the IMGT database (www.imgt.org) for the plasmablasts observed after acute antigenic
human IGH locus at 14q32 indicate that most stimulation such as influenza vaccination
humans, depending on their haplotypes, have usually show mutation levels in the range of
123 to 129 IGHV gene segments, of which 43 to 5 to 15% in the IGHV segment, while memory
46 are able to form functional rearrangements; B cells show somewhat lower mutation levels
27 IGHD gene segments (23 functional); and (14). In unusual circumstances such as chronic
9 IGHJ segments (6 functional) (8, 9). The exposure to viral antigens in the perturbed
IGK locus at 2p11 encodes 76 IGKV in most immune systems of HIV-infected individuals,
individuals (31 to 36 functional), and 5 IGKJ much higher mutation levels (over 30%) can
gene segments (5 functional), while the IGL be observed in some antibody lineages (15).
locus at 22q11 contains 73 to 74 IGLV segments Mutational events are targeted relatively pre-
(29 to 33 functional) and 7 to 11 IGLJ segments cisely in a region extending from upstream
(4 to 5 functional) (8, 9). Allelic and copy of, or within, the leader sequence through the V
number variation in Ig loci in different human (D)J rearrangement, and taper off in the intron
populations were identified in earlier litera- separating the J segments from the constant
ture, but the pace of identification of new regions (16, 17). Notably, leader sequences in
variants has now accelerated, making it clear some loci may be less mutated than the rest of
that the currently curated variants represent the Ig gene rearrangement, as best documented
only a small sampling of the total variation that in mouse kappa light chains (17).
is likely to be present across all human groups
(7, 10, 11, 12).
The genetic structure of V gene segments HIGH-THROUGHPUT DNA SEQUENCING
includes a small upstream exon encoding INSTRUMENTS
most of a leader peptide, followed by a short
intron and a second exon encoding the rest Soon after the completion of the first human
of the leader peptide and the V segment genome draft sequence in 2001, a technolog-
itself. In the unrearranged human germ line ical race between several different companies
loci, the V, D, and J regions are separated and academic laboratories led to the develop-
from each other by kilobases of sequence, ment of a handful of competing platforms for
and the constant regions, encoded by single determining DNA sequences in a more highly
exons (for kappa or lambda) or several parallel, miniaturized, and efficient manner
exons (for the heavy chain isotypes) are than was possible with Sanger sequencing.
located kilobases downstream of the J Common features among these methods were
segments, with the exception of the lambda the use of miniaturized microwell plates
locus, where a few J segments are inter- or flow cells that could capture DNA mole-
spersed among the most upstream constant cules at particular spatial positions, methods
CHAPTER 20 HTS Analysis of Antibody Repertoires 349

of generating locally amplified template from polymer tracts of a particular nucleotide are
single DNA molecules for sequencing, and a sequenced with decreased accuracy, because
method of detection of nucleotide sequence it is more difficult to distinguish between the
that could occur in parallel for thousands to levels of signal produced by, for example,
millions of distinct templates at the same time. 14 incorporation events compared with 15
The platforms that have been most widely incorporation events in a sequencing step.
used for Ig sequencing in the published Error rates are discussed in greater detail
literature have been those from Roche/454, below. Current versions of these instruments
Illumina, and Ion Torrent. A brief overview of give approximately one million sequences
the features of these instruments is given with read lengths of over 500 bases in the
below. Other promising technologies, includ- case of Roche/454, and 10 million sequences of
ing true single-molecule sequencing approaches, 100 bases for the Ion Torrent PGM. The
have been less widely used so far. majority of the published literature on HTS
In brief, the Roche/454 and Ion Torrent of immunoglobulin genes has used the Roche/
methods spatially separate and amplify 454 methodology, but Roche has indicated
single-template molecules by limiting dilu- that it is no longer developing the 454 platform
tion of the template in aqueous solution and will not support it in the future.
followed by production of an aqueous-in-oil In contrast, the Illumina platform, which
emulsion. The template is diluted so that has now become the dominant HTS method
less than one template molecule is present for most applications, performs template
on average per aqueous droplet. The aque- separation and amplification of diluted
ous phase contains the enzymes, primers, template DNA on the surface of a flow cell
nucleotides, and buffers for PCR, as well as (20). The flow cell is functionalized with
capture beads to which the amplified tem- oligonucleotides that capture the template
plate becomes attached. After PCR, the and prime bridging PCR amplifications that
emulsion is broken and the beads are use linker sequences introduced at the ends
positioned in microfabricated wells of a of the template during library construction.
plate. Both the Roche/454 and Ion Torrent The bridging PCR yields focal clusters of
platforms use a sequencing-by-synthesis amplified template molecules that are cova-
approach in which reaction mixtures con- lently attached to the flow cell. The amplified
taining only one of the deoxyribonucleotide clusters are then sequenced with cycles of
triphosphates (dATP, dCTP, dGTP, or single-base extension sequencing-by-synthesis.
dTTP) are sequentially used in cycles of In this strategy, each nucleotide is labeled with
extension of a DNA strand complementary a fluorophore that identifies the incorporated
to the template. In 454 sequencing, incor- nucleotide and blocks further extension of the
poration of one or more nucleotides during template once the nucleotide is added to the
an extension step is detected by a coupled growing DNA strand. A mixture of all four
enzymatic reaction in the sequencing plate nucleotide triphosphates is used in each cycle
well, in which pyrophosphate liberated from of synthesis. Nucleotide incorporation into
the nucleotide triphosphate added to the each template cluster during each sequencing
growing DNA strand drives the generation cycle can be detected by imaging the flow cell
of photons of light by luciferase enzyme (18). and detecting what fluorophore is present.
The Ion Torrent platform detects nucleotide Once the flow cell is imaged, the fluorescent
incorporation during DNA synthesis via labels are cleaved off and the next cycle of
miniaturized ion sensors in the sequencing extension and imaging begins. Until recently,
plate that detect the release of protons occur- a disadvantage of the Illumina instruments
ring upon nucleotide addition (19). In both the was their relatively short read lengths (initially
Roche/454 and Ion Torrent protocols, homo- 35 bases), but continual improvements in the
350 BOYD AND JOSHI

methodology have led to current instruments or any tissue containing B cells. In order to
and kits that give total read lengths of sequence identify sequences that are derived from B cells
that are comparable to the those of the 454 that have undergone somatic mutation, the
platform. The Illumina MiSeq instrument can sequences in such libraries can be evaluated for
sequence approximately 15 million templates the presence of mutated nucleotide positions
per run, yielding up to 300 bases of sequence in V, D, or J gene segments (21). However, a
from each end of the template molecules, while number of specialized B-cell subsets in
the HiSeq 2500 model can sequence 2 billion humans, such as memory B cells, plasmablasts,
templates, reading 125 bases from each end of transitional B cells, and suppressor B cells,
the template. among others, have been defined on the basis
of cell surface receptors, intracellular protein
EXPERIMENTAL STRATEGIES FOR expression, or other phenotypic features, and
SEQUENCING Ig REPERTOIRES there can be additional experimental value
added by using flow cytometry or other
Sequencing Ig V(D)J rearrangements in methods to sort out particular B-cell or plasma
human samples is conceptually straightfor- cell subsets of interest prior to generating
ward. As a result of the close juxtaposition libraries (14, 22, 23, 24). It is clear, however,
of V, (D), and J gene segments following that the cell subset definitions and the func-
genomic rearrangement, short V(D)J PCR tions ascribed to them are subject to progres-
amplicons (<500 bp, excluding the lengths sive refinement and revision over time in the
of sequencing instrument linkers and immunological literature, meaning that
barcodes) can be amplified efficiently from comparisons of Ig repertoire data based on
genomic DNA or complementary DNA from cell subsets should be made with caution
samples containing B cells. The kilobases of unless identical protocols and definitions are
sequence that separate the gene segments in used (22). As a practical matter, sorting of small
the unrearranged genomic DNA prevent cell subsets can also make it more difficult to
significant amplification from non-B-cell isolate nucleic acids for sequencing with the
templates in the sample. The incorporation use of standard methods and can require
of oligomer nucleotide barcodes in the additional rounds of PCR to generate enough
primers or linkers used in library prepara- amplified material for accurate quantitation
tion can be used for sample multiplexing prior to sequencing. Another consideration in
in a single sequencing run with any of the human studies is that sometimes samples that
HTS instruments, permitting samples to be are precious because of the rarity of the clinical
sequenced together but allowing the sequenc- phenotype, pathogen exposure, or other im-
ing data to be assigned back to the samples by munological stimulus may only be available in
sorting based on the unique sample barcodes. the form of frozen nonviable cells or leukocyte-
Beyond these similarities, a number of impor- or PBMC-derived RNA or DNA, owing to the
tant experimental design choices influence the sample collection and storage protocols or
kinds of interpretation that can be reliably resource limitations in clinical studies. In such
made from sequencing libraries generated by cases, valuable data can still be obtained from
using different approaches, as detailed below. total Ig repertoires without cell surface pheno-
type information, and antibody isotype expres-
sion and mutation status can be interpreted.
Cell Populations
High-quality DNA sequencing libraries of
Targeted PCR versus 5 RACE
V(D)J rearrangements can be produced from
templates isolated from total leukocytes, The 5 rapid amplification of cDNA ends (5
peripheral blood mononuclear cells (PBMCs), RACE) for library preparation requires only
CHAPTER 20 HTS Analysis of Antibody Repertoires 351

a single primer to hybridize to a known mutated antibody lineages such as broadly


region of the target mRNA. cDNA synthesis neutralizing anti-HIV antibodies (31, 32).
proceeds until the 5 end of the target
mRNA is reached, and then one of several
Choice of Template
approaches can be used to add a known,
unrelated primer sequence to the 3 end of Ig sequencing libraries can be generated
the cDNA strand, permitting subsequent from genomic DNA template, typically with
PCR amplification (25). This method has V-and J-segment primers, or from cDNA
begun to be applied to generate Ig HTS template using V-leader primers, internal V
libraries, and may be less subject to primer primers, J primers, or constant region primers
bias and multiplexed PCR artifacts that can (Fig. 2). In addition to the considerations
result from using primers within the V, J, or related to potential effects of somatic mutation
constant regions, while also being better at on primer binding, as outlined above, there are
amplifying heavily mutated Ig sequences several other reasons to select either genomic
where primer binding sites may be altered DNA template, cDNA template, or both,
(26, 27). Limitations of 5 RACE methods depending on the goals of a particular exper-
are the requirement for RNA as the starting iment. One advantage of genomic DNA tem-
material, precluding analysis of genomic plate is that it permits analysis of both
DNA rearrangements, and more variable productively rearranged Ig sequences that
performance with RNA of suboptimal qual- yield a protein product, as well as unpro-
ity or limiting quantity. The alternative ductively rearranged loci where a stop codon
approach of using multiplexed primer or reading frame incompatibility between the
mixtures targeting the V, D, J, or constant V segment and the downstream gene segments
regions, and performing PCR from either has been introduced during V(D)J rearrange-
cDNA or genomic DNA template, has been ment. While not giving rise to functional
the predominant method used in the liter- protein, the unproductive rearrangements
ature to date (Fig. 2). This method is subject provide a record of the features of the Ig
to a few potential problems, including PCR locus rearrangement, such as gene-segment
amplification bias, as a result of different rearrangement frequencies, segment chew-
primer annealing efficiencies to particular back by exonucleases, and nontemplated
gene segments; the need to select primers base addition levels prior to selection of the
that can be multiplexed without producing B cell based on expressed Ig protein. This can
off-target primer dimers or other artifacts; and be useful in evaluating immunological pheno-
potential loss of highly mutated sequences types related to receptor gene rearrangements
from sequencing libraries as a result of or B-cell selection, such as in immunodefi-
the somatic mutation preventing primer ciency disorders (K. Roskin, submitted for
annealing. Selection of primers for human Ig publication). In addition, the fact that there is
rearrangements has focused on the framework only one productively rearranged IGH, IGK,
(FR) regions FR1, FR2, and FR3, which are or IGL locus per cell can be used to evaluate
relatively less mutated than the CDR se- the sizes of B-cell clones in the cell populations
quences in B cells that survive to be observed studied, because multiple replicate libraries
in human samples, likely because mutated for sequencing can be generated from inde-
framework regions that destabilize the anti- pendent aliquots of genomic DNA template,
body structure are not compatible with the and V(D)J rearrangements from different
persistence of the B cell in the body (28, 29, members of the same clone of B cells can
30). Primers targeting the leader sequences later be identified in data analysis (21, 33). In
have also been used in a number of pub- contrast, the many copies of Ig mRNA pro-
lications, particularly those studying highly duced within a single B cell, and the differing
352 BOYD AND JOSHI

FIGURE 2 IgH library production from genomic DNA (gDNA) or complementary DNA (cDNA). The top
diagram shows the rearranged gDNA encoding an antibody heavy chain. Primer sets designed to
hybridize in the framework 1, 2, or 3 (FR1, FR2, FR3) regions (labeled with circled numbers 1, 2, and 3),
together with a primer complementary to the J gene segments (labeled 4), can be used for PCR to
amplify the VDJ gene rearrangement. Multiple primers are shown for the framework primer sets,
indicating the different primers required for amplication of V segments belonging to different families.
The leader peptide exon is separated from the V segment by a short intron in the genomic DNA. The
lower diagram shows cDNA generated from spliced mRNA encoding a heavy chain. Primers hybridizing
to the constant region (labeled 6) can be used as the initial gene-specic primer for 5 RACE protocols
(see main text), or else can be used in PCR with primers in the leader sequences (labeled 5), or framework
primers, to amplify the VDJ gene rearrangements. The constant region isotype associated with the VDJ
gene rearrangement can be identied in such libraries. doi:10.1128/microbiolspec.AID-0017-2014.f2

levels of mRNA expression for Ig genes in is lost during purification owing to less than
different subsets of B cells, such as relatively 100% yield, there will not be a linear reduction
low expression in nave cells and high expres- in the number of B-cell clones represented in
sion in plasmablasts, mean that it is not the sample, as is the case with genomic DNA.
possible to reliably distinguish between the Finally, cDNA contains more amplifiable Ig
presence of cells with high Ig mRNA expres- templates per nanogram of template ampli-
sion versus the presence of a clone containing fied, compared with genomic DNA where the
many cells with lower Ig mRNA expression, if rearranged Ig loci are a tiny fraction of the
only a single RNA sample is available for total DNA quantity. Because there is a limit to
analysis. However, using RNA as the starting the amount of DNA template that can be added
material for repertoire sequencing experi- to a PCR reaction, more complex libraries of Ig
ments has other important advantages, chiefly rearrangements therefore can be generated
that it enables identification of the heavy chain with fewer PCR reactions by the use of the
isotype associated with a particular V(D)J cDNA template compared with the genomic
rearrangement. Another potential advantage DNA template.
of RNA template is that it should preserve a
more complete representation of the B-cell
Multiplexing and Chimeric Sequences
clones present in the cell sample, because each
B cell can contribute multiple mRNA copies of Another kind of artifact that can arise in
its Ig rearrangements, so that, if some template library preparation is the chimeric sequence,
CHAPTER 20 HTS Analysis of Antibody Repertoires 353

usually generated when a DNA strand is exception of polynucleotide tract regions


incompletely extended in one PCR cycle, and in data gathered with 454/Roche or Ion
can then hybridize to an unrelated template Torrent instruments. A recent comparison
and be extended further in a subsequent cycle. of error rates of benchtop HTS instruments
If PCR amplifications are performed with sequencing Escherichia coli genomic DNA
several different heavy chain isotype primers found that the insertion and deletion (indel)
in the same reaction, such chimeras may lead rate was 0.38% per base for the 454 GS
to errors of assignment of particular VDJ Junior, 1.5% per base for the Ion Torrent
rearrangements to particular isotypes. Other- PGM, and <0.001% per base for the Illumina
wise, they can yield apparent regions of highly MiSeq (36). In this comparison, the substi-
increased somatic mutation in the V(D)J tution error rate of the MiSeq instrument
rearrangement, and other artifactual results. was the lowest measured, at 0.1 per 100
Bioinformatic filtering to remove sequences bases. The quality of the sequence data with
with such unusual features is one way of all of these instruments decreases toward
attempting to minimize such effects, but other the distal ends of the reads. When applied
approaches such as performing PCR with to Ig templates, reported error rates for the
templates at limiting dilution in aqueous-in- combination of PCR and sequencing error,
oil emulsions or microtiter plates have been excluding indels, have been reported in the
proposed and used as alternatives, although literature and seen in our own experiments,
these dilution methods raise other challenges to be approximately 0.1 to 0.3% per base for
of sample throughput (34, 35). the 454 platform and 0.1 to 0.2% for
Illumina sequencing (21, 29, 36, 37, 38).
One approach to detecting potential PCR or
Replicate Library Preparation
sequencing errors, and distinguishing these
As noted above, the preparation of multiple from true somatic mutation positions or
libraries from a sample by using indepen- allelic variants of gene segments, is to obtain
dent template aliquots (either by using a manyfold more sequencing reads than the
genomic DNA template, or by separating number of template molecules in the original
cells into separate aliquots prior to isolating sample. Errors occurring early in PCR cannot
RNA template) permits reliable detection of be corrected with this approach, but, together
expanded clonal B-cell populations and with the evaluation of sequences derived from
distinction between an expanded clone known templates (such as sequences cloned
of cells versus a single cell expressing into plasmids), this approach can be used to
high levels of Ig mRNA (21, 33). As with correct many sequencing-derived errors, and
any experimental methodology, conducting give an upper bound of remaining PCR and
experiments with replicate sampling and sequencing errors in the data. An alternative
library generation helps to distinguish be- method of detecting errors from amplifica-
tween real signals compared with statistical tion or sequencing has been adapted for Ig
noise in the dataset and also gives a basis sequencing from earlier approaches of adding
for assessing the robustness of any other highly diverse sequence tags to nucleic acid
secondary results derived from the data. templates prior to amplification of libraries
from the templates (39, 40, 41, 42). Incorpora-
tion of a randomized sequence tag in the primer
Error Correction Strategies
used for reverse transcription of Ig mRNA
The combined frequency of PCR errors and enables later comparison of the data from reads
sequencing errors for Ig amplicon libraries sharing the same sequence of randomized tag,
sequenced with current HTS platforms are and helps the inference of which variant bases
usually less than 0.5% per base, with the are likely to be errors in such sequences (38).
354 BOYD AND JOSHI

Paired Heavy and Light Chain scripts used for analysis, should enable appro-
Sequencing priate verification of reported results. One
overall limitation of many published articles to
A long-awaited experimental breakthrough
date has been the relatively low number of
in Ig repertoire studies has been the effi-
samples analyzed and the rare to absent
cient high-throughput sequencing of native
consideration of statistical correction for mul-
pairs of heavy and light chain (H+L) se-
tiple hypothesis testing in data sets that have
quences from large numbers of individual B
many different features that can be compared
cells. In 2013, the proof of concept of a
between samples.
method of generating paired H+L sequence
libraries from tens of thousands of individual B
cells was described, making use of high- Sequence Barcode Analysis, Filtering,
density microtiter plates with 125-pl well Primer Trimming, and Quality Score Use
volumes to separate the cells and enable
As with other amplicon-sequencing exper-
capture of mRNA from each cell on an oligo-
iments, data analysis of Ig libraries typically
dT functionalized polymer bead, followed by
includes early steps of identifying the
reverse transcription and joining PCR to
sequence barcodes that identify the sample
covalently link the heavy and light chain
of origin of a sequence. Many groups have
sequences (43). While the data sets from
designed barcodes that are relatively resis-
these initial experiments contained thousands
tant to sequencing error, in that they are
of natively paired sequences, it is likely that
different from each other at two or more
the protocols will improve their throughput
positions (i.e., are separated by a Hamming
with the use of emulsion strategies or poten-
distance of two or more). If gene-specific
tially even the use of microfluidic devices (44).
primers are used in the amplification strat-
egy and a primer is not a perfect match to
DATA ANALYSIS APPROACHES the gene segment it amplified, trimming of
primers is necessary to avoid spurious
A variety of data analysis methods have introduction or loss of point mutations.
been applied to Ig sequence data sets, with Quality scores for each nucleotide position
many laboratories developing their own are a feature calculated by all of the HTS
pipelines of publicly available software, cus- platforms and agree fairly well with actual
tomized scripts and programs, and databases error rates in experiments performed with
to manage the data. Owing to the numerous known template sequences (36). Quality
experimental strategies for library preparation scores can be used to exclude sequences of
being explored in the literature, and the excessively poor quality from further anal-
idiosyncrasies of each groups data analysis ysis; as an alternative approach, such reads
approaches, direct comparison of reported can be carried forward in the analysis and
results is challenging. This is not an insur- removed from consideration later if they do
mountable problem, so long as the experimen- not align well enough to gene segments of
tal protocols are described in sufficient detail, the locus of interest.
and the raw sequence data are made available
to other investigators who wish to evaluate the
Alignment and Parsing Programs
results by using their own analysis pipeline. It
is not realistic to expect that all research In the place of traditional mapping of reads
laboratories will converge on a single exper- to a reference scaffold, as is performed for
imental and data analysis approach, but full genome or exome sequence analysis, the
transparency about library preparation meth- next step in Ig sequence analysis is deter-
ods and software, including sharing of the mining which regions of the rearranged Ig
CHAPTER 20 HTS Analysis of Antibody Repertoires 355

align with germ line V, D, and J gene helpful to collapse identical sequences into a
segments and constant region sequences in single representative, to minimize PCR ampli-
the locus that was amplified, parsing the fication biases or stochastic PCR jackpots that
nontemplated regions at the segment junc- cause some sequences to appear in increased
tions and determining the positions of somatic copy numbers in the sequencing data. Overall
mutation in the sequence. A number of features of Ig repertoires, or the repertoires
programs have been devised for this purpose, derived from particular B-cell subsets, can
including IgBlast, V-QUEST, and the hidden be readily determined from the parsed se-
Markov model-based programs iHMMune- quence output of any of the alignment utili-
align and SoDA2 (45, 46, 47, 48). In response ties described above. Features that have been
to the increasing demand for higher-through- reported in many studies include gene seg-
put analysis of large sequence data sets, the ment usage frequencies; length, composition,
IMGT website (www.imgt.org) has intro- and amino acid sequences of CDR3 regions;
duced a new next-generation sequencing mutation levels and distributions; and heavy
data set portal for sequence alignment and chain isotype associated with particular V(D)J
analysis, HighV-QUEST (49). Many of these rearrangements, among other metrics.
published analysis programs claim superiority Recent articles have provided more de-
for their approach to sequence analysis, but, in our tailed insights into baseline human Ig
laboratory, evaluation of the alignments gen- repertoires than were previously known
erated by IgBlast, V-QUEST, and iHMMune- from lower-throughput data, including the
align are identical or very similar for most individual specific usage frequencies of V, D,
sequences, and differ most in junctional and J gene segments, and the presence of copy
parsing of sequences where the true answer number variants and allelic variants within
may be unknowable. For example, IGH se- particular haplotypes (11, 29, 50). Glanville et
quences in which the D segment is very short al. observed a strong influence of the germ line
owing to exonuclease digestion during rear- genome on V, D, and J gene segment usage
rangement, so that the true germ line D (probably combining the effects of receptor
segment identity cannot be determined rearrangement frequencies, and selection act-
would be such a case. An important consider- ing on the expressed protein) in repertoires
ation for any of these utilities is the choice of sequenced from two pairs of identical twins
germ line V, D, and J gene segment repertoires (51). HTS has also enabled better measure-
used for alignment, because current curated ment of the frequencies of uncommon features
databases are not complete, so depending on in Ig repertoires, such as rearrangements
the population group studied, rarer and unre- using two D segments in the heavy chain and
ported alleles can appear to carry somatic indels generated as a by-product of somatic
mutations as a result of misalignment to other hypermutation (52, 53, 54). The distinct
gene segments (11, 12). features of the repertoires of different B-cell
subsets, such as class-switched or IgM-
expressing memory cells, compared with
HTS Ig DATA ANALYSIS: APPLICATIONS nave B cells, at the level of gene segment
IN INFECTIOUS DISEASE RESEARCH usage and junctional features, have been
reported by several groups (21, 24, 30, 55).
A wide variety of secondary analyses can be Analysis of somatic mutation patterns in Ig
performed on Ig repertoire data once the rearrangements, and detection of evidence of
sequences are aligned and parsed, depending selection, has also been enhanced in part by
on the experimental question being asked. the availability of larger data sets for evalua-
Often, particularly if replicate libraries have tion, and has prompted the development of
been generated from a sample, it can be new tools and interfaces to facilitate analysis
356 BOYD AND JOSHI

of larger sequence sets (56, 57). The influx of insights into many other infectious diseases
much larger datasets of Ig sequences that and chronic or recurrent infections.
often contain many representatives of clonally Quantitation or normalization of the
expanded B-cell lineages has also stimulated contribution of clonally expanded B-cell
new approaches for inferring the relationships populations to an observed repertoire can
of descent and genetic inheritance between be performed from multiple-replicate li-
individual cells within the clone (58). brary data, using a modified form of the
Determining whether sequences derived Gini-Simpson index adapted for HTS data;
from the same B-cell clone are present in we have termed this a clonality score
different samples of a longitudinal time or coincidence index, in recent articles.
course, or different tissue sites, sample types, This measure can distinguish between in-
or B-cell subsets, is often of interest and can be dividuals responding to acute Dengue virus
approached in several different ways. For infection from those who are convalescent
initial analyses, we often use a clone definition or uninfected (21, 33). The presence of
for comparing two heavy chain sequences expanded B-cell clones that persist in the
requiring that the V and J segments be the circulation of individuals, as measured with a
same, or of the same subgroup, and that the similar clonality score from samples collected
CDR3 sequence be the same length and match a year apart, is significantly associated with
at 80% of the nucleotides. Depending on the EBV infection status in healthy subjects (21).
experimental question being asked, different We have also observed that this measure of
thresholds for clonal similarity may be more clonality is closely correlated with the increase
appropriate. For example, in searching for in numbers of plasmablasts observed in the
members of a clonal lineage that matures from blood 7 days after vaccination with inactivated
the germ line sequence state to a highly trivalent influenza vaccine, and that the
mutated state (as in the case of some broadly clonality score is correlated with serocon-
neutralizing antibodies against HIV), a more version following vaccination. Other recent
permissive definition of CDR3 similarity could studies of vaccination for influenza or pneu-
be used, and then the putative clone members mococcus have highlighted sequences that
could be tested for other inherited features appear at higher copy number in sequencing
such as somatic hypermutation positions. libraries following vaccination, which may
Ensuring that the conclusions drawn in a correspond to expanded clones stimulated by
study are not overly sensitive to minor differ- the vaccine, or cells producing higher levels of
ences in clonal definitions is a conservative Ig mRNA, or both (37, 38, 65).
and prudent approach. A number of groups Estimation of the size of the B-cell
have made striking progress in recent years by repertoire has been attempted in a number
using HTS to track the clonal evolution and of prior studies, but is subject to high degrees
mutational and selection histories of B-cell of uncertainty and underbias when extrapo-
lineages making antibodies specific for partic- lated from peripheral blood samples that
ular pathogens or vaccine components. Such typically contain millions of B cells,
analyses have been performed most extensive- representing less than 0.1% of the approxi-
ly in studies of HIV, identifying putative germ mately 100 billion B cells in an individuals
line ancestral sequences of highly mutated body (29). Parametric approaches that make
antibody lineages that acquire virus neutrali- assumptions about the distribution of clone
zation breadth only after many months to years sizes in the repertoire may be particularly
of chronic infection, and identifying shared unreliable, but even nonparametric estimates
sequence features of antibodies that bind to such as the lower bound of the Chao 2
particular epitopes of HIV (31, 32, 58, 59, 60, metric are likely to underestimate the min-
61, 62, 63, 64). Similar approaches may offer imal size of the repertoire (Y. Liu, personal
CHAPTER 20 HTS Analysis of Antibody Repertoires 357

communication, and reference 66). In our populations of biological or medical interest


view, currently published estimates of nave in any immunological context. Initial stud-
B-cell and T-cell receptor diversity are ies in infectious disease topics have provid-
underestimates of the true values; it will ed new knowledge about the numbers and
await larger data sets collected with multiple diversity of B-cell clones stimulated by
independent samples from each human sub- infection or vaccination by a variety of
ject to arrive at better estimates in the future. agents, and have been especially useful in
In light of the enormous size of the documenting the pathways of mutation and
potential antibody repertoire, it has been selection followed by antibodies responding
an open question whether different humans to HIV. Clear and explicit documentation of
raise similar antibodies in response to the the experimental methods used and data
same infectious or vaccination challenge. analysis approaches applied to Ig HTS data
Previous studies using Sanger sequencing sets, along with sharing of primer se-
identified some circumstances where highly quences, scripts for analysis, and other
similar convergent antibodies could be important experimental features along
detected in different people, such as immu- with raw and processed data, will help to
nization with Haemophilus influenzae type ensure the reproducibility and broader
b polysaccharide vaccine or pneumococcal scientific value of studies in this area. As
vaccine (67, 68). HTS of Ig repertoires has with any other research area generating and
greatly increased the ability to detect such analyzing large and complex molecular
convergent sequences, and pathogen-specific datasets, Ig sequence analysis will benefit
antibody rearrangements have now been from a greater emphasis on the use of
detected in patients infected with Dengue sample sets with larger numbers of individ-
virus and HIV (32, 33, 63) as well as in subjects ual patients or subjects, collection of longi-
vaccinated with trivalent inactivated influenza tudinal repeat sampling from individuals
(69). It appears that the stimulation of con- when possible, appropriate statistical anal-
vergent antibodies that are specific for partic- ysis to correct for multiple hypothesis testing,
ular pathogens is a widespread phenomenon. and validation of results with training and test
Once more data are gathered for a variety of sets of data. A particularly promising area is
different pathogens and antigens, it may the coupling of HTS studies of immune
become possible to read an individuals history response genetics (Ig and TCR sequencing,
of pathogen exposure from the Ig sequences in as well as analysis of other host genetic
their memory B-cell repertoire. features) with equally powerful genetic anal-
ysis of the populations of pathogens seeking to
infect the host and evade the immune system.
CONCLUSIONS AND The coming years should offer a ringside seat
FUTURE DIRECTIONS to researchers intent on observing these
fascinating battles.
The application of HTS methods to the
study of B-cell responses and immunoglob-
ACKNOWLEDGMENT
ulin structure-function relationships is now
well started, and most of the cost limitations Conflicts of interest: We disclose no conflicts.
and technological shortcomings of the ear-
lier generations of HTS instruments have
CITATION
been overcome. Well-designed experimen-
tal protocols and data analysis approaches Boyd SD, Joshi SA. 2014. High-throughput
promise to provide unprecedentedly accu- DNA sequencing analysis of antibody reper-
rate, extensive, and exact tracking of B-cell toires. Microbiol Spectrum 2(5):AID-0017-2014
358 BOYD AND JOSHI

REFERENCES 13. Klein U, Dalla-Favera R. 2008. Germinal centres:


role in B-cell physiology and malignancy. Nat Rev
1. Gonzaga-Jauregui C, Lupski JR, Gibbs RA. Immunol 8:2233.
2012. Human genome sequencing in health and 14. Wrammert J, Smith K, Miller J, Langley WA,
disease. Annu RevMed 63:3561. Kokko K, Larsen C, Zheng NY, Mays I, Garman
2. Boyd SD. 2013. Diagnostic applications of L, Helms C, James J, Air GM, Capra JD, Ahmed
high-throughput DNA sequencing. Annu Rev R, Wilson PC. 2008. Rapid cloning of high-
Pathol 8:381410. affinity human monoclonal antibodies against
3. Jung D, Giallourakis C, Mostoslavsky R, Alt influenza virus. Nature 453:667671.
FW. 2006. Mechanism and control of V(D)J 15. Mascola JR, Haynes BF. 2013. HIV-1 neutral-
recombination at the immunoglobulin heavy izing antibodies: understanding natures
chain locus. Annu Rev Immunol 24:541570. pathways. Immunol Rev 254:225244.
4. Janeway CAJ, Travers P, Waport M, Shlomchik 16. Lebecque SG, Gearhart PJ. 1990. Boundaries
MJ. 2001. Immunobiology: The Immune System of somatic mutation in rearranged immuno-
in Health and Disease, 5th ed. Garland Science, globulin genes: 5 boundary is near the promo-
New York. ter, and 3 boundary is approximately 1 kb from
5. Matsuda F, Ishii K, Bourvagnet P, Kuma K, V(D)J gene. J Exp Med 172:17171727.
Hayashida H, Miyata T, Honjo T. 1998. The 17. Rada C, Gonzalez-Fernandez A, Jarvis JM,
complete nucleotide sequence of the human Milstein C. 1994. The 5 boundary of somatic
immunoglobulin heavy chain variable region hypermutation in a V kappa gene is in the
locus. J Exp Med 188:21512162. leader intron. Eur J Immunol 24:14531457.
6. Abecasis GR, Altshuler D, Auton A, Brooks 18. Margulies M, Egholm M, Altman WE, Attiya
LD, Durbin RM, Gibbs RA, Hurles ME, S, Bader JS, Bemben LA, Berka J, Braverman
McVean GA. 2010. A map of human genome MS, Chen YJ, Chen Z, Dewell SB, Du L,
variation from population-scale sequencing. Fierro JM, Gomes XV, Godwin BC, He W,
Nature 467:10611073. Helgesen S, Ho CH, Irzyk GP, Jando SC,
7. Watson CT, Steinberg KM, Huddleston J, Alenquer ML, Jarvie TP, Jirage KB, Kim JB,
Warren RL, Malig M, Schein J, Willsey AJ, Joy Knight JR, Lanza JR, Leamon JH, Lefkowitz
JB, Scott JK, Graves TA, Wilson RK, Holt RA, SM, Lei M, Li J, Lohman KL, Lu H,
Eichler EE, Breden F. 2013. Complete haplotype Makhijani VB, McDade KE, McKenna MP,
sequence of the human immunoglobulin heavy- Myers EW, Nickerson E, Nobile JR, Plant R,
chain variable, diversity, and joining genes and Puc BP, Ronan MT, Roth GT, Sarkis GJ,
characterization of allelic and copy-number var- Simons JF, Simpson JW, Srinivasan M,
iation. Am J Hum Genet 92:530546. Tartaro KR, Tomasz A, Vogt KA, Volkmer
8. Lefranc MP. 2011. IMGT, the International GA, Wang SH, Wang Y, Weiner MP, Yu P,
ImMunoGeneTics Information System. Cold Begley RF, Rothberg JM. 2005. Genome
Spring Harb Protoc 2011:595603. sequencing in microfabricated high-density
9. Lefranc MP, Lefranc G. 2001. The Immu- picolitre reactors. Nature 437:376380.
noglobulin FactsBook. Academic Press, New 19. Rothberg JM, Hinz W, Rearick TM, Schultz
York, NY. J, Mileski W, Davey M, Leamon JH, Johnson
10. Wang Y, Jackson KJ, Gaeta B, Pomat W, Siba K, Milgrew MJ, Edwards M, Hoon J, Simons
P, Sewell WA, Collins AM. 2011. Genomic JF, Marran D, Myers JW, Davidson JF,
screening by 454 pyrosequencing identifies a new Branting A, Nobile JR, Puc BP, Light D, Clark
human IGHV gene and sixteen other new IGHV TA, Huber M, Branciforte JT, Stoner IB,
allelic variants. Immunogenetics 63:259265. Cawley SE, Lyons M, Fu Y, Homer N, Sedova
11. Boyd SD, Gaeta BA, Jackson KJ, Fire AZ, M, Miao X, Reed B, Sabina J, Feierstein E,
Marshall EL, Merker JD, Maniar JM, Zhang Schorn M, Alanjary M, Dimalanta E, Dressman
LN, Sahaf B, Jones CD, Simen BB, Hanczaruk D, Kasinskas R, Sokolsky T, Fidanza JA,
B, Nguyen KD, Nadeau KC, Egholm M, Namsaraev E, McKernan KJ, Williams A, Roth
Miklos DB, Zehnder JL, Collins AM. 2010. GT, Bustillo J. 2011. An integrated semiconductor
Individual variation in the germline Ig gene device enabling non-optical genome sequencing.
repertoire inferred from variable region gene Nature 475:348352.
rearrangements. J Immunol 184:69866992. 20. Bentley DR, Balasubramanian S, Swerdlow
12. Wang Y, Jackson KJ, Sewell WA, Collins AM. HP, Smith GP, Milton J, Brown CG, Hall KP,
2008. Many human immunoglobulin heavy-chain Evers DJ, Barnes CL, Bignell HR, Boutell
IGHV gene polymorphisms have been reported in JM, Bryant J, Carter RJ, Keira Cheetham R,
error. Immunol Cell Biol 86:111115. Cox AJ, Ellis DJ, Flatbush MR, Gormley NA,
CHAPTER 20 HTS Analysis of Antibody Repertoires 359

Humphray SJ, Irving LJ, Karbelashvili MS, tion on human B cell repertoires. J Immunol
Kirk SM, Li H, Liu X, Maisinger KS, Murray 192:603611.
LJ, Obradovic B, Ost T, Parkinson ML, Pratt 22. Jackson SM, Wilson PC, James JA, Capra
MR, Rasolonjatovo IM, Reed MT, Rigatti R, JD. 2008. Human B cell subsets. Adv Immunol
Rodighiero C, Ross MT, Sabot A, Sankar SV, 98:151224.
Scally A, Schroth GP, Smith ME, Smith VP, 23. Mauri C, Blair PA. 2010. Regulatory B cells in
Spiridou A, Torrance PE, Tzonev SS, autoimmunity: developments and controver-
Vermaas EH, Walter K, Wu X, Zhang L, sies. Nat Rev Rheumatol 6:636643.
Alam MD, Anastasi C, Aniebo IC, Bailey DM, 24. Mroczek ES, Ippolito GC, Rogosch T, Hoi
Bancarz IR, Banerjee S, Barbour SG, KH, Hwangpo TA, Brand MG, Zhuang Y, Liu
Baybayan PA, Benoit VA, Benson KF, Bevis CR, Schneider DA, Zemlin M, Brown EE,
C, Black PJ, Boodhun A, Brennan JS, Georgiou G, Schroeder HW Jr. 2014. Differences
Bridgham JA, Brown RC, Brown AA, in the composition of the human antibody
Buermann DH, Bundu AA, Burrows JC, repertoire by B cell subsets in the blood. Front
Carter NP, Castillo N, Chiara ECM, Chang Immunol 5:96. doi:10.3389/fimmu.2014.00096.
S, Neil Cooley R, Crake NR, Dada OO, 25. Frohman MA, Dush MK, Martin GR. 1988.
Diakoumakos KD, Dominguez-Fernandez B, Rapid production of full-length cDNAs from
Earnshaw DJ, Egbujor UC, Elmore DW, rare transcripts: amplification using a single
Etchin SS, Ewan MR, Fedurco M, Fraser gene-specific oligonucleotide primer. Proc
LJ, Fuentes Fajardo KV, Scott Furey W, Natl Acad Sci USA 85:89989002.
George D, Gietzen KJ, Goddard CP, Golda 26. Aoki-Ota M, Torkamani A, Ota T, Schork N,
GS, Granieri PA, Green DE, Gustafson DL, Nemazee D. 2012. Skewed primary Igkappa
Hansen NF, Harnish K, Haudenschild CD, repertoire and V-J joining in C57BL/6 mice:
Heyer NI, Hims MM, Ho JT, Horgan AM, implications for recombination accessibility and
Hoschler K, Hurwitz S, Ivanov DV, Johnson receptor editing. J Immunol 188:23052315.
MQ, James T, Huw Jones TA, Kang GD, 27. Choi NM, Loguercio S, Verma-Gaur J,
Kerelska TH, Kersey AD, Khrebtukova I, Degner SC, Torkamani A, Su AI, Oltz EM,
Kindwall AP, Kingsbury Z, Kokko-Gonzales Artyomov M, Feeney AJ. 2013. Deep sequencing
PI, Kumar A, Laurent MA, Lawley CT, Lee SE, of the murine IgH repertoire reveals complex
Lee X, Liao AK, Loch JA, Lok M, Luo S, regulation of nonrandom v gene rearrangement
Mammen RM, Martin JW, McCauley PG, frequencies. J Immunol 191:23932402.
McNitt P, Mehta P, Moon KW, Mullens JW, 28. van Dongen JJ, Langerak AW, Bruggemann
Newington T, Ning Z, Ling Ng B, Novo SM, M, Evans PA, Hummel M, Lavender FL,
ONeill MJ, Osborne MA, Osnowski A, Ostadan Delabesse E, Davi F, Schuuring E, Garcia-
O, Paraschos LL, Pickering L, Pike AC, Chris Sanz R, van Krieken JH, Droese J, Gonzalez
Pinkard D, Pliskin DP, Podhasky J, Quijano VJ, D, Bastard C, White HE, Spaargaren M,
Raczy C, Rae VH, Rawlings SR, Chiva Gonzalez M, Parreira A, Smith JL, Morgan
Rodriguez A, Roe PM, Rogers J, Rogert GJ, Kneba M, Macintyre EA. 2003. Design
Bacigalupo MC, Romanov N, Romieu A, Roth and standardization of PCR primers and
RK, Rourke NJ, Ruediger ST, Rusman E, protocols for detection of clonal immunoglob-
Sanches-Kuiper RM, Schenker MR, Seoane ulin and T-cell receptor gene recombinations
JM, Shaw RJ, Shiver MK, Short SW, Sizto NL, in suspect lymphoproliferations: report of the
Sluis JP, Smith MA, Ernest Sohna Sohna J, BIOMED-2 Concerted Action BMH4-CT98-
Spence EJ, Stevens K, Sutton N, Szajkowski L, 3936. Leukemia 17:22572317.
Tregidgo CL, Turcatti G, Vandevondele S, 29. Boyd SD, Marshall EL, Merker JD, Maniar
Verhovsky Y, Virk SM, Wakelin S, Walcott JM, Zhang LN, Sahaf B, Jones CD, Simen BB,
GC, Wang J, Worsley GJ, Yan J, Yau L, Zuerlein Hanczaruk B, Nguyen KD, Nadeau KC,
M, Mullikin JC, Hurles ME, McCooke NJ, West Egholm M, Miklos DB, Zehnder JL, Fire
JS, Oaks FL, Lundberg PL, Klenerman D, AZ. 2009. Measurement and clinical monitoring
Durbin R, Smith AJ. 2008. Accurate whole of human lymphocyte clonality by massively
human genome sequencing using reversible parallel VDJ pyrosequencing. Sci Transl Med
terminator chemistry. Nature 456:5359. 1:12ra23. doi:10.1126/scitranslmed.3000540.
21. Wang C, Liu Y, Xu LT, Jackson KJ, Roskin 30. Briney BS, Willis JR, McKinney BA, Crowe
KM, Pham TD, Laserson J, Marshall EL, Seo JE Jr. 2012. High-throughput antibody sequenc-
K, Lee JY, Furman D, Koller D, Dekker CL, ing reveals genetic evidence of global regulation of
Davis MM, Fire AZ, Boyd SD. 2014. Effects of the naive and memory repertoires that extends
aging, cytomegalovirus infection, and EBV infec- across individuals. Genes Immun 13:469473.
360 BOYD AND JOSHI

31. Wu X, Zhou T, Zhu J, Zhang B, Georgiev I, toire sequencing. Proc Natl Acad Sci USA
Wang C, Chen X, Longo NS, Louder M, 110:1346313468.
McKee K, ODell S, Perfetto S, Schmidt SD, 39. Shiroguchi K, Jia TZ, Sims PA, Xie XS. 2012.
Shi W, Wu L, Yang Y, Yang ZY, Yang Z, Digital RNA sequencing minimizes sequence-
Zhang Z, Bonsignori M, Crump JA, Kapiga dependent bias and amplification noise with
SH, Sam NE, Haynes BF, Simek M, Burton optimized single-molecule barcodes. Proc Natl
DR, Koff WC, Doria-Rose NA, Connors M, Acad Sci USA 109:13471352.
Mullikin JC, Nabel GJ, Roederer M, Shapiro 40. Miner BE, Stoger RJ, Burden AF, Laird CD,
L, Kwong PD, Mascola JR. 2011. Focused Hansen RS. 2004. Molecular barcodes detect
evolution of HIV-1 neutralizing antibodies redundancy and contamination in hairpin-
revealed by structures and deep sequencing. bisulfite PCR. Nucleic Acids Res 32:e135.
Science 333:15931602. doi:10.1093/nar/gnh132.
32. Scheid JF, Mouquet H, Ueberheide B, Diskin 41. McCloskey ML, Stoger R, Hansen RS, Laird
R, Klein F, Oliveira TY, Pietzsch J, Fenyo D, CD. 2007. Encoding PCR products with batch-
Abadir A, Velinzon K, Hurley A, Myung S, stamps and barcodes. Biochem Genet 45:761767.
Boulad F, Poignard P, Burton DR, Pereyra F, 42. Kinde I, Wu J, Papadopoulos N, Kinzler KW,
Ho DD, Walker BD, Seaman MS, Bjorkman Vogelstein B. 2011. Detection and quantification
PJ, Chait BT, Nussenzweig MC. 2011. Se- of rare mutations with massively parallel sequenc-
quence and structural convergence of broad ing. Proc Natl Acad Sci USA 108:95309535.
and potent HIV antibodies that mimic CD4 43. DeKosky BJ, Ippolito GC, Deschner RP,
binding. Science 333:16331637. Lavinder JJ, Wine Y, Rawlings BM, Varadarajan
33. Parameswaran P, Liu Y, Roskin KM, Jackson N, Giesecke C, Dorner T, Andrews SF, Wilson
KK, Dixit VP, Lee JY, Artiles KL, Zompi S, PC, Hunicke-Smith SP, Willson CG, Ellington
Vargas MJ, Simen BB, Hanczaruk B, AD, Georgiou G. 2013. High-throughput se-
McGowan KR, Tariq MA, Pourmand N, Koller quencing of the paired human immunoglobulin
D, Balmaseda A, Boyd SD, Harris E, Fire AZ. heavy and light chain repertoire. Nat Biotechnol
2013. Convergent antibody signatures in human 31:166169.
dengue. Cell Host Microbe 13:691700. 44. Georgiou G, Ippolito GC, Beausang J, Busse
34. Rubelt F, Sievert V, Knaust F, Diener C, Lim CE, Wardemann H, Quake SR. 2014. The
TS, Skriner K, Klipp E, Reinhardt R, Lehrach promise and challenge of high-throughput
H, Konthur Z. 2012. Onset of immune senescence sequencing of the antibody repertoire. Nat
defined by unbiased pyrosequencing of human Biotechnol 32:158168.
immunoglobulin mRNA repertoires. PloS One 7: 45. Munshaw S, Kepler TB. 2010. SoDA2: a
e49774. doi:10.1371/journal.pone.0049774. Hidden Markov Model approach for identifi-
35. Tan YC, Blum LK, Kongpachith S, Ju CH, Cai cation of immunoglobulin rearrangements.
X, Lindstrom TM, Sokolove J, Robinson WH. Bioinformatics 26:867872.
2014. High-throughput sequencing of natively 46. Gaeta BA, Malming HR, Jackson KJ, Bain
paired antibody chains provides evidence ME, Wilson P, Collins AM. 2007. iHMMune-
for original antigenic sin shaping the anti- align: hidden Markov model-based alignment and
body response to influenza vaccination. Clin identification of germline genes in rearranged
Immunol 151:5565. immunoglobulin gene sequences. Bioinformatics
36. Loman NJ, Misra RV, Dallman TJ, Constantinidou 23:15801587.
C, Gharbia SE, Wain J, Pallen MJ. 2012. 47. Giudicelli V, Brochet X, Lefranc MP. 2011.
Performance comparison of benchtop high- IMGT/V-QUEST: IMGT standardized analysis
throughput sequencing platforms. Nat Biotechnol of the immunoglobulin (IG) and T cell receptor
30:434439. (TR) nucleotide sequences. Cold Spring Harb
37. Jiang N, He J, Weinstein JA, Penland L, Protoc 2011:695715.
Sasaki S, He XS, Dekker CL, Zheng NY, 48. Ye J, Ma N, Madden TL, Ostell JM. 2013.
Huang M, Sullivan M, Wilson PC, Greenberg IgBLAST: an immunoglobulin variable domain
HB, Davis MM, Fisher DS, Quake SR. 2013. sequence analysis tool. Nucleic Acids Res 41:
Lineage structure of the human antibody W34W40.
repertoire in response to influenza vaccina- 49. Li S, Lefranc MP, Miles JJ, Alamyar E,
tion. Sci Transl Med 5:171ra119. doi:10.1126/ Giudicelli V, Duroux P, Freeman JD, Corbin
scitranslmed.3004794. VD, Scheerlinck JP, Frohman MA, Cameron
38. Vollmers C, Sit RV, Weinstein JA, Dekker PU, Plebanski M, Loveland B, Burrows SR,
CL, Quake SR. 2013. Genetic measurement Papenfuss AT, Gowans EJ. 2013. IMGT/
of memory B-cell recall using antibody reper- HighV QUEST paradigm for T cell receptor
CHAPTER 20 HTS Analysis of Antibody Repertoires 361

IMGT clonotype diversity and next generation Lu X, Yu JS, Hwang KK, Gao F, Markowitz
repertoire immunoprofiling. Nat Commun M, Heath S L, Bar KJ, Goep fert PA,
4:2333. doi:10.1038/ncomms3333. Montefiori DC, Shaw GC, Alam SM, Margolis
50. Kidd MJ, Chen Z, Wang Y, Jackson KJ, DM, Denny TN, Boyd SD, Marshal E, Egholm
Zhang L, Boyd SD, Fire AZ, Tanaka MM, M, Simen BB, Hanczaruk B, Fire AZ, Voss G,
Gaeta BA, Collins AM. 2012. The inference of Kelsoe G, Tomaras GD, Moody MA, Kepler
phased haplotypes for the immunoglobulin H TB, Haynes BF. 2011. Initial antibodies bind-
chain V region gene loci by analysis of VDJ gene ing to HIV-1 gp41 in acutely infected subjects
rearrangements. J Immunol 188:13331340. are polyreactive and highly mutated. J Exp
51. Glanville J, Kuo TC, von Budingen HC, Guey Med 208:22372249.
L, Berka J, Sundar PD, Huerta G, Mehta GR, 60. Kwong PD, Mascola JR. 2012. Human anti-
Oksenberg JR, Hauser SL, Cox DR, Rajpal A, bodies that neutralize HIV-1: identification,
Pons J. 2011. Naive antibody gene-segment structures, and B cell ontogenies. Immunity
frequencies are heritable and unaltered by chronic 37:412425.
lymphocyte ablation. Proc Natl Acad Sci USA 61. Liao HX, Lynch R, Zhou T, Gao F, Alam SM,
108:2006620071. Boyd SD, Fire AZ, Roskin KM, Schramm CA,
52. Briney BS, Willis JR, Crowe JE Jr. 2012. Zhang Z, Zhu J, Shapiro L, Mullikin JC,
Location and length distribution of somatic Gnanakaran S, Hraber P, Wiehe K, Kelsoe G,
hypermutation-associated DNA insertions and Yang G, Xia SM, Montefiori DC, Parks R,
deletions reveals regions of antibody structural Lloyd KE, Scearce RM, Soderberg KA, Cohen
plasticity. Genes Immun 13:523529. M, Kamanga G, Louder MK, Tran LM, Chen
53. Briney BS, Willis JR, Hicar MD, Thomas JW Y, Cai F, Chen S, Moquin S, Du X, Joyce MG,
II, Crowe JE Jr. 2012. Frequency and genetic Srivatsan S, Zhang B, Zheng A, Shaw GM,
characterization of V(DD)J recombinants in Hahn BH, Kepler TB, Korber BT, Kwong PD,
the human peripheral blood antibody reper- Mascola JR, Haynes BF. 2013. Co-evolution of
toire. Immunology 137:5664. a broadly neutralizing HIV-1 antibody and
54. Larimore K, McCormick MW, Robins HS, founder virus. Nature 496:469476.
Greenberg PD. 2012. Shaping of human 62. Zhou T, Zhu J, Wu X, Moquin S, Zhang B,
germline IgH repertoires revealed by deep Acharya P, Georgiev IS, Altae-Tran HR,
sequencing. J Immunol 189:32213230. Chuang GY, Joyce MG, Do Kwon Y, Longo
55. Wu YC, Kipling D, Leong HS, Martin V, NS, Louder MK, Luongo T, McKee K,
Ademokun AA, Dunn-Walters DK. 2010. Schramm CA, Skinner J, Yang Y, Yang Z,
High-throughput immunoglobulin repertoire Zhang Z, Zheng A, Bonsignori M, Haynes BF,
analysis distinguishes between human IgM Scheid JF, Nussenzweig MC, Simek M,
memory and switched memory B-cell popula- Burton DR, Koff WC, Mullikin JC, Connors
tions. Blood 116:10701078. M, Shapiro L, Nabel GJ, Mascola JR, Kwong
56. Uduman M, Yaari G, Hershberg U, Stern JA, PD. 2013. Multidonor analysis reveals structural
Shlomchik MJ, Kleinstein SH. 2011. Detecting elements, genetic determinants, and maturation
selection in immunoglobulin sequences. Nucleic pathway for HIV-1 neutralization by VRC01-class
Acids Res 39:W499W504. antibodies. Immunity 39:245258.
57. Yaari G, Uduman M, Kleinstein SH. 2012. 63. Zhu J, Wu X, Zhang B, McKee K, ODell
Quantifying selection in high-throughput Im- S, Soto C, Zhou T, Casazza JP, Mullikin
munoglobulin sequencing data sets. Nucleic JC, Kwong PD, Mascola JR, Shapiro L. 2013.
Acids Res 40:e134. doi:10.1093/nar/gks457. De novo identification of VRC01 class HIV-1-
58. Sok D, Laserson U, Laserson J, Liu Y, neutralizing antibodies by next-generation
Vigneault F, Julien JP, Briney B, Ramos A, sequencing of B-cell transcripts. Proc Natl
Saye KF, Le K, Mahan A, Wang S, Kardar M, Acad Sci USA 110:E4088E4097.
Yaari G, Walker LM, Simen BB, St John EP, 64. Doria-Rose NA, Schramm CA, Gorman J,
Chan-Hui PY, Swiderek K, Kleinstein SH, Alter Moore PL, Bhiman JN, Dekosky BJ,
G, Seaman MS, Chakraborty AK, Koller D, Ernandes MJ, Georgiev IS, Kim HJ, Pancera
Wilson IA, Church GM, Burton DR, Poignard M, Staupe RP, Altae-Tran HR, Bailer RT,
P. 2013. The effects of somatic hypermutation on Crooks ET, Cupo A, Druz A, Garrett NJ, Hoi
neutralization and binding in the PGT121 family of KH, Kong R, Louder MK, Longo NS, McKee
broadly neutralizing HIV antibodies. PLoS Pathog K, Nonyane M, ODell S, Roark RS, Rudicell
9:e1003754. doi:10.1371/journal.ppat.1003754. RS, Schmidt SD, Sheward DJ, Soto C, Wibmer
59. Liao HX, Chen X, Munshaw S, Zhang R, CK, Yang Y, Zhang Z, Nisc Comparative
Marshall DJ, Vandergrift N, Whitesides JF, Sequencing Program, Mullikin JC, Binley JM,
362 BOYD AND JOSHI

Sanders RW, Wilson IA, Moore JP, Ward AB, lar polysaccharide of Streptococcus pneumo-
Georgiou G, Williamson C, Abdool Karim SS, niae type 23F. Infect Immun 70:40834091.
Morris L, Kwong PD, Shapiro L, Mascola JR. 68. Lucas AH, McLean GR, Reason DC, OConnor
2014. Developmental pathway for potent V1V2- AP, Felton MC, Moulton KD. 2003. Molecular
directed HIV-neutralizing antibodies. Nature ontogeny of the human antibody repertoire to the
509:5562. Haemophilus influenzae type B polysaccharide:
65. Ademokun A, Wu YC, Martin V, Mitra R, expression of canonical variable regions and their
Sack U, Baxendale H, Kipling D, Dunn- variants in vaccinated infants. Clin Immunol
Walters DK. 2011. Vaccination-induced changes 108:119127.
in human B-cell repertoire and pneumococcal 69. Jackson KJL, Liu Y, Roskin KM, Glanville J,
IgM and IgA antibody at different ages. Aging Hoh RA, Seo K, Marshall EL, Gurley TC, Moody
Cell 10:922930. MA, Haynes BF, Walter EB, Liao H, Albrecht
66. Chao A. 1987. Estimating the population size RA, Garca-Sastre A, Chaparro-Riggers J, Rajpal
for capture-recapture data with unequal catch- A, Pons J, Simen BB, Hanczaruk B, Dekker CL,
ability. Biometrics 43:783791. Laserson J, Koller D, Davis MM, Fire AZ, Boyd
67. Zhou J, Lottenbach KR, Barenkamp SJ, SD. 2014. Human responses to influenza vaccina-
Lucas AH, Reason DC. 2002. Recurrent var- tion show seroconversion signatures and conver-
iable region gene usage and somatic mutation gent antibody rearrangements. Cell Host Microbe
in the human antibody response to the capsu- doi:10.1016/j.chom.2014.05.013.
Antibody Informatics: IMGT,
the International ImMunoGeneTics
Information System

MARIE-PAULE LEFRANC1
21
INTRODUCTION

The efficiency of the adaptive immune response and its capability of recognizing
a large number of different antigens depend on the huge diversity of the antigen
receptors, immunoglobulins (IG), or antibodies of the B lymphocytes and T cell
receptors (TR) of the T lymphocytes. The genes that code the IG and TR are
highly polymorphic and are organized in clusters in several loci (three loci for IG
and four for TR in humans) located on different chromosomes (four in humans) in
the genome (1, 2). The molecular synthesis of the IG and TR chains is particularly
complex and unique. It includes several mechanisms that occur at the DNA level:
combinatorial rearrangements of the variable (V), diversity (D), and joining (J)
genes that code the IG and TR variable domain, exonuclease trimming at the ends
of the V, D, and J genes, and the random addition of nucleotides by the terminal
deoxynucleotidyltransferase (TdT) that create the junction N diversity and,
for IG, somatic hypermutations (1, 2). The IG and TR repertoires show an
extraordinary diversity with a potential of 1012 IG and 1012 TR per individual, and
the only limiting factor is the number of genetically programmed B and T cells for
an organism. Therefore, the analysis of the IG and TR genes and of their

1
IMGT, the international ImMunoGeneTics information system, Laboratoire dImmunoGntique Molculaire
LIGM, Universit Montpellier 2, Institut de Gntique Humaine IGH, UPR CNRS 1142, Montpellier, 34396 cedex 5,
France.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2014 American Society for Microbiology. All rights reserved.
doi:10.1128/microbiolspec.AID-0001-2012

363
364 LEFRANC

expression represents a crucial challenge for Information (NCBI) (20), in Entrez Gene
the understanding of the immune response in (NCBI, USA) when this database (now desig-
normal and pathological situations. nated as Gene) superseded LocusLink (21),
IMGT, the international ImMunoGeneTics in Ensembl at the European Bioinformatics
information system (http://www.imgt.org) Institute (EBI) (22), and in the Vertebrate
(3), was created in 1989 at Montpellier, France Genome Annotation (Vega) Browser (23) at
(CNRS and Universit Montpellier 2) to the Wellcome Trust Sanger Institute (UK).
standardize the immunogenetics data and to HGNC, Gene (NCBI), Ensembl, and Vega have
manage the huge diversity of the antigen direct links to IMGT/GENE-DB (18), which is
receptors, IG and TR (1, 2). New concepts, the global reference for IG and TR alleles
databases, and tools were developed by IMGT assigned by the WHO-IUIS-IMGT Nomencla-
to explore and to analyze the particular ture Subcommittee and, so far, the only
properties of the IG and TR, compared with database managing IG and TR alleles. Since
conventional genes and proteins. They led to 2008, IMGT gene and allele names have been
the emergence of a new science, immuno- used by the WHO-International Nonpropri-
informatics, and, owing to the huge develop- etary Names (WHO-INN) program in the
ment of therapeutic antibodies, to antibody definition of therapeutic monoclonal anti-
informatics, the part of immunoinformatics bodies (mAb, INN suffix -mab), fusion proteins
dealing with IG. for immune applications (FPIA, INN suffix
-cept), and composite proteins for clinical
applications (CPCA) with Fc for increased
half-life (24, 25), and the corresponding se-
THE BASICS FOR ANTIBODY
quences and data have been entered in IMGT/
INFORMATICS
2Dstructure-DB (26) and in IMGT/mAb-DB
(27) (http://www.imgt.org).
IMGT Gene and Allele Nomenclature
The accuracy and the consistency of the IMGT
IMGT Unique Numbering
data are based on IMGT-ONTOLOGY, the
first and, so far, unique ontology for immuno- A second IMGT-ONTOLOGY revolutionizing
genetics and immunoinformatics (4, 5, 6, 7, 8, 9, concept was the IMGT unique numbering
10). IMGT-ONTOLOGY defined, for the first (28, 29, 30, 31, 32, 33), which, with its two-
time, the concept of genes for the IG and TR, dimensional (2D) representation, the IMGT
which led to their gene and allele nomencla- Collier de Perles (34, 35, 36, 37, 38), bridged for
ture (e.g., IGHV1-2*01) and allowed their entry the first time, and definitively, the gap between
in databases and tools (11). The IMGT IG and sequences and structures for the variable (V)
TR gene nomenclature (1, 2, 12, 13) was and constant (C) domains of the immunoglob-
approved at the international level by the ulin superfamily (IgSF) proteins (28, 29, 30, 31,
Human Genome Organisation (HUGO) No- 32), and for the groove (G) domains of the
menclature Committee (HGNC) in 1999 (14, major histocompatibility (MH) superfamily
15) and endorsed by the World Health Orga- (MhSF) (32, 33). The V domains include the
nization - International Union of Immunolog- V-DOMAIN of the IG and TR and the V-LIKE-
ical Societies (WHO-IUIS) Nomenclature DOMAIN of the IgSF other than IG and TR; the
Committee (16, 17). The IMGT IG and TR C domains include the C-DOMAIN of the IG
gene names are the official reference for the and TR and the C-LIKE-DOMAIN of the IgSF
genome projects and, as such, have been other than IG and TR; whereas the G domains
entered in IMGT/GENE-DB (18), in the include the G-DOMAIN of the MH and the
Genome Database (GDB) (19), in LocusLink G-LIKE-DOMAIN of the MhSF other than
at the National Center for Biotechnology MH (28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38).
CHAPTER 21 Antibody Informatics: IMGT 365

IMGT Anchors for V and C Domains V-DOMAIN), forming a barrel structure or


One of the IMGT unique numbering major sandwich with a hydrophobic inner core,
breakthroughs for antibody informatics was to and three loops (designated as complementar-
define anchors in the V and C domains, a new ity determining regions (CDR) or hyper-
and basic concept for both sequences and variable regions in a V-DOMAIN) (28, 29, 30)
structures, that definitively solved the hetero- (Fig. 1A). Compared with a V domain, a C
geneity and complexity of previous num- domain has seven antiparallel beta strands
berings, and also is valid for both V and C instead of nine, two loops instead of three, and
domains (28, 29, 30, 31). A V domain is made of a characteristic CD transverse strand (in the
nine antiparallel beta strands on two layers absence of a C strand, CC loop, and C
(designated as framework regions (FR) in a strand) (31) (Fig. 1B).

FIGURE 1 IMGT Collier de Perles for V-DOMAIN and C-DOMAIN. A, VH and V-KAPPA. B, CH1 and C-KAPPA. The VH,
V-KAPPA, CH1, and C-KAPPA of the motavizumab antibody are shown as examples. IMGT Colliers de Perles are
shown on one layer (top), on two layers (middle) (INN, from IMGT/2Dstructure-DB, http://www.imgt.org), and on
two layers with hydrogen bonds (bottom) (3ixt_H and 3ixt_L, from IMGT/3Dstructure-DB, http://www.imgt.org)
(26, 48). In the V-DOMAIN, anchors (positions 26 and 39, 55 and 66, 104 and 118) support the three CDR (CDR1-
IMGT, CDR2-IMGT, and CDR3-IMGT that correspond to the BC, CC, and FG loops, respectively) (29, 30, 32). The
VH and V-KAPPA CDR-IMGT lengths are [10.7.12] and [5.3.9], respectively. In the C-DOMAIN, anchors (positions
26 and 39, 104 and 118) support the BC and FG loops as in a V-DOMAIN, whereas in the absence of a C strand, CC
loops, and C strand, anchors 45 and 77 delimit the CD transverse strand (31, 32). A ban symbol indicates an
error (K instead of CH1 R120) in the 3D structure PDB le (3ixt), detected by using the IMGT unique numbering
and by comparison with the INN sequence. A similar type of error (A instead of CH1 V121) has also been detected
by IMGT for the 3D structure of the anti-HIV b12 antibody (1hzh). doi:10.1128/microbiolspec.AID-0001-2012.f1
366 LEFRANC

The IMGT anchors define the positions of Highly Conserved Amino Acids
the V or C domain strands that support the In the IMGT unique numbering, amino acids
loops. Anchors are shown in squares in IMGT always have the same position number. Highly
Colliers de Perles (Fig. 1). In a V domain, these conserved amino acids of an IG or TR V and C
anchors comprise the positions 26 and 39, 55 domains are written in red online, in the IMGT
and 66, 104 and 118 that support the BC, CC, Colliers de Perles. They comprise five positions
and FG loops. In an IG or TR V-DOMAIN, these in a V-DOMAIN: cysteine C23 (1st-CYS), tryp-
anchors are the positions of the FR-IMGT that tophan W41 (CONSERVED-TRP), hydropho-
support the CDR-IMGT (28, 29, 30). In a C bic amino acid 89, cysteine C104 (2nd-CYS), and
domain, four anchors are identical to those of a phenylalanine F118 (J-PHE) or tryptophan
V domain (positions 26 and 39, 104 and 118 that W118 (J-TRP) (that belongs to the F/W-G-X-G
support the BC and FG loops) and two anchors motif encoded by the J-REGION) (Fig. 1A). In a
(positions 45 and 77) delimit the CD strand (31). Cdomain,therearefourhighlyconservedamino
acids: C23 (1st-CYS), W41 (CONSERVED-
CDR-IMGT Lengths for Antibody TRP), hydrophobic 89, and C104 (2nd-CYS),
Humanization by Grafting instead of five.
By allowing a correct delimitation of the FR-
IMGT and CDR-IMGT, anchors led to the
IMGT Standardized and
CDR-IMGT lengths concept. The CDR-
Integrated System
IMGT lengths are indicated between brackets
and separated by dots, e.g., VH [10.7.12] and In 2013, IMGT comprised seven databases,
V-KAPPA [5.3.9] (see legend to Fig. 1). They seventeen online tools, and more than 15,000
are essential information for antibody char- pages of Web resources and is the reference
acterization, used in IMGT databases and in immunogenetics and immunoinformatics.
tools and in the INN mAb definitions (24) and, IMGT-ONTOLOGY concepts (identification,
experimentally, for antibody humanization. description, classification (11), numerotation
The CDR-IMGT lengths avoid unnecessary (32, 37), and detailed protocols (IMGT/V-
extrapolation on canonical structures if the QUEST [44], IMGT/JunctionAnalysis [45],
antibody has not yet been crystallized, while, IMGT/DomainGapAlign [47], IMGT/
at the same time, they provide the structural 3Dstructure-DB [48], IMGT/Collier-de-
delimitations for the V-domain interactions Perles [38]) have been published in Cold
withtheantigen(39,40,41,42),asdemonstrated Spring Harbor Protocols (IMGT booklet
by the contact analysis and paratope/epitope freely downloadable in IMGT references, http://
details of known 3D structures (40, 41, 42). www.imgt.org).
Moreover, the CDR-IMGT lengths are used The next sections illustrate the use of IMGT
whatever the chain type (e.g., heavy, light kappa tools for the standardized IG sequences and
or lambda for IG) and the species (e.g., human, structures analysis for the IG repertoire anal-
mouse, rabbit, camel), which is particularly ysis in infectious diseases, antibody engineer-
useful for domain comparison in antibody ing and humanization, and study of antibody/
humanization by CDR grafting. For all these antigen interactions. Nucleotide sequences
reasons, the CDR-IMGT lengths are key criteria from deep sequencing (Next Generation
for a good grafting in antibody humanization. Sequencing [NGS] or High Throughput Se-
Grafting CDR-IMGT on a germline-derived quencing [HTS]) of antibody V domains are
human FR-IMGT represents the definitive analyzed with IMGT/HighV-QUEST (43), the
answer to preserve the specificity and affinity high-throughput version of IMGT/V-QUEST
oftheoriginalantibodyandtoobtain,atthesame and IMGT/JunctionAnalysis (44, 45, 46).
time, a humanized antibody that is the most Amino acid sequences of V and C domains are
human as possible (39, 40, 41, 42). represented with the IMGT/Collier-de-Perles
CHAPTER 21 Antibody Informatics: IMGT 367

tool (38) and analyzed with IMGT/Domain


IMGT/HighV-QUEST for
GapAlign (26, 47). Three-dimensional (3D)
Deep Sequencing
structures (including contact analysis and
paratope/epitope) are described in IMGT/ Since October 2010, the analysis of IG and TR
3Dstructure-DB (26, 48). Based on a friendly nucleotide sequences obtained from deep
interface, IMGT/mAb-DB (27) contains thera- sequencing (NGS or HTS) can be performed
peutic monoclonal antibodies (and also FPIA with IMGT/HighV-QUEST (43, 46) (http://
and CPCA with Fc) that can be queried on their www.imgt.org) that analyzes 500,000 se-
specificity, for example, in infectious diseases, quences per run and performs statistical
on bacterial or viral targets. analysis on the results (version July 2012).
The IMGT/HighV-QUEST Search page is
very similar to the IMGT/V-QUEST Search
page (44, 46). The results are provided in a
ANALYSIS OF ANTIBODY V-DOMAIN
downloadable main folder with eleven files in
NUCLEOTIDE SEQUENCES
CSV format (results equivalent to those of the
Excel file of IMGT/V-QUEST online), and one
IMGT/V-QUEST
folder with the individual files (up to 500,000)
Since 1997, IMGT/V-QUEST (44, 46), a highly of all the sequences results (results identical
customized and integrated IMGT online tool, for each analyzed sequence to those of IMGT/
has been the reference for the standardized V-QUEST online in Text, and corresponding
analysis of IG and TR rearranged V-domain to Detailed view) (43, 44, 46). Text and CSV
nucleotide sequences. IMGT/V-QUEST iden- formats were chosen to facilitate statistical
tifies the V, D, and J genes and alleles by studies for further interpretation and knowl-
alignment with the germline IG and TR gene edge extraction. The eleven files of the main
and allele sequences of the IMGT reference folder provide, per analysis, a total of 513 to 576
directory and, for the IG, the nucleotide (nt) columns of results (Table 1) and demonstrate
mutations and amino acid (AA) changes that IMGT standards, based on IMGT-
resulting from somatic hypermutations. The ONTOLOGY concepts, fit remarkably well
tool identifies the hot-spot positions in the V- with extensive and detailed NGS data analysis
REGION of the closest germline V gene. for antibody informatics. The results include
IMGT/V-QUEST integrates IMGT/ the identification of the closest V and J genes
JunctionAnalysis (45), for the detailed analysis and alleles (and D genes for VH), detailed
of the V-J and V-D-J junctions and IMGT/ IMGT/JunctionAnalysis results, analysis of
Automat for a complete sequence annotation the nucleotide (nt) sequence mutations, and
of the sequences, and also provides IMGT analysis of the amino acid (AA) changes in the
Collier de Perles (37, 38). Customized sequence translations. The AA changes are
parameters and results provided by IMGT/ described for the hydropathy (3 classes),
V-QUEST and IMGT/JunctionAnalysis have volume (5 classes), and IMGT physicochem-
been detailed for antibody informatics (39, 40, ical properties (11 classes that comprise ali-
41, 42). The IMGT reference directories phatic [A, V, I, L], acid [D, E], basic [H, K, R],
against which the tools compare user se- amide [N, Q], hydroxyl [S, T], sulfur [C, M], G,
quences are regularly updated with new IG P, Y, F, and W) (49) (IMGT Aide-mmoire, in
and TR data from recently sequenced ge- the Amino acids section, http://www.imgt.
nomes (rat, rabbit, etc.), annotated in IMGT/ org). Thus, S40>G (+ + ) means that the two
LIGM-DB, the IMGT nucleotide sequence AA involved in the change (S>G) at codon 40
database (http://www.imgt.org), and IMGT/ belong to the same hydropathy (+) and volume
GENE-DB, the IMGT gene and allele database (+) classes but to different physicochemical
(18). properties () classes (49). It is the first time
368
TABLE 1 List of the IMGT/HighV-QUESTa results les with number of columns and results content
File number File name Number of columns Results contentb

LEFRANC
1 Summary 2529 Identity percentage with the closest V, D, and J genes and alleles
FR-IMGT and CDR-IMGT lengths
Amino acid (AA) JUNCTION
Description of insertions and deletions if any
2 IMGT-gapped- 18 Nucleotide (nt) sequences gapped according to the IMGT unique numbering for V-D-J-REGION, V-J-REGION,
nt-sequences V-REGION, FR1-IMGT, CDR1-IMGT, FR2-IMGT, CDR2-IMGT, FR3-IMGT
nt sequences of CDR3-IMGT, JUNCTION, J-REGION and FR4-IMGT
3 Nt-sequences 6378 nt sequences of all labels that can been automatically annotated by IMGT/Automat.
4 IMGT-gapped- 18 AA sequences gapped according to the IMGT unique numbering for the labels V-D-J-REGION, V-J-REGION,
AA-sequences V-REGION, FR1-IMGT, CDR1-IMGT, FR2-IMGT, CDR2-IMGT, FR3-IMGT
AA sequences of CDR3-IMGT, JUNCTION, J-REGION and FR4-IMGT
5 AA-sequences 18 Same columns as IMGT-gapped-AA-sequences (#4), but sequences of labels are without IMGT gaps.
6 Junction 33, 46, 66, or 77 Results of IMGT/JunctionAnalysis (33 columns for IGL and IGK (also for TRA and TRG) sequences, 46 (if one D),
66 (if two D) or 77 (if 3 D) columns for IGH (also for TRB and TRD) sequences
7 V-REGION-mutation- 11 List of mutations (nt mutations, AA changes, AA class identity (+) or change ()) for V-REGION, FR1-IMGT,
and-AA-change table CDR1-IMGT, FR2-IMGT, CDR2-IMGT, FR3-IMGT, and germline CDR3-IMGT
8 V-REGION-nt- 130 Number (nb) of nt positions including IMGT gaps, nb of nt, nb of identical nt, total nb of mutations, nb of
mutation-statistics silent mutations, nb of nonsilent mutations, nb of transitions (a>g, g>a, c>t, t>c) and nb of transversions
(a>c, c>a, a>t, t>a, g>c, c>g, g>t, t>g) for V-REGION, FR1-IMGT, CDR1-IMGT, FR2-IMGT, CDR2-IMGT, FR3-IMGT,
and germline CDR3-IMGT
9 V-REGION-AA- 189 nb of AA positions including IMGT gaps, nb of AA, nb of identical AA, total nb of AA changes, nb of AA
change-statistics changes according to AAclassChangeType (+ + +, + + , + +, + , + , +, ), and nb of AA class
changes according to AAclassSimilarityDegree (nb of Very similar, nb of Similar, nb of Dissimilar, nb of Very
dissimilar) for V-REGION, FR1-IMGT, CDR1-IMGT, FR2-IMGT, CDR2-IMGT, FR3-IMGT, and germline CDR3-IMGT
10 V-REGION-mutation- 8 Hot spots motifs ((a/t)a, t(a/t), (a/g)g(c/t)(a/t), (a/t)(a/g)c(c/t) detected in the closest germline V-REGION with
hot spots positions in FR-IMGT and CDR-IMGT
11 Parameters Date of the analysis
IMGT/V-QUEST program version, IMGT/V-QUEST reference directory release
Parameters used for the analysis: species, receptor type or locus, IMGT reference directory set, and Advanced
parameters.
a
IMGT/HighV-QUEST (43), freely available for academics on the IMGT Home page (http://www.imgt.org), analyzes up to 500,000 sequences per run. Results in the 11 les are equivalent to
those of the Excel le of IMGT/V-QUEST online (46). The statistical analysis (tables and histograms) is performed on the Summary le content and on the CDR3-IMGT results (nt and AA) of
results of several runs up to 500,000 sequences and provided as PDF les and PNG gures (detailed in reference 43)
b
Files 1 to 10 comprise systematically sequence identication (name, functionality, and names of the closest V, D, and J genes and alleles).
CHAPTER 21 Antibody Informatics: IMGT 369

that such qualification of AA replacement is heavy (VH) and V light (VL) domains, the VL
provided. This has led us to identify four types being V kappa (V-KAPPA) or V lambda
of AA changes: very similar (+ + +), similar (++ (V-LAMBDA) in higher vertebrates, and V iota
, + +), dissimilar ( +, + , + ) and very (V-IOTA) in fish. The C domains correspond
dissimilar ( ) (44) (file 9 in Table 1). either to a complete C-REGION (C-KAPPA of
IMGT/HighV-QUEST performs statistical the IG-Light-Kappa chains or C-LAMBDA of
analysis on the results of several runs up to the IG-Light-Lambda chains) or to part of it (e.g.,
500,000 sequences. Detailed statistical analy- CH1, CH2, and CH3 of the IG-Heavy-Gamma
sis tables and histograms (e.g., V, D, and J chains) (28, 29, 30, 31,32).
usage, CDR3-IMGT [nt and AA lengths]), In IMGT Collier de Perles (Fig. 1), amino
provided as PDF reports and separate graph- acids are shown in the one-letter abbreviation.
ical elements (figures in PNG format), are Anchor positions are shown in squares.
described elsewhere (43). Hatched circles correspond to missing posi-
IMGT/HighV-QUEST achieved the same tions according to the IMGT unique number-
degree of resolution and high-quality results for ing (29, 30, 31, 32). Strand positions at which
antibody sequences analysis as IMGT/V- hydrophobic amino acids (hydropathy index
QUEST. Both tools use the same algorithm, with positive value: I, V, L, F, C, M, A) and
and the user can evaluate the quality of his/her tryptophan (W) are found in more than 50% of
sequences before IMGT/HighV-QUEST anal- sequences are shown with a blue background
ysis, by checking the results obtained with color. Arrows indicate the direction of the beta
IMGT/V-QUEST on a few sequences. The strands and their designations in 3D struc-
eventual limitations of the IG V domain nu- tures. The CDR-IMGT of the V-DOMAIN are
cleotide sequence analysis using IMGT/HighV- colored according to the IMGT color menu
QUEST are currently not on the antibody which indicates the type of rearrangement, V-
informatics side, but rather on the experimental J or V-D-J (1, 2). Thus, the IMGT color menu
and sequencing conditions that need to be for CDR1-IMGT, CDR2-IMGT, and CDR3-
improved (sequence length, avoiding PCR or IMGT is online red, orange, and purple for VH
sequencing errors) for a fully reliable and (encoded by a V-D-J-REGION resulting from
meaningful biological interpretation of the a V-D-J rearrangement), and blue, green, and
NGS or HTS results. By providing warnings green-blue for V-KAPPA or V-LAMBDA (en-
and classification of sequences in categories (1 coded by a V-J-REGION resulting from a V-J
copy, More than 1, single allele, several rearrangement) (Fig. 1A).
alleles), IMGT/HighV-QUEST helps the The IMGT Colliers de Perles of the V and C
user in evaluating his/her own data quality domains can be displayed on two layers (Fig. 1,
and therefore represents the reference tool for middle row) to get a graphical representation
antibody-standardized analysis of NGS or HTS closer to the 3D structure, and on two layers
data. with hydrogen bonds (Fig. 1, bottom row) if
the mAb has been crystallized and data are in
IMGT/3Dstructure-DB (26, 48).
ANTIBODY V- AND C-DOMAIN 2D
REPRESENTATIONS The IMGT/Collier-de-Perles Tool
The IMGT/Collier-de-Perles tool (38) (IMGT
IMGT Collier de Perles 2D Representation
Home page, http://www.imgt.org) allows the
IMGT Collier de Perles (34, 35, 36, 37, 38) is a users to draw IMGT Colliers de Perles, on
graphical 2D representation of domain, based one or two layers, starting from their own
on the IMGT unique numbering (28, 29, 30, 31, domain amino acid sequences. Sequences
32, 33). For the IG, the V domains include the V have to be gapped according to the IMGT
370 LEFRANC

unique numbering (using, for example, CH1, without light chain): V42>(F,Y), G49>(E,
IMGT/DomainGapAlign [47]). The IMGT/ Q), L50>(C,R) and W52>(F,G,L,W) (IMGT
Collier-de-Perles tool can be customized to Gene table in IMGT Repertoire, and IMGT
display the CDR-IMGT according to the Biotechnology page, http://www.imgt.org/
IMGT color menu or the amino acids ac- IMGTbiotechnology/Camel_IgG.html),
cording to their hydropathy, volume, or the 11 4. to predict important AA interactions,
IMGT physicochemical classes (49) (IMGT even in the absence of 3D structures, by
Aide-Mmoire, in the Amino acids section, analyzing the display of conserved hydrogen
http://www.imgt.org). bonds in crystallized V domains and C domains
(Fig. 1) (IMGT Collier de Perles on two layers
in IMGT/3Dstructure-DB [26, 48]). Of impor-
Importance for Antibody Engineering
tance for antibody humanization are the two
and Antibody Humanization
FR-IMGT positions, 39 and 40, that have
The IMGT Colliers de Perles are used in hydrogen bonds with a CDR2-IMGT and a
antibody engineering and antibody humaniza- CDR3-IMGT position, respectively: anchor 39
tion, and for the evaluation of the immunoge- with CDR2-IMGT 56 (in VH) or 57 (in VL), and
nicity of therapeutic monoclonal antibodies position 40 with CDR3-IMGT 105 (in both VH
(40, 41, 42). The information is particularly and VL). These positions eventually need to be
useful: kept from the original antibody (26, 48), and
1. to precisely define the CDR1-IMGT, 5. to localize the landmarks of the CH
CDR2-IMGT, and CDR3-IMGT to be grafted domain. These include the conserved N-
in antibody humanization design based on glycosylation site in the CH2 of the IGHG1,
CDR grafting. Amino acids of the CDR-IMGT IGHG2, IGHG3, and IGHG4, on the aspara-
loops are those involved in the interactions gine (Asn) N84.4 at the top of the DE turn.
with the antigen (40, 41, 42), There is also a polymorphic N-glycosylation
2. to compare the physicochemical prop- site N79 in the CH3 strand D of several alleles
erties of AA at given positions in the user of the IGHG3 (replaced by a lysine [Lys] K in
V-region sequences with those of the IMGT the other alleles) (Alignment of alleles in
Collier de Perles statistical profiles for the IMGT Repertoire, http://www.imgt.org). The
human expressed IGHV, IGKV, and IGLV positions used for the knobs-into-holes engi-
repertoires (49), and to identify AA that, given neering for heavy-chain dimerization and the
their positions, could be potentially immuno- production of bispecific IgG antibodies involve
genic in chimeric or poorly humanized anti- CH3 positions threonine (Thr) T22 (strand B)
bodies. The IMGT statistical profiles comprise and tyrosine (Tyr) Y86 (strand E) with the
61 positions with conserved properties, among following AA changes, T22>Y on one CH3
which 13 participate to the inner core, 10 to the domain and Y86>T on the other one (IMGT
inner surface of the ABED and GFCCC beta Biotechnology page, http://www.imgt.org).
sheet (inside of the sandwich), 21 to the outer
surface of the ABED beta sheet and of the F
strand, and 10 to turns (exposed to solvent),
ANALYSIS OF ANTIBODY V- AND
and seven to the VH-VL interface (detailed in
C-DOMAIN AMINO ACID SEQUENCES
Table 3 of reference 49),
3. to localize unusual amino acid changes,
IMGT/DomainGapAlign
for example, the four AA changes observed
between camel IGHV1 genes expressed in IMGT/DomainGapAlign (26, 47) analyzes the
conventional IgG1 and those expressed in amino acid sequences of the IG and TR
nonconventional IgG2 and IgG3 (dimer of variable (V) and constant (C) domains. Several
heavy gamma2 or gamma3 chains with deleted amino acid sequences can be analyzed
CHAPTER 21 Antibody Informatics: IMGT 371

simultaneously, provided that they belong to constituted by the closest human germline
the same domain type (V or C). IMGT/ IGHV2-70*01 and IGHJ6*03, whereas the
DomainGapAlign analyzes the user amino framework of the V-KAPPA has 5 AA differ-
acid domain sequences by comparison with ences (84/89 identical AA) with the frame-
the IMGT reference directory sets (translation work constituted by the closest human
of the germline V and J genes and of the C gene germline IGKV1-5*01 and IGKJ4*01 (Fig.
domains from IMGT/GENE-DB [18]). These 2A) (in a humanization with a good IMGT
reference amino acid sequences can be dis- grafting, the number of AA differences in the
played by querying IMGT/DomainDisplay framework is minimal and usually between 0
(IMGT Home page, http://www.imgt.org). and 2).
The IMGT/DomainGapAlign results in-
Antibody V Domain Amino Acid clude tables with the characteristics of the
Sequence Analysis AA changes (49) shown in strands and loops,
IMGT/DomainGapAlign identifies the closest and in FR-IMGT and CDR-IMGT. The IMGT
germline V-REGION and J-REGION alleles. Collier de Perles of the analyzed VH or VL
IMGT/DomainGapAlign displays the V region domain (V-D-J region or V-J region, respec-
amino acid sequences of the user aligned with tively) is also available with highlighted AA
the closest V and J regions (Fig. 2A) with differences (in pink circles online) with the
IMGT gaps and delimitations of the strands closest germline sequence.
(FR-IMGT) and loops (CDR-IMGT), ac-
cording to the IMGT unique numbering for Antibody C Domain Amino Acid
V domain (28, 29, 30, 32). For instance, for Sequence Analysis
motavizumab, the V-REGION of the VH IMGT/DomainGapAlign displays the C-
domain is identified as having 86.9% identity domain amino acid sequences of the user
with the Homo sapiens IGHV2-70*01 and the sequence aligned with the closest C-DOMAIN
V-REGION of the V-KAPPA as having 83.0% alleles (Fig. 2B) with IMGT gaps and deli-
identity with the H. sapiens IGKV1-5*01. If mitations of the strands, turns and loops,
several closest alleles are identified, the user according to the IMGT unique numbering for
can select the display of each corresponding C domain (31, 32). The IMGT unique number-
alignment. The amino acid sequence is also ing for C-DOMAIN contributes to antibody
displayed online, according to the IMGT color engineering and humanization by providing
menu, with the delimitations of the V-RE- a standardized description of the Gm, Am,
GION, J-REGION, and for VH domains, (N- and Km allotypes, and by establishing, for
D)-REGION. the first time, the correlation between the
The number of amino acid differences in G1m, G2m, G3m, A2m, and Km allotypes, and
the FR-IMGT and CDR-IMGT is one of the the IGHG1, IGHG2, IGHG3, IGHA2, and
criteria to evaluate the potential immuno- IGKC alleles, respectively (50).
genicity (39, 40, 41, 42). The framework of a For instance, IMGT/DomainGapAlign
VH domain comprises 91 positions (25, 17, shows that the C-REGION of motavizumab is
38, and 11 positions for FR1-, FR2-, FR3-, 100% identical with the IGHG1*03 allele in its
and FR4-IMGT, respectively, represented three CH domains and hinge region. The
as [25.17.38.11]), whereas the framework of a IGHG1*03 allele corresponds to the G1m3
VL domain comprises 89 positions (26, 17, allele (50), characterized by the G1m3 allo-
36, 10 positions for FR1-, FR2-, FR3-, and type (CH1 arginine [Arg] R120, associated to
FR4-IMGT, respectively, represented as CH1 isoleucine [Ileu] I103) (Fig. 2B), and to
[26.17.36.10]) (26, 47). Thus, for motavizumab, the isoallotypes nG1m17 (CH1 R120) and
the framework of the VH has 10 AA differ- nG1m1 (CH3 glutamate [Glu] E12 and methi-
ences (81/91 identical AA) with the framework onine [Met] M14) (50). The C-KAPPA of
372 LEFRANC

FIGURE 2 IMGT/DomainGapAlign alignments. A, VH and V-KAPPA. B, CH1 and C-KAPPA. The closest V-REGION
and J-REGION identied at the amino acid level are aligned with the user sequence (here, motavizumab INN
8693, as example). The VH and V-KAPPA are identied as having 86.9% and 83% identity at the amino acid level
with the Homo sapiens IGHV2-70*01 and IGKV1-5*01, respectively (% identity is shown in an upper section
online). Amino acid differences are indicated below the V and J alignments. The FR-IMGT and CDR-IMGT, strands
and loops are according to the IMGT unique numbering for V domain (28, 29, 30, 32). The CH1 and C-KAPPA are
identied as having 100% identity at the amino acid level with the H. sapiens IGHG1*03 CH1 and IGKC*01,
respectively. IMGT/DomainGapAlign displays the C-domain amino acid sequence of the user, with IMGT gaps
and delimitations of the strands, turns, and loops, according to the IMGT unique numbering (31, 32).
doi:10.1128/microbiolspec.AID-0001-2012.f2
CHAPTER 21 Antibody Informatics: IMGT 373

motavizumab is 100% identical to the IGKC*01 IMGT file, IMGT numbering comparison,
allele (Fig. 2B). IGKC*01 is one of the four References and links, and Printable card.
IGKC alleles that correspond to a Km3 allele,
characterized by an alanine (Ala) A45.1 (.1 for
IMGT/3Dstructure-DB Chain Details
the first position in the transverse CD strand)
and a valine (Val) V101 (50). The section Chain details of the IMGT/
The IMGT/DomainGapAlign results in- 3Dstructure-DB card comprises, for each
clude tables with the characteristics of the chain, information first on the chain itself,
AA changes (49) shown in strands, turns, and then per domain.
loops. The IMGT Collier de Perles of the
1. The information for each chain includes:
analyzed C domain is also available with
highlighted AA differences (in pink circles Chain ID (e.g., 3ixt_H),
online) with the closest allele domain Chain length in amino acids (e.g., 225),
sequence. IMGT chain description with the delimit-
ations of the different domains (e.g., VH-
CH1 = VH (1-120) [D1] + CH1 (121-210) [D2]),
Chain sequence with delimitations of the
ANTIBODY 3D STRUCTURES
regions and domains, highlighting of AA
(in orange color) that are different from
IMGT/3Dstructure-DB Card
the closest genes and alleles, and links to
IMGT/3Dstructure-DB (26, 48) (http://www. Sequence in FASTA format and to Sequence
imgt.org), the IMGT 3D structure database, is in IMGT format.
queried through a user-friendly interface and
2. The information for a V-DOMAIN, as an
different displays for the results can be chosen
example, includes:
from the database home page (48). Each entry
in the database is detailed in an IMGT/ IMGT domain description (e.g., VH
3Dstructure-DB card that provides access to (1-120) [D1]),
all sequence data related to that entry, and, in IMGT gene and allele name with the
particular, for antibodies, the paired heavy and percentage of identity for the V and a link
light chains, and for IG/antigen (Ag) com- to Alignment details
plexes, the ligands (if peptides or proteins). IMGT gene and allele name with the
IMGT uses the PDB code (4 letters and/or percentage of identity for the J as well as
numbers) as IMGT entry ID. An additional other alleles giving the same percentage of
letter separated by an underscore (_) identi- identity), and a link to Alignment details,
fies the different chains in a 3D structure. For 2D representation: links to IMGT Collier
example, the 3ixt entry that corresponds to the de Perles on one layer or IMGT Collier de
3D structure of an IG/antigen (Ag) complex Perles on two layers,
(motavizumab Fab in complex with the anti- Contact analysis: a link to Domain con-
gen ligand Fusion glycoprotein F1) comprises tacts (overview),
two crystallographic complexes (as indicated CDR-IMGT lengths (e.g., [10.7.12]),
in the column CC with the colors red and blue), Sheet composition (e.g., [ABDE][ACCC
with for one of them (CC1, red), the following FG]),
chains: 3ixt_H (VH-CH1) and 3ixt_L (L- the domain amino acid sequence with
KAPPA) for the IG Fab, and 3ixt_P for the CDR-IMGT delimitations and high-
antigen (ligand). Eight tabs are available at lighting of AA (in orange color) that are
the top of each card: Chain details, Contact different from the closest V and J genes
analysis, Paratope and epitope, 3D visuali- and alleles,
zation Jmol or QuickPDB, Renumbered the link to IMGT/DomainGapAlign results.
374 LEFRANC

IMGT/3Dstructure-DB Contact Analysis ture, Phi and Psi angles [in degrees] and
accessible surface area [ASA] [in square
The IMGT/3Dstructure-DB Contact analysis
angstroms]). The IMGT Residue@Position
(26, 48) provides extensive information on the
cards can be accessed directly from the amino
atom pair contacts between domains and/or
acid sequences of the IMGT/3Dstructure-DB
chains and on the internal contacts in an
card or from the IMGT Colliers de Perles, by
IMGT/3Dstructure-DB entry. Atom pair con-
clicking on one AA.
tacts are obtained in IMGT/3Dstructure-DB by
a local program in which atoms are considered
to be in contact when no water molecule can IMGT Paratope and Epitope
take place between them and are characterized
In an IG/Ag complex, the amino acids in
by their atom contact types (Noncovalent,
contact at the interface between the IG and
Polar, Hydrogen bond, etc.) and their atom
the Ag constitute the paratope on the IG
contact categories ([BB] Backbone/backbone,
surface, and the epitope on the Ag surface
[SS] Side chain/side chain, etc.) (26, 48).
(Fig. 3C). In IMGT/3Dstructure-DB, the
This information can be obtained at three
IMGT paratope and epitope of IG/Ag com-
different levels:
plexes is determined automatically by combin-
1. Domain contacts (overview), ing contact analysis with an interaction scoring
2. Domain pair contacts (DomPair) that function and are described in a standardized
provides information on the contacts be- way. Thus, the IG paratope of 3ixt
tween a pair of domains or between a domain (motavizumab Fab) (Fig. 3C) comprises
and a ligand (e.g., between the VH domain of amino acids of VH (3ixt_H chain) and of
motavizumab (3ixt_H chain) and the ligand V-KAPPA (3ixt_L chain). Fifteen amino acids
(3ixt_P chain) (Fig. 3A), or between the V- of the antibody, 8 from VH and 7 from
KAPPA domain of motavizumab (3ixt_L V-KAPPA, form the paratope (Fig. 3C). Eight
chain) and the ligand (3ixt_P chain) (Fig. 3B), of the 8 positions belong to the VH CDR-IMGT
3. Residue pair contacts, or more precisely, [10.7.12] (A35 to the CDR1-IMGT; W58, D59,
IMGT Residue@Position (R@P) pair con- and K64 to the CDR2-IMGT; and I109, F110,
tacts. N112, and F113 to the CDR3-IMGT). Seven of
the 7 positions belong to the V-KAPPA CDR-
R@P represents one of the major break- IMGT [5.3.9] (G37 and Y38 to the CDR1-IMGT;
throughs of the IMGT unique numbering for D56 to the CDR2-IMGT; and G107, S108, G109,
3D structures. Indeed, in IMGT, any residue and Y114 to the CDR3-IMGT). These results
(amino acid) that belongs to an antibody domain emphasize the importance of using the IMGT
(and more generally, any amino acid that unique numbering for standardized antibody
belongs to a V or C domain of an IgSF protein, analysis and confirm that the CDR-IMGT
or to a G domain of a MhSF protein [32]) is structural delimitations correspond to the V-
characterized by its position in the domain. An domain interactions with the antigen.
R@P is defined by the IMGT position number- The Ag epitope of 3ixt (Fig. 3C) comprises
ing, the residue name, the IMGT domain AA of the Fusion glycoprotein F1 (3ixt_P).
description, and the IMGT chain ID (e.g., 35 Eleven amino acids form the Ag epitope. Each
ALA (A) VH 3ixt_H). Structural information amino acid that belongs to the epitope is
and contacts for a given R@P are provided in defined by its position in the chain in the 3D
the IMGT Residue@Position card that includes structure (if the AA is part of a V, C, or G
general information (PDB file numbering, domain, the position is given according to the
IMGT file numbering, residue full name and IMGT unique numbering (32).
formula), and structural information IMGT Clicking on a residue in Paratope details
LocalStructure@Position (secondary struc- or in Epitope details (Fig. 3C) provides the
CHAPTER 21 Antibody Informatics: IMGT 375

FIGURE 3 IMGT/3Dstructure-DB Domain pair contacts and IMGT paratope and epitope details. A, IMGT/
3Dstructure-DB Domain pair contacts between the VH of motavizumab (3ixt_H) and the Fusion
glycoprotein F1 (ligand) (3ixt_P). B, IMGT/3Dstructure-DB Domain pair contacts between the V-KAPPA
of motavizumab (3ixt_L) and the Fusion glycoprotein F1 (ligand) (3ixt_P). Polar, Hydrogen bonds, and
Nonpolar were selected before display, in Atom contact types. Amino acids belonging to the CDR1-
IMGT, CDR2-IMGT, and CDR3-IMGT are colored online according to the IMGT color menu (red, orange,
and purple, respectively, for VH; blue, green, and green-blue, respectively, for V-KAPPA). In this 3D
structure, all but one amino acid that contact the antigen belong to the CDR-IMGT. Clicking on R@P gives
access to the IMGT Residue@Position cards (26, 48). C, IMGT paratope and epitope details of the IG/Ag
complex 3ixt is shown. Each AA that belongs to the IG paratope is characterized by its position in the V
domains according to the IMGT unique numbering (29, 30, 32). Thus, A (35V1_A) means that the alanine
(A) is at position 35 of the V domain 1 of 3ixt_A (VH). In the same way, G(107V1_B) means that the
glycine (G) is at position 107 of the V domain 1 of 3ixt_B (V-KAPPA). Each AA that belongs to the
antigenic determinant (epitope) is characterized by its position (here, position in the chain, in the 3D
structure). For example, S (3_C) means that the serine (S) is at position 3 of the Fusion glycoprotein F1
ligand (3ixt_C), whereas SN (23-24_C) means that the serine (S), asparagine (N) are at positions 23, 24.
The IMGT paratope and epitope analysis of the IG/Ag 3D structure (3ixt) is from IMGT/3Dstructure-DB
(http://www.imgt.org). doi:10.1128/microbiolspec.AID-0001-2012.f3
376 LEFRANC

R@P card for each AA that belongs to the sequences of the paired heavy and light chains
paratope or epitope, respectively. of antibodies from WHO-INN (24, 25) and
those from Kabat (annotated by IMGT and for
which there are no available nucleotide se-
IMGT/StructuralQuery and IMGT/
quences, the others already being in IMGT/
DomainSuperimpose
LIGM-DB). IMGT/2Dstructure-DB entries
IMGT/DomainSuperimpose (3) (IMGT also include the INN amino acid sequences of
Home page, http://www.imgt.org) allows one FPIA (suffix -cept) and those of CPCA with a
to superimpose the 3D structures of two Fc (for increased half-life) (prefix ef-).
domains from IMGT/3Dstructure-DB, and Queries can be made on an individual entry,
demonstrate that the same IMGT numbering using the Entry ID (e.g., for an INN entry, the
is found at the same positions of the frame- 4-number code, e.g., 8693) or the Molecule
work of the heavy and light chains and for the name (e.g., motavizumab). Search can be made
lower part of the CDR-IMGT loops of the two on Entry type (e.g., INN), and on criteria
domains. IMGT/StructuralQuery (3) allows detailed in the IMGT/2Dstructure-DB Query
the retrieval the IMGT/3Dstructure-DB page, http://www.imgt.org). In the results
entries containing a V, C, or G domain, based page, clicking on an IMGT entry ID gives
on specific structural characteristics of the access to the IMGT/2Dstructure-DB card
intramolecular interactions: phi and psi angles, (42). The IMGT/2Dstructure-DB card
accessible surface area, type of atom contacts, provides standardized IMGT information on
distance in angstroms between amino acids, chains and domains and IMGT Colliers de
R@P pair contacts, and, for the V domain, Perles on one or two layers, identical to that of
CDR-IMGT length or pattern. an IMGT/3Dstructure-DB card; however, the
information on experimental structural data
(hydrogen bonds in IMGT Collier de Perles on
IMGT/2Dstructure-DB AND
two layers, Contact analysis) is only available
IMGT/mAb-DB
in the corresponding IMGT/3Dstructure-DB
In a further effort to bridge the gap between card, if the antibodies have been crystallized
sequences and 3D structures, a new extension (26, 48). For therapeutic antibodies, queries
of IMGT/3Dstructure-DB, designated as can be made from the IMGT/mAb-DB user-
IMGT/2Dstructure-DB, was created to de- friendly interface (http://www.imgt.org) on
scribe and analyze amino acid sequences of search criteria, such as characteristics (conju-
paired chains of heavy and light chains of gated, radiolabeled), IG classes or subclasses
antibodies for which no 3D structures are for complete IG or format (Fab, scFv, etc.) for
available. These amino acid sequences are IG fragments, specificity (target name and
analyzed and managed with the IMGT criteria species), clinical indication, development sta-
of standardized gene and allele nomenclature tus, etc.
(classification), standardized labels (descrip-
tion), and IMGT unique numbering and IMGT
Colliers de Perles (numerotation) (10). IMGT/ TOWARD TARGETED AND CUSTOMIZED
2Dstructure-DB uses the IMGT/3Dstructure- THERAPEUTIC ANTIBODIES
DB informatics frame and interface that allows
one to analyze, manage, and query antibodies IMGT provides antibody informatics with a
as polymeric receptors made of several chains, standardized system for sequences and struc-
in contrast to the IMGT/LIGM-DB sequence tures (6, 7), from amino acid characteristics
database that analyzes and manages IG se- at given positions (49), to IG gene and allele
quences, individually. The current IMGT/ identification, and to 3D structures analysis
2Dstructure-DB entries include amino acid (antibody/antigen contact analysis, paratope/
CHAPTER 21 Antibody Informatics: IMGT 377

epitope description). Since its creation in 1989, (IMIA). IMGT was funded in part by the
and owing to a strong expertise and back- BIOMED1 (BIOCT930038), Biotechnology
ground in immunogenetics, IMGT has defined BIOTECH2 (BIO4CT960037), 5th PCRDT
IG alleles and developed databases and tools Quality of Life and Management of Living
and web resources for IG polymorphism, Resources (QLG2-2000-01287), and 6th
as demonstrated by IMGT/GENE-DB (18), PCRDT Information Science and Technology
IMGT Alignments of alleles and the corre- (ImmunoGrid, FP6 IST-028069) programs of
spondences between IGHG and IGKC genes the European Union (EU), and by the Agence
and the C-domain Gm and Km allotypes (50). Nationale de la Recherche ANR (BIOSYS06_
The extension of the IMGT unique numbering 135457, FLAVORES). IMGT is currently
to the IgSF and to the MhSF proteins other supported by the Centre National de la Re-
than IG or TR has opened new perspectives cherche Scientifique (CNRS), the Ministre de
for the standardized description of the poly- lEnseignement Suprieur et de la Recherche
morphism of the antigens (epitopes belonging (MESR), the Universit Montpellier 2, the GIS
to V, C, or G domains) and of the Fc recep- IBiSA, the Rgion Languedoc-Roussillon
tors (FCGR of the IgSF, FCGRT of the MhSF) (Grand Plateau Technique pour la Recher-
and for the characterization of their inter- che [GPTR], GEPETOS), and the Labex
actions (antibody/antigen, FcR/antibody). MAbImprove (ANR-10-LABX-53). This work
Given the importance of these interactions was granted access to the HPC resources of
in the antibody specificity and affinity on CINES under the allocations 2010-2014-036029
the one hand and in the antibody pharmaco- made by GENCI (Grand Equipement National
kinetics/pharmacodynamics and half-life on de Calcul Intensif).
the other hand, the IMGT integrated and Conflicts of interest: I declares no conflicts.
standardized approach provides the genetic
knowledge for allowing antibody informatics
CITATION
to answer the needs of targeted and custom-
ized therapy in the context of personalized Lefranc M-P. 2014. Antibody informatics:
medicine. IMGT, the International ImMunoGeneTics
Information System. Microbiol Spectrum
2(2):AID-0001-2012.
AVAILABILITY AND CITATION
REFERENCES
Authors who use IMGT databases and tools
are encouraged to cite this article and to quote 1. Lefranc M-P, Lefranc G. 2001. The Immunoglob-
the IMGT Home page (http://www.imgt.org). ulin FactsBook. Academic Press, London, United
Kingdom.
Online access to IMGT databases and tools is 2. Lefranc M-P, Lefranc G. 2001. The T Cell
freely available for academics and under Receptor FactsBook. Academic Press, London,
licenses and contracts for companies. United Kingdom.
3. Lefranc M-P, Giudicelli V, Ginestoux C, Jabado-
Michaloud J, Folch G, Bellahcene F, Wu Y,
ACKNOWLEDGMENTS Gemrot E, Brochet X, Lane J, Regnier L,
Ehrenmann F, Lefranc G, Duroux P. 2009.
I thank Grard Lefranc for helpful discussions, IMGT, the international ImMunoGeneTics in-
Sophia Kossida and Souphatta Sasorith for formation system. Nucl Acids Res 37:D1006
reading the manuscript, Denis Moreno for the D1012.
4. Giudicelli V, Lefranc M-P. 1999. Ontology for
figures, and the IMGT team for its expertise and
immunogenetics: IMGT-ONTOLOGY. Bioinfor-
constant motivation. IMGT is a registered matics 15:10471054.
trademark of CNRS. IMGT is a member of the 5. Lefranc M-P, Giudicelli V, Ginestoux C, Bosc N,
International Medical Informatics Association Folch G, Guiraudou D, Jabado-Michaloud J,
378 LEFRANC

Magris S, Scaviner D, Thouvenin V, Combres K, receptors report August 2007, 13th International
Girod D, Jeanjean S, Protat C, Yousfi Monod M, Congress of Immunology, Rio de Janeiro, Brazil.
Duprat E, Kaas Q, Pommi C, Chaume D, Dev Comp Immunol 32:461463.
Lefranc G. 2004. IMGT-ONTOLOGY for immu- 18. Giudicelli V, Chaume D, Lefranc M-P. 2005.
nogenetics and immunoinformatics. In Silico Biol IMGT/GENE-DB: a comprehensive database for
4:1729. human and mouse immunoglobulin and T cell
6. Lefranc M-P, Clment O, Kaas Q, Duprat E, receptor genes. Nucl Acids Res 33:D256D261.
Chastellan P, Coelho I, Combres K, Ginestoux C, 19. Letovsky SI, Cottingham RW, Porter CJ, Li PW.
Giudicelli V, Chaume D, Lefranc G. 2005. IMGT- 1998. GDB: the Human Genome Database. Nucl
Choreography for Immunogenetics and Immuno- Acids Res 26:9499.
informatics. In SilicoBiol 5:4560. 20. Maglott DR, Katz KS, Sicotte H, Pruitt KD.
7. Lefranc M-P, Giudicelli V, Regnier L, Duroux P. 2000. NCBIs LocusLink and RefSeq. Nucl Acids
2008. IMGT, a system and an ontology that Res 28:126128.
bridge biological and computational spheres in 21. Maglott D, Ostell J, Pruitt KD, Tatusova T.
bioinformatics. Brief Bioinform 9:263275. 2007. Entrez Gene: gene-centered information at
8. Duroux P, Kaas Q, Brochet X, Lane J, Ginestoux NCBI. Nucl Acids Res 35:D26D31.
C, Lefranc M-P, Giudicelli V. 2008. IMGT- 22. Stabenau A, McVicker G, Melsopp C, Proctor G,
Kaleidoscope, the formal IMGT-ONTOLOGY Clamp M, Birney E. 2004. The Ensembl core
paradigm. Biochimie 90:570583. software libraries. Genome Res 14:929933.
9. Giudicelli V, Lefranc M-P. 2012. IMGT-ONTOL- 23. Wilming LG, Gilbert JG, Howe K, Trevanion S,
OGY 2012. Front Genet 3:79. doi:10.3389/fgene. Hubbard T, Harrow JL. 2008. The vertebrate
2012.00079. genome annotation (Vega) database. Nucl Acids
10. Lefranc M-P. 2013. IMGT-ONTOLOGY, p 964 Res 36:D753D760.
972. In Dubitzky W, Wolkenhauer O, Cho K, 24. Lefranc M-P. 2011. Antibody nomenclature: from
Yokota H (ed), Encyclopedia of Systems Biology, IMGT-ONTOLOGY to INN definition. mAbs 3:12.
doi:10.1007/978-1-4419-9863-7. Springer Science 25. World Health Organization. 2011. General poli-
+Business Media, LLC012, Rueil-Malmaison, cies for monoclonal antibodies, p 810. In Inter-
France. national Nonproprietary Names (INN) for Biological
11. Lefranc M-P. 2011. From IMGT-ONTOLOGY and Biotechnological Substances, INN Working
CLASSIFICATION Axiom to IMGT standardized Document 05.179, Update 2011. http://www.who.
gene and allele nomenclature: for immuno- int/medicines/services/inn/en.
globulins (IG) and T cell receptors (TR). Cold 26. Ehrenmann F, Kaas Q, Lefranc M-P. 2010. IMGT/
Spring Harb Protoc 6:627632. pii: pdb.ip84. 3Dstructure-DB and IMGT/DomainGapAlign: a
doi:10.1101/pdb.ip84. database and a tool for immunoglobulins or
12. Lefranc M-P. 2001. Nomenclature of the human antibodies, T cell receptors, MHC, IgSF and
immunoglobulin genes, p A.1P.1A.1P.37. In MhcSF. Nucl Acids Res 38:D301D307.
Coligan JE, Bierer BE, Margulies DE, Shevach 27. Poiron C, Wu Y, Ginestoux C, Ehrenmann F,
EM, Strober W (ed), Current Protocols in Immu- Duroux P, Lefranc M-P. 2010. IMGT/mAb-DB:
nology. John Wiley and Sons, Hoboken, NJ. the IMGT database for therapeutic monoclonal
13. Lefranc M-P. 2001. Nomenclature of the human antibodies, poster 101. 11mes Journes Ouvertes
T cell receptor genes, p A.1O.1A.1O.23. In Coligan de Biologie, Informatique et Mathmatiques
JE, Bierer BE, Margulies DE, Shevach EM, (JOBIM), Montpellier, France, 7 to 9 September
Strober W (ed), Current Protocols in Immunology. 2010.
John Wiley and Sons, Hoboken, NJ. 28. Lefranc M-P. 1997. Unique database numbering
14. Wain HM, Bruford EA, Lovering RC, Lush MJ, system for immunogenetic analysis. Immunol
Wright MW, Povey S. 2002. Guidelines for Today 18:509.
human gene nomenclature. Genomics 79:464 29. Lefranc M-P. 1999. The IMGT unique numbering
470. for Immunoglobulins, T cell receptors and Ig-like
15. Bruford EA, Lush MJ, Wright MW, Sneddon domains. The Immunologist 7:132136.
TP, Povey S, Birney E. 2008. The HGNC Data- 30. Lefranc M-P, Pommi C, Ruiz M, Giudicelli V,
base in 2008: a resource for the human genome. Foulquier E, Truong L, Thouvenin-Contet V,
Nucl Acids Res 36:D445D448. Lefranc G. 2003. IMGT unique numbering for
16. Lefranc M-P. 2007. WHO-IUIS Nomenclature immunoglobulin and T cell receptor variable
Subcommittee for immunoglobulins and T cell domains and Ig superfamily V-like domains. Dev
receptors report. Immunogenetics 59:899902. Comp Immunol 27:5577.
17. Lefranc M-P. 2008. WHO-IUIS Nomenclature 31. Lefranc M-P, Pommi C, Kaas Q, Duprat E, Bosc
Subcommittee for immunoglobulins and T cell N, Guiraudou D, Jean C, Ruiz M, Da Piedade I,
CHAPTER 21 Antibody Informatics: IMGT 379

Rouard M, Foulquier E, Thouvenin V, Lefranc 42. Lefranc MP, Ehrenmann F, Ginestoux C,


G. 2005. IMGT unique numbering for immuno- Duroux P, Giudicelli V. 2012. Use of IMGT
globulin and T cell receptor constant domains and databases and tools for antibody engineering and
Ig superfamily C-like domains. Dev Comp humanization. In Chames P (ed), Antibody Engi-
Immunol 29:185203. neering. Humana Press, Springer, New York, NY.
32. Lefranc M-P. 2011. IMGT Unique Numbering for Meth Mol Biol 907:337.
the Variable (V), Constant (C), and Groove (G) 43. Alamyar E, Giudicelli V, Shuo L, Duroux P,
Domains of IG, TR, MH, IgSF, and MhSF. Cold Lefranc M-P. 2012. IMGT/HighV-QUEST: the
Spring Harb Protoc 6:633642. pii: pdb.ip85. IMGT web portal for immunoglobulin (IG) or
doi:10.1101/pdb.ip85. antibody and T cell receptor (TR) analysis from
33. Lefranc M-P, Duprat E, Kaas Q, Tranne M, NGS high throughput and deep sequencing.
Thiriot A, Lefranc G. 2005. IMGT unique Immunome Res 8:26.
numbering for MHC groove G-DOMAIN and 44. Giudicelli V, Brochet X, Lefranc M-P. 2011.
MHC superfamily (MhcSF) G-LIKE-DOMAIN. IMGT/V-QUEST: IMGT Standardized Analysis
Dev Comp Immunol 29:917938. of the Immunoglobulin (IG) and T Cell Receptor
34. Ruiz M, Lefranc M-P. 2002. IMGT gene identi- (TR) Nucleotide Sequences. Cold Spring Harb
fication and Colliers de Perles of human Protoc 6:695715. pii: pdb.prot5633. doi:10.1101/
immunoglobulins with known 3D structures. pdb.prot5633.
Immunogenetics 53:857883. 45. Giudicelli V, Lefranc M-P. 2011. IMGT/
35. Kaas Q, Lefranc M-P. 2007. IMGT Colliers de JunctionAnalysis: IMGT Standardized Analysis
Perles: standardized sequence-structure repre- of the V-J and V-D-J Junctions of the Rearranged
sentations of the IgSF and MhcSF superfamily Immunoglobulins (IG) and T Cell Receptors (TR).
domains. Curr Bioinformatics 2:2130. Cold Spring Harb Protoc 6:716725. pii: pdb.
36. Kaas Q, Ehrenmann F, Lefranc M-P. 2007. IG, prot5634. doi:10.1101/pdb.prot5634.
TR and IgSf, MHC and MhcSF: what do we learn 46. Alamyar E, Duroux P, Lefranc M-P, Giudicelli
from the IMGT Colliers de Perles? Brief Funct V. 2012. IMGT tools for the nucleotide analysis of
Genomic Proteomic 6:253264. immunoglobulin (IG) and T cell receptor (TR) V-
37. Lefranc M-P. 2011. IMGT Collier de Perles for the (D)-J repertoires, polymorphisms, and IG muta-
Variable (V), Constant (C), and Groove (G) tions: IMGT/V-QUEST and IMGT/HighV-
Domains of IG, TR, MH, IgSF, and MhSF. Cold QUEST for NGS. In Christiansen F, Tait B (ed),
Spring Harb Protoc 6:643651. pii: pdb.ip86. Immunogenetics. Humana Press, Springer, New
doi:10.1101/pdb.ip86. York, NY. Meth Mol Biol 882:569604.
38. Ehrenmann F, Giudicelli V, Duroux P, Lefranc 47. Ehrenmann F, Lefranc M-P. 2011. IMGT/
M-P. 2011. IMGT/Collier-de-Perles: IMGT Stan- DomainGapAlign: IMGT Standardized Analysis
dardized Representation of Domains (IG, TR, and of Amino Acid Sequences of Variable, Constant,
IgSF Variable and Constant Domains, MH and and Groove Domains (IG, TR, MH, IgSF, MhSF).
MhSF Groove Domains). Cold Spring Harb Protoc Cold Spring Harb Protoc 6:737749. pii: pdb.
6:726736. pii: pdb.prot5635. doi:10.1101/pdb. prot5636. doi:10.1101/pdb.prot5636.
prot5635. 48. Ehrenmann F, Lefranc M-P. 2011. IMGT/
39. Lefranc M-P. 2004. IMGT, the international 3Dstructure-DB: Querying the IMGT Database
ImMunoGenetics information system. In Lo for 3D Structures in Immunology and Immuno-
BKC (ed), Antibody Engineering: Methods and informatics (IG or Antibodies, TR, MH, RPI, and
Protocols, 2nd edition. Humana Press, Totowa, FPIA). Cold Spring Harb Protoc 6:750761. pii:
NJ. Meth Mol Biol 248:2749. pdb.prot5637. doi:10.1101/pdb.prot5637.
40. Lefranc M-P. 2009. Antibody databases and tools: 49. Pommi C, Levadoux S, Sabatier R, Lefranc M-
The IMGT experience, p 91114. In An Z (ed), P. 2004. IMGT standardized criteria for statistical
Therapeutic Monoclonal Antibodies: from Bench to analysis of immunoglobulin V-REGION amino
Clinic. John Wiley and Sons, Hoboken, NJ. acid properties. J Mol Recognit 17:1732.
41. Ehrenmann F, Duroux P, Giudicelli V, Lefranc 50. Lefranc M-P, Lefranc G. 2012. Human Gm, Km
M-P. 2010. Standardized sequence and struc- and Am allotypes and their molecular character-
ture analysis of antibody using IMGT, p 1131. ization: a remarkable demonstration of poly-
In Kontermann R, Dbel S (ed), Antibody Engi- morphism. In Christiansen F, Tait B (ed),
neering, vol 2. Springer-Verlag, Heidelberg, Immunogenetics. Humana Press, Springer, New
Germany. York, NY. Meth Mol Biol 882:635680.
Probing Antibody-Antigen
Interactions

GUOCHENG YANG,1 STEFANIE N. VELGOS,2 SHANTA P. BODDAPATI,3 and


MICHAEL R. SIERKS1
22
PROBING ANTIBODY-ANTIGEN INTERACTIONS

Antibodies bind antigens via noncovalent bonds, such as hydrogen bonds, ionic,
hydrophobic, and Van der Waals forces, and their interactions depend strongly
on the distance between two interacting molecules. While each individual bond
is weak, the collective noncovalent bonds between the antibody and antigen can
be strong when all the interacting molecules work together synergistically.
Because there are a very large number of these interactions and the antigen and
antibody are large, flexible, dynamic molecules, binding between an antibody
and antigen is a very complex process. The binding interactions may also be
time dependent, because formation of an antibody-antigen complex may
involve a sequential series of interactions which induce conformational changes
that generate some bonds while breaking others. Because of the dynamic and
transient state of antibody-antigen interactions, measurements of the anti-
body-antigen interactions can be quite complicated and inconsistent, and the
results may vary depending on the sample treatment conditions and technique
utilized (1).

1
Department of Chemical Engineering, Arizona State University, Tempe, AZ 85287-6006; 2Mayo Clinic Arizona, Phoenix,
AZ 85054; 3Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0010-2013

381
382 YANG ET AL.

Classical immunoassays such as radioim- of the analytes is unstable or cannot be im-


munoassay (RIA) and enzyme-linked immu- mobilized.
nosorbent assay (ELISA) are widely used to
determine the affinity and specificity of anti-
SPR: a Label-Free Detection System
bodies to their target antigens. These assays
typically involve the immobilization of the SPR is an optical technique that offers real-
antigen or antibody onto a solid support, time, label-free detection of binding inter-
followed by the introduction of labeled actions between the immobilized biomole-
analyte at varying concentrations and the cule and the free-flowing analyte. SPR
measurement of bound analyte to determine measures changes in mass at the sensor
the binding constant (KD) of the antibody. surface as the analyte interacts with the
While these methods are widely utilized, immobilized biomolecule, and this increase
there are some limitations in their application. in mass is proportional to the increase in
For example, RIA and ELISA generally re- refractive index (discussed later). A typical
quire large amounts of sample, chemical SPR instrument comprises three main com-
modification to the antigen or antibody for ponents: (i) a microfluidic cartridge, (ii) a
immobilization, and the need of a label. sensor chip, and (iii) an optical detection
Consequently, modern immunochemical unit (Fig. 1A). The microfluidic assembly
techniques, which include label-free detec- allows the analyte to pass over the sensor
tion, a single-molecule detection level, and a surface in a continuous and controlled flow,
low sample volume system, have been devel- while maintaining a constant analyte con-
oped to overcome these limitations and to centration. The sensor chip is usually made
complement the classical methods to study of a glass support with a thin gold-sensing
antibody-antigen interactions. Here, we will layer (2). The gold-sensing surface of the
review three conceptually different tech- chip is in direct contact with a flow cell (the
niques that can be used to study and quantify sample side), while the glass support is
bulk and single-molecule-level antibody-anti- placed directly adjacent to a glass prism on
gen interactions: (i) surface plasmon reso- the detector side (Fig. 1B). A light source,
nance (SPR), a label-free method to measure typically a light-emitting diode, emits a
both on and off rates of antibody-antigen wedge of polarized light (3) onto the prism,
interactions; (ii) fluorescence-activated cell which has the same refractive index as the
sorting (FACS) that enables high-throughput glass substrate. The angle of incident from
screening of surface-bound proteins; and (iii) the light-emitting diode source is increased
atomic force microscopy (AFM) that enables a until the prism reflects light at or beyond
molecular-level detection of individual anti- the critical angle of the sensor surface such
body-antigen binding. These techniques differ that total internal reflection (TIR) occurs
from traditional immunoassays in that they do (Fig. 1B).
not require high sample volumes or concen- TIR occurs when a light beam is propagat-
trations, SPR and AFM do not require addi- ed through two nonabsorbing media of differ-
tional labeling, SPR provides real-time in situ ent refractive indices (like glass-buffer or
detection of antibody-antigen interactions, glass-air) (4). This reflected energy from the
and AFM can provide detailed structural light beam excites the electrons of the gold
information of the target antigen. These atoms in the outer orbitals, thus causing the
techniques expand the capabilities of classical excited electrons to become delocalized and to
immunoassays and are particularly useful form plasmons (4). The delocalized electrons
when the quantity of antibody or antigen is (plasmons) resonate on the gold surface and
limited, or when the purification process is emit evanescent waves, which propagate from
tedious and difficult, or when the structure the glass sensor chip (high refractive index) to
CHAPTER 22 Probing Antibody-Antigen Interactions 383

FIGURE 1 Schematic illustration of a BIAcore SPR instrument. (A) The BIAcore system contains three main
elements crucial to its operation: the integrated microuidic cartridge (IFC) (light gray L-shaped block),
sensor chip, and optical detection unit. (B) A simplistic overview of how SPR works. (C) The
transformation of the shift in incident angles to the response signal in an SPR sensogram.
doi:10.1128/microbiolspec.AID-0010-2013.f1

the medium (low refractive index). The the target and analyte, to determine the
resonance energy transfer between the eva- kinetics of binding interactions (on and off
nescent wave and the surface plasmons on the rates), and to determine the concentrations of
metal surface causes a decrease in the inci- the amount of analyte in a sample (Fig. 2).
dent light intensity (3). TIR only occurs at a Because the detection sensitivity of SPR
specific angle of the incident light reflection mainly depends on the distance of the analyte
(5), and the SPR-reflected light angle depends from the sensor surface, although some
on the refractive index of the medium close to electrostatic attraction or conformational
the gold film surface (3). As mass accumulates changes may induce changes in SPR angles
at the sensor chip surface owing to biomolec- (6), SPR can detect virtually any interacting
ular interactions, it causes a change in molecules regardless of their nature.
localized solute concentration, which corre- Several SPR experimental setups can be
sponds to a change in the refractive index of used to obtain data, including direct binding,
the medium. This translates into a change in surface competition, or inhibition-in-solution
the SPR angle (5) and the SPR angle is often assays (7). For immobilization of antigen or
reported as resonance units (RUs) (Fig. 1C) antibody, a variety of different surface chem-
(3). By monitoring the SPR signal, it is istries (e.g., dextran, carboxylation, and strep-
possible to quantify the interaction between tavidin coating) are available so that binding
384 YANG ET AL.

FIGURE 2 An SPR sensogram depicting the different stages of a binding event. After the target has been
immobilized, a baseline RU is established by using only running buffer in the ow system. Upon injection of the
analyte, the RU on the sensogram gradually increases, indicating that the system is in the association phase
where the analyte binds to the target. It is important to note that some bound molecules have already begun to
dissociate during analyte injection. The RU reaches saturation at steady state, where the associating and
dissociating molecules are in equilibrium. Once the analyte injection is completed and is replaced by running
buffer, the system is in the pure dissociation phase, which is marked by a decrease in RU. The sensor chip
regeneration is performed, and baseline RU should be restored. doi:10.1128/microbiolspec.AID-0010-2013.f2

artifacts and high nonspecific binding can be addition to antibody-antigen interactions, SPR
avoided (8). has also been used to measure proteinpeptide
SPR has important benefits over conven- and protein-DNA interactions, cellular liga-
tional immunofluorescence or ELISA-type tion, and DNA hybridization (9, 10).
assays for studying interactions between
immobilized biomolecules and a solution-
FACS Offers High-Throughput Screening
phase analyte. SPR measurements are based
of Antibodies
on refractive index changes and do not require
the need of a label or the need for a multistep FACS is an application of flow cytometry that
protocol (9). Also, since SPR measurements uses fluorescently labeled reagents to detect
are performed in real time instead of by using antigens expressed on cells or other particles.
equilibrium endpoints, it provides useful ther- This technique can be effectively used to
modynamic and kinetic information of the determine antibody-antigen interactions. One
system, including binding enthalpies and en- major advantage of using FACS for studying
tropies, and the kinetic on and off rates. In antibody-antigen interactions is that the target
CHAPTER 22 Probing Antibody-Antigen Interactions 385

antigen does not need to be modified or proportional (fixed constant ratio) to the
immobilized. In FACS, cells are first labeled number of binding events, since both rates of
directly or indirectly with fluorophore-conju- binding and dissociation are equal at equilibri-
gated antibodies. The labeled cell-antibody um (Eq. 1). With the use of single-site first-
mixture is suspended in a stream of fluid, and order binding kinetics, and assuming that the
the cells can then be sorted and analyzed concentration of free scFv is much larger than
individually. Laser light is directed onto the the number of binding sites (12), the KD and
stream of cells and the scattering charac- maximum fluorescence values (Fmax) values
teristics of the cells are used to analyze cell can be obtained by using Eq. 1 and Eq. 2,
size, granularity, and expression of the antigen respectively. Equation 2 is generally referred to
of interest on the cell surface (11). Thus, as a Lineweaver-Burke plot. A similar ap-
multiple antibodies can be used to simulta- proach was also used to quantify the KD values
neously detect the expression of several anti- of botulinum toxin specific antibodies using
gens of interest on the cell surface (Fig. 3). Cells flow cytometry (13). FACS-based selection
with desired characteristics can be collected techniques were also used to select for scFvs
for further growth and characterization. against several proteins and phosphopeptides
Flow cytometry was first used to determine by using a nonimmune yeast surface display
the equilibrium binding constant KD for a library where the scFvs were displayed on the
recombinant single-chain antibody fragment yeast cell surface by genetically coupling the
(scFv) binding to the CD34 antigen (12). In this scFv gene to a yeast agglutinin receptor (14). KD
case, yeast cells expressing the scFv were values of the scFvs calculated using FACS
conjugated with a labeled antibody and the were similar to values obtained by using SPR
antigen concentration was varied. The antigen- (15).
antibody binding can be quantified using flow
cytometry by measuring the mean fluorescence
intensity (MFI) for the antibody and antigen koff AbAg
binding over a range of concentrations. The KD KD 1
kon AbAg
can be calculated assuming MFI is directly

FIGURE 3 Schematic illustration of a FACS system. (A) A ow cytometer showing the light path, sensors,
optics, and lters used (gure adapted from Applied Microbiology and Biotechnology [11] with permission
of with permission of Beckman-Coulter). (B) A simplied illustration of the principles of ow cytometry
with the use of FACS. FL, uorescent light. doi:10.1128/microbiolspec.AID-0010-2013.f3
386 YANG ET AL.

where KD = equilibrium binding con- protein, it can be used at the stage of selection
stant, and screening of antibodies. In addition,
kon = association constant, screening methods that use FACS analysis
koff = dissociation constant, will be able to differentiate positive clones
[Ab] = antibody concentration, based on affinity. Unlike other selection meth-
[Ag] = antigen concentration, and ods that collect all positive binders for further
[AbAg] = antibody-antigen complex con- rounds of amplification, high and low binders
centration can be separately sorted for further amplifica-
tion.
1 1 KD 1
2
FFb Fmax Fmax scFv Single-Molecule Antigen-Antibody
Detection Using AFM
where Fb = background fluorescence, The previous techniques describe methods
Fmax = maximum fluorescence, and that can be used to analyze bulk antigen-
[scFv] = concentration of scFv antibody interactions. However, sometimes it
FACS-based analysis can also be used to is beneficial to study single-molecule interac-
screen and select for antibodies from second- tions. Antibody-antigen binding interactions
generation libraries containing mutations of a are complex and dynamic processes, and
parent antibody (16, 17, 18). FACS selection was proteins may have a multitude of variable
used to isolate higher-affinity variants from a conformations complicating or preventing
library of clones generated from a parent anti- bulk measurements. AFM is a powerful tech-
fluorescein 4-4-20 antibody (19). The dissoci- nique that can be used to measure single-
ation of antigen from scFv follows a first-order molecule antibody-antigen interactions.
kinetic model since the dissociation rate (koff) Three major techniques have been used to
depends on the amount of antigen bound to the explore single molecular interactions: optical
surface-displayed scFv on the yeast (Eq. 3). A tweezers, magnetic tweezers, and AFM.
plot of the exponential decay of MFI with time Among them, optical tweezers and magnetic
enables calculation of the amount of bound tweezers use either a laser optical trap or a
antigen providing the system is initially satu- magnetic force field to move a particle with
rated with excess antigen (Eq. 4). Antibody molecules linked on it, while AFM uses a tiny
clones can be selected from the library based stylus of a few nanometers in radius to directly
on koff values, reducing the number of clones probe and move the single molecule in three
that need to be tested for further screening or dimensions. An excellent review describing
characterization. the different principles and instrumentation
setups and discussing the resolution and other
dF limitations is available (20). Although optical
koff F 3 tweezer and magnetic tweezer techniques are
dt
considered to be noninvasive, they are not
particularly well suited for measuring protein
Ft F0 ek off t 4 interactions because of their low pico-Newton
measurement range (0.1 to 100 pN). AFM,
however, can be effectively used to study
where Ft = MFI at time, t, and Fo = initial interactions between molecules because it
MFI. has a relatively large measurement range, is
FACS-based analysis of antigen-antibody simple to operate, and has a unique ability to
binding is a convenient and reliable method. simultaneously image the sample and measure
Because it does not need purified form of the force interactions.
CHAPTER 22 Probing Antibody-Antigen Interactions 387

FIGURE 4 Schematic illustration of an AFM. (A) A typical AFM setup comprises an AFM cantilever, a laser
source, a photodetector, and a piezoelectric scanner. (B) AFM topographical imaging is acquired when
the tip raster scans the sample surface in the X-Y direction, while the tip deection is recorded in the Z
direction. (C) AFM force-distance curve. The force plot is generated when the tip (i) approaches the
surface (red dash line), (ii) contacts the surface and moves with the surface up to a preset point. In the
retracting phase (green solid line), the tip retracts from the surface and experiences (iii) one or (iv) more
bond-breaking events before completely dissociating from the surface and (v) returning to its original
point. doi:10.1128/microbiolspec.AID-0010-2013.f4

AFM was derived from scanning probe reflected as a lateral or vertical shift of the laser
microscopy, which was invented by Binnig et spot on the photodetector. This shift is used to
al. in 1984 (21). AFM typically contains four adjust the X-Y-Z extension of the scanner, to
main parts: (i) a tip attached to a flexible maintain the distance between the sample and
cantilever, (ii) a laser signal and a photodetector the scanning tip at the prescribed set point
sensing the movement of the tip, (iii) a piezo- value, which corresponds to the specific inter-
electric scanner controlling the X-Y-Z position action force between the tip and sample. The X-
of the sample, and (iv) a feedback loop control- Y-Z position of the sample generated from this
ling the movement of these three parts (Fig. 4). process is recorded as the topography of the
While the tip raster scans the sample surface, sample, also known as the height image. This
the laser beam is projected off the back of the primary imaging mode of AFM is also known as
cantilever onto the photodetector. The tip contact-mode AFM, in which the tip is in
deflection caused by tip-sample interaction is constant contact with the sample. To reduce
388 YANG ET AL.

interactions between the tip and sample, flexible the tip is, the more difficult it is to
Hansma et al. invented a new way of oscillating obtain good images. Contact mode imaging
a tip at a high frequency over the sample within and noncontact mode imaging usually use
a short distance, which is known as intermittent cantilevers of different spring constants, and
contact or tapping mode AFM (22). not all of them are suitable for force interaction
AFM is able to produce high-resolution studies because of sensitivity limits.
images of sample topography in aqueous/ To prevent nonspecific binding interactions,
liquid environments with the use of minimal the protein of interest can be attached onto the
sample preparation and without the need to cantilever tip. Two common approaches are
stain the sample. Images with nanometer used, either esterification or silanization. In
resolution laterally and Angstrom resolution general, 3-aminophenyl-trimethoxysilane is
vertically can be readily obtained (23, 24, 25). used for silanization and ethanolamine is used
In addition to generating high-resolution for esterification (26). Both of these approaches
images, AFM is also particularly useful for add functional amine groups onto the end the
measuring interaction forces between mole- tip. The amine groups are sequentially
cules. Since the tip on the cantilever is free to connected with glutaraldehyde, which can
move above the sample surface, it can be then be used to connect the target protein
modeled as a spring with a spring constant of onto the tip. If needed, an extended polyethyl-
k. According to Hookes law, the force exerted ene glycol linker can be added to allow more
between the tip and sample can be calculated flexibility for the attached protein to minimize
from the deflection of the tip (z) multiplied steric constraints.
by the spring constant k (Eq. 5). For any sudden AFM was initially used to study the binding
change in the force interaction between the tip interaction between streptavidin and biotin,
and surface due to the rupture of a bond or one of the strongest protein/protein bonds
unfolding of a molecule, a sharp change in the known in nature. With the use of AFM, the
force curve will be observed. Such special force interaction between streptavidin and
force curves will have multiple peaks or a biotin was calculated to be 0.35 nN (27) and
sawtooth pattern, which is characteristic of between the essentially similar avidin and
nonspecific binding events. biotin to be 0.16 nN (28). The difference in
these two calculated values may result from
the different experimental setups used where
Ftip kDz 5 one protocol utilized a flat surface to immobi-
lize the protein, and the other used curved
agarose beads to minimize the potential for
where k = spring constant and z = tip multiple binding events.
deflection. Nonspecific binding interactions are a
Most cantilevers and tips used for AFM problem in all antibody-antigen techniques,
imaging are made either of silicon nitride and this is equally true for single-molecule
(Si3N4) or silicon (Si), which provides excel- measurements using AFM. The use of nonre-
lent mechanical elasticity. By fabricating the active blocking agents and detergents to
cantilevers in different geometric shapes and minimize nonspecific interactions has been
sizes, tips with different spring constants can extensively applied in traditional antibody-
be constructed. Selecting the cantilever with antigen assays such as ELISA. Similar ap-
the right spring constant is critically important proaches can be used for single molecule
for performing protein interaction measure- force spectroscopy to differentiate specific
ments. The smaller the spring constant, the antibody-antigen interaction from nonspecific
more sensitive the cantilever is and the weaker backgrounds as demonstrated on a ferritin and
force it can detect; on the other hand, the more antiferritin system, by using bovine serum
CHAPTER 22 Probing Antibody-Antigen Interactions 389

albumin and Tween 20 as unreactive proteins body-binding domains can be displayed on the
and detergents, respectively (29). surface of cells or virus particles, enabling the
It is important to point out that single- selection of displayed antibody fragments that
molecule interaction forces as measured by have particular binding properties or that have
AFM at a specific pulling rate represent a improved affinity compared with a parent
single point in a spectrum of binding strength. antibody. In the following section, we will
It is expected that different forces will be briefly describe how the different aforemen-
observed at different pulling rates. This prin- tioned techniques can also be used to select for
ciple was elegantly demonstrated by measur- antibody fragments with improved binding
ing the same streptavidin-biotin attractive characteristics.
forces with the use of a technique called
surface biomembrane force probe (30). The
SPR-Based Antibody Selection
biomembrane force probe is a single-molecule
force apparatus, from which the interaction SPR utilizes a microfluidic platform to first
force is detected from the deformation of the bind and then elute antibodies from antigens
vesicle or cell membrane with molecules immobilized on the chip surface. This feature
attached. The measured bond strengths varied can also be utilized to select for higher binding
from 5 pN to 170 pN, depending on the rate at variants of antibodies from a pool of anti-
which the forces were applied. By analyzing bodies. For example, in a study by Yuan et al., a
the slope of the force versus the loading rate, it library of higher binding variants of a parent
is possible to estimate the activation barriers of antibody binding the protein -amyloid (A)
the interaction complex at a specific loading were selected by using SPR (32). On average,
rate. antibody clones selected by SPR had fivefold
Similar experiments using AFM have also better dissociation rates than clones selected
been performed to determine the binding force by conventional means, and the best dissocia-
between the Fv fragments of antilysozyme and tion rate obtained with SPR was almost 10
lysozyme (31). At a loading rate of 5,000 pN/s, times better than that obtained conventionally
multiples of characteristic unbinding force of (32).
50 pN were detected from 900 binding SPR technology has also been used to
curves, which likely represents the rupture of characterize binding epitopes of two different
a single Fv and lysozyme complex. As expected, scFvs isolated against A (33). While tradi-
the measured pulling force varied at different tional immunoassays such as immunoblot,
loading rates. It is interesting to note that the ELISA, and immunoprecipitations can be
kinetic off-rate dissociation constant extrapo- used for epitope mapping, SPR does not
lated at zero force is 1 103 s1, which is in require a washing step to remove unbound
the same range as the value obtained using SPR material, which also makes it possible to
(3 103 s1). characterize low-affinity and transient inter-
actions (33). In this study by Liu et al., different
peptide epitopes of A (A1-16, A17-28, A29-
SELECTING ANTIBODIES WITH 42) were immobilized to SPR substrates and
DIFFERENT SPECIFICITIES BY USING SPR, different scFvs were injected over the chips.
FACS, AND AFM Binding specificity can be readily detected by
the association curves (Fig. 5). The association
While the various techniques described above (ka) and dissociation (kd) rate constants and the
can be effectively used to measure and study dissociation constants (KD = ka/kd) for the
antibody-antigen interactions, the same tech- entire AB peptide were also determined (Fig.
niques can also be used to select for antibodies 6) by fitting the data to a single binding site
with target-specific binding properties. Anti- model. The higher affinity of H1v2 [KD = (2.47
390 YANG ET AL.

FIGURE 5 Association curves demonstrate that H1v2 binds AB17-28 (A) and C1 binds AB29-40 (B).
doi:10.1128/microbiolspec.AID-0010-2013.f5

0.122) 107 M] compared with C1 [KD = 1.63 isolate cells expressing antibody fragments
0.74) 106 M] is primarily due to differences with high affinity for the given target antigen.
in the association rate constant, ka, rather than By the use of this protocol, antibody fragments
in the dissociation rate constant, kd. with high affinity against antigens, including
epidermal growth factor receptor, hen egg
lysozyme, and p-53 phosphopeptides were
FACS-Based Antibody Selection
readily isolated (14). A schematic for the
FACS can also be used to select for surface process of selection is shown in Fig. 7. The
display antibodies with increased binding KD values can be determined as described
affinities. Proteins such as antibody fragment earlier. One of the major advantages of using
libraries can be displayed on the surface of flow cytometry-based techniques is the ability
yeast cells by fusing the antibody fragment to a to distinguish between binders with high and
yeast agglutinin receptor (14). Cells can be low affinities. Negative selection steps can also
tagged with fluorescently labeled markers to be included in the selection process to facili-
identify the number of antibody fragments tate isolation of antibody fragments with
displayed and the number of antigen mole- specific binding properties. For example, an
cules bound (15). Cells can then be sorted to scFv that selectively inhibited only one side of
CHAPTER 22 Probing Antibody-Antigen Interactions 391

FIGURE 6 Association (ka) and dissociation (kd) rate constants and dissociation constants (KD = ka/kd) of
H1v2 (A) and C1 (B) to monomeric AB1-40. doi:10.1128/microbiolspec.AID-0010-2013.f6

AFM-Based Antibody Selection


a target proteolytic cleavage site on the
amyloid precursor protein was isolated by Since individual protein-binding events can be
first eliminating all cells that bound to a highly monitored by AFM, this provides a powerful
immunogenic site on one side of the cleavage tool to facilitate the isolation of antibody
site corresponding to the amino terminus of fragments with very selective binding spe-
the A protein implicated in Alzheimers cificities. For example, antibodies that recog-
disease (34). Monitoring the panning process nize specific protein variants or morphologies
by using flow cytometry ensured that the can have important implications for studying,
negative selection successfully eliminated diagnosing and treating different diseases. We
cells binding the nondesired antigen. Other developed an AFM-based biopanning protocol
panning techniques can be used to evaluate that utilizes the nanoscale imaging capability of
the individual clones for antigen binding. the AFM and the diversity of phage display
FACS enables the panning process to be antibody libraries to isolate antibody fragments
monitored in real time, greatly facilitating with very specific binding properties (35) (Fig.
the efficient selection of desired antibody 8). With the use of this AFM-based method,
fragments. antibody fragments against selected protein
392 YANG ET AL.

FIGURE 7 Illustration of magnetic bead-based (MACS) biopanning. Yeast cells expressing antibody
fragments (scFvs) are grown in medium containing galactose to induce expression and incubated with
biotinylated antigen and streptavidin or neutravidin magnetic beads. Yeast cells are loaded into a
magnetic column, and those expressing antigen-binding scFvs are retained. After multiple washes,
bound yeast cells can be plunged out and the process can be repeated several times to enrich for antigen
binders. doi:10.1128/microbiolspec.AID-0010-2013.f7

morphology targets can be used with high use of this AFM-based biopanning technique,
efficiency with the use of minimal sample antibody fragments with specificity for a variety
volumes and without the need to modify or of target protein morphologies were isolated,
label the antigen. including different oligomeric and fibrillar
In AFM biopanning, the target antigen is forms of -synuclein (36, 37) and A (38, 39, 40).
first immobilized on mica and then imaged by Most antibody selection methods require
using AFM to confirm that the desired antigen significant amounts of purified antigen target
morphology is present. A phage library is then for selection and screening and involve several
added to the antigen-coated mica to select for rounds of amplification to enrich the number of
the phage clones that have the strongest positive clones present in the eluted mixture.
affinity to that particular protein morphology However, many biologically important antigen
(Fig. 9). AFM is used to monitor each step of targets are either scant in availability, difficult to
the biopanning process, including binding, purify, or may be unstable. In order to isolate
washing, and elution, greatly increasing the antibodies to such antigens, a more direct
efficiency of the panning process and ensuring method would be to directly fish out the
that a high percentage of recovered clones desired antibodies. By the use of the AFM
have the desired binding specificity. With the cantilever as the fishing pole, a single-phage
CHAPTER 22 Probing Antibody-Antigen Interactions 393

FIGURE 8 Overview of the AFM-biopanning process. The target antigen is rst adsorbed on the mica
surface, and the selection of phage is performed by using multiple rounds of biopanning. Any unbound
phage is subsequently washed and removed, and bound phages are eluted from the mica surface. The
eluted phages are then amplied, puried, and used for subsequent rounds of panning. doi:10.1128/
microbiolspec.AID-0010-2013.f8

recovery technique using AFM was developed antibodies on one platform by using minimal
based on the principles of the AFM biopanning amounts of target protein, this AFM technology
technology (41). An image of the target protein- is very useful to generate affinity reagents to a
scFv complex is first acquired to determine the variety of target protein morphologies of inter-
presence of a desired target protein-phage. est that may be difficult to isolate by conven-
Once the protein-scFv complex of interest is tional methods such as ELISA. This protocol
determined, the AFM tip is positioned over the enables endless possibilities to detect the pre-
target and the applied force is increased in order sence of various transient intermediate protein
to pick up the phage-displaying scFv bound to structures that occur in vivo and that may be
the target protein. The DNA of the scFv can be involved in various human diseases.
recovered and amplified from the single-phage
molecule on the AFM tip by using PCR. The
advantages of this AFM single-phage recovery SUMMARY
technology include (i) selection for morpholo-
gy-specific scFvs from a mixture of different Antibody-antigen binding is a complex and
antigens, (ii) detection and recovery of anti- dynamic process driven by various noncovalent
bodies of transiently stable protein morpho- forces including hydrogen bond, van der Waals,
logies, and (iii) recovery of antibodies from ionic, and hydrophobic interactions. Since both
different regions of the same protein aggregate, antigen and antibody are flexible molecules that
e.g., the sides, ends, or along the axis of a fibril. are constantly moving, the different forces
With the ability to screen, select, and recover between the antibody and antigen are also
394 YANG ET AL.

FIGURE 9 Single-phage recovery using AFM. (Top) Phage DNA recovery on the AFM tip using PCR. (Bottom) AFM
image of the sample before (A) and after (B) phage pickup. doi:10.1128/microbiolspec.AID-0010-2013.f9

constantly changing during binding and un- Here, we present three additional tech-
binding events to accommodate for the changes niques, SPR, FACS, and AFM, that can also be
to the structure of each molecule. Therefore, it used to characterize antibody-antigen interac-
is crucial to understand and determine the tions. Each technique has unique detection
thermodynamic properties of antibody-antigen methods and provides some advantages over
interactions, such as the equilibrium binding traditional immunoassays. SPR offers a real-
(KD), association (kon), and dissociation rate time, label-free detection system by measuring
(koff) constants, in order to elucidate the affinity the changes in refractive index of the sensor
and specificity of an antibody to its target chip surface. This allows for continuous
antigen. Traditional immunoassays such as monitoring of the kinetic profile between an
RIA and ELISA have long been used to immobilized agent and free-flowing analyte,
determine the steady-state equilibrium inter- and thermodynamic constants can be easily
actions between an antibody and antigen and obtained from the SPR sensograms. In addi-
are excellent techniques for large-volume, tion, different sensor surface chemistries are
high-throughput screening of antibodies available for antigen or antibody immobiliza-
against an array of antigens. However, these tion, which increases the overall number and
techniques are not suitable for more intricate variety of targets that can be analyzed. FACS
analysis such as a precise characterization of an utilizes fluorescently labeled antibodies for
antibody-antigen interaction, the determina- characterization of binding events on cell
tion of antibody specificity for rare and unstable surfaces in order to sort and count cells that
antigens, or for a low-volumehigh-throughput have the desired properties. Additionally, the
screening of antibody libraries. use of multiple labeled antibodies is possible in
CHAPTER 22 Probing Antibody-Antigen Interactions 395

FACS, which allows for the simultaneous REFERENCES


analysis of multiple targets on a single cell. 1. Reverberi R, Reverberi L. 2007. Factors affecting
Because thousands of particles per second can the antigen-antibody reaction. Blood Transfus
be easily counted and sorted by using FACS, 5:227240.
this technique is very efficient for drug or 2. Ritzefeld M, Sewald N. 2012. Real-time analysis
antibody screening. Finally, AFM provides a of specific protein-DNA Interactions with sur-
face plasmon resonance. J Amino Acids 2012:
high-sensitivity detection method to detect 816032.
antibody-antigen interactions without the 3. Zhu G, Yang B, Jennings RN. 2000. Quantitation
need of a label. Individual molecular forces of basic fibroblast growth factor by immunoassay
between an antibody and antigen can be using BIAcore 2000. J Pharm Biomed Anal
elucidated by using the AFM, as well as the 24:281290.
4. Jonsson U, Fgerstam L, Ivarsson B, Johnsson
folding and unfolding of a biomolecule by B, Karlsson R, Lundh K, Lfs S, Persson B, Roos
varying the loading rates. In addition to force H, Ronnberg I, et al. 1991. Real-time biospecific
measurements, the intricate structures or interaction analysis using surface plasmon reso-
morphologies of biomolecules can be obtained nance and a sensor chip technology. Biotechniques
11:620627.
by using the AFM with high resolution. These
5. Guo X. 2012. Surface plasmon resonance based
capabilities can facilitate more precise anti- biosensor technique: a review. J Biophotonics 5:
body selection methods against different target 483501.
protein morphologies. 6. Mannen T, Yamaguchi S, Honda J, Sugimoto S,
The demand for antibodies with higher Kitayama A, Nagamune T. 2001. Observation of
affinity and specificity is increasingly important charge state and conformational change in
immobilized protein using surface plasmon reso-
for both diagnostic and therapeutic appli-
nance sensor. Anal Biochem 293:185193.
cations to eliminate nonspecific and off-target 7. Karlsson R, Kullman-Magnusson M, Hamalainen
interactions. In order to design and synthesize MD, Remaeus A, Andersson K, Borg P, Gyzander
the next-generation set of antibodies, analytical E, Deinum J. 2000. Biosensor analysis of drug-
methods are needed to characterize, screen, target interactions: direct and competitive binding
assays for investigation of interactions between
sort, select, and determine the binding kinetics
thrombin and thrombin inhibitors. Anal Biochem
of the antibodies against an array of antigens. 278:113.
The techniques discussed in this article are 8. Kruger R, Kuhn W, Mller T, Woitalla D,
among some of the modern analytical tools Graeber M, Ksel S, Przuntek H, Epplen JT,
that can be used to study antibody-antigen Schls L, Riess O. 1998. Ala30Pro mutation in the
interactions. They are good alternatives or gene encoding alpha-synuclein in Parkinsons
disease. Nat Genet 18:106108.
complementary techniques to the classical
9. Daghestani HN, Day BW. 2010. Theory and
immunoassays in providing a fast, label-free, applications of surface plasmon resonance, reso-
low-volume, high-throughput screening of nant mirror, resonant waveguide grating, and dual
antibodies with high specificity and a molec- polarization interferometry biosensors. Sensors
ular level of detection resolution. (Basel) 10:96309646.
10. Boozer C, Kim G, Cong S, Guan H, Londergan T.
2006. Looking towards label-free biomolecular
interaction analysis in a high-throughput format: a
ACKNOWLEDGMENT review of new surface plasmon resonance tech-
nologies. Curr Opin Biotechnol 17:400405.
Conflicts of interest: We disclose no conflicts.
11. Rieseberg M, Kasper C, Reardon KF, Scheper T.
2001. Flow cytometry in biotechnology. Appl
Microbiol Biotechnol 56:350360.
CITATION 12. Benedict CA, MacKrell AJ, Anderson WF. 1997.
Determination of the binding affinity of an anti-
Yang G, Velgos SN, Boddapati SP, Sierks MR. CD34 single-chain antibody using a novel, flow
2014. Probing antibody-antigen interactions. cytometry based assay. J Immunol Methods 201:
Microbiol Spectrum 2(1):AID-0010-2013 223231.
396 YANG ET AL.

13. Lou J, Geren I, Garcia-Rodriguez C, Forsyth imaging of the human trigeminal ganglion. Neuro-
CM, Wen W, Knopp K, Brown J, Smith T, Smith image 14:13481352.
LA, Marks JD. 2010. Affinity maturation of 25. Nag K, Munro JG, Hearn SA, Rasmusson J,
human botulinum neurotoxin antibodies by light Petersen NO, Possmayer F. 1999. Correlated
chain shuffling via yeast mating. Protein Eng Des atomic force and transmission electron microsco-
Sel 23:311319. py of nanotubular structures in pulmonary sur-
14. Feldhaus MJ, Siegel RW, Opresko LK, Coleman factant. J Struct Biol 126:115.
JR, Feldhaus JM, Yeung YA, Cochran JR, 26. Ebner A, Hinterdorfer P, Gruber HJ. 2007.
Heinzelman P, Colby D, Swers J, Graff C, Comparison of different aminofunctionalization
Wiley HS, Wittrup KD. 2003. Flow-cytometric strategies for attachment of single antibodies to
isolation of human antibodies from a nonimmune AFM cantilevers. Ultramicroscopy 107:922927.
Saccharomyces cerevisiae surface display library. 27. Lee GU, Kidwell DA, Colton RJ. 1994. Sensing
Nat Biotechnol 21:163170. discrete streptavidin-biotin interactions with
15. Chao G, Lau WL, Hackel BJ, Sazinsky SL, atomic force microscopy. Langmuir 10:354357.
Lippow SM, Wittrup KD. 2006. Isolating and 28. Florin EL, Moy VT, Gaub HE. 1994. Adhesion
engineering human antibodies using yeast surface forces between individual ligand-receptor pairs.
display. Nat Protoc 1:755768. Science 264:415417.
16. Li B, Zhao L, Guo H, Wang C, Zhang X, Wu L, 29. Wakayama J, Sekiguchi H, Akanuma S, Ohtani
Chen L, Tong Q, Qian W, Wang H, Guo Y. 2009. T, Sugiyama S. 2008. Methods for reducing
Characterization of a rituximab variant with nonspecific interaction in antibody-antigen assay
potent antitumor activity against rituximab-resis- via atomic force microscopy. Anal Biochem
tant B-cell lymphoma. Blood 114:50075015. 380:5158.
17. Geuijen CA, Clijsters-van der Horst M, Cox F, 30. Merkel R, Nassoy P, Leung A, Ritchie K, Evans
Rood PM, Throsby M, Jongeneelen MA, Backus E. 1999. Energy landscapes of receptor-ligand
HH, van Deventer E, Kruisbeek AM, Goudsmit bonds explored with dynamic force spectroscopy.
J, de Kruif J. 2005. Affinity ranking of antibodies Nature 397:5053.
using flow cytometry: application in antibody 31. Berquand A, Xia N, Castner DG, Clare BH,
phage display-based target discovery. J Immunol Abbott NL, Dupres V, Adriaensen Y, Dufrene
Methods 302:6877. YF. 2005. Antigen binding forces of single
18. Daugherty PS, Chen G, Olsen MJ, Iverson BL, antilysozyme Fv fragments explored by atomic
Georgiou G. 1998. Antibody affinity maturation force microscopy. Langmuir 21:55175523.
using bacterial surface display. Protein Eng 11:825 32. Yuan B, Schulz P, Liu R, Sierks MR. 2006.
832. Improved affinity selection using phage display
19. Boder ET, Midelfort KS, Wittrup KD. 2000. technology and off-rate based selection. Electron J
Directed evolution of antibody fragments with Biotechnol 9:171175.
monovalent femtomolar antigen-binding affinity. 33. Liu R, Yuan B, Emadi S, Zameer A, Schulz P,
Proc Natl Acad Sci USA 97:1070110705. McAllister C, Lyubchenko Y, Goud G, Sierks
20. Neuman KC, Nagy A. 2008. Single-molecule force MR. 2004. Single chain variable fragments against
spectroscopy: optical tweezers, magnetic tweezers beta-amyloid (Abeta) can inhibit Abeta aggrega-
and atomic force microscopy. Nature Methods 5: tion and prevent abeta-induced neurotoxicity.
491505. Biochemistry 43:69596967.
21. Binnig G, Rohrer H, Gerber C, Weibel E. 1982. 34. Boddapati S, Levites Y, Sierks MR. 2011. Inhib-
Surface studies by scanning tunneling microsco- iting -secretase activity in Alzheimers disease
py. Phys Rev Lett 49:5761. cell models with single-chain antibodies specifi-
22. Hansma PK, Cleveland JP, Radmacher M, cally targeting APP. J Mol Biol 405:436447.
Walters DA, Hillner PE, Bezanilla M, Fritz M, 35. Barkhordarian H, Emadi S, Schulz P, Sierks MR.
Vie D, Hansma HG, Prater CB, Massie J, 2006. Isolating recombinant antibodies against
Fukunaga L, Gurley J, Elings V. 1994. Tapping specific protein morphologies using atomic force
mode atomic-force microscopy in liquids. Appl microscopy and phage display technologies. Pro-
Phys Lett 64:17381740. tein Eng Des Sel 19:497502.
23. Matsko N. 2007. Atomic force microscopy applied 36. Emadi S, Barkhordarian H, Wang MS, Schulz P,
to study macromolecular content of embedded Sierks MR. 2007. Isolation of a human single
biological material. Ultramicroscopy 107:95105. chain antibody fragment against oligomeric alpha-
24. Melling M, Hochmeister S, Blumer R, Schilcher synuclein that inhibits aggregation and prevents
K, Mostler S, Behnam M, Wilde J, Karimian- alpha-synuclein-induced toxicity. J Mol Biol 368:
Teherani D. 2001. Atomic force microscopy 11321144.
CHAPTER 22 Probing Antibody-Antigen Interactions 397

37. Emadi S, Kasturirangan S, Wang MS, Schulz P, oligomeric -amyloid stabilizes nontoxic form.
Sierks MR. 2009. Detecting morphologically dis- Neurobiol Aging 33:13201328.
tinct oligomeric forms of alpha-synuclein. J Biol 40. Marcus WD, Wang H, Lindsay SM, Sierks MR.
Chem 284:1104811058. 2008. Characterization of an antibody scFv that
38. Zameer A, Kasturirangan S, Emadi S, recognizes fibrillar insulin and beta-amyloid using
Nimmagadda SV, Sierks MR. 2008. Anti-oligo- atomic force microscopy. Nanomedicine 4:17.
meric Abeta single-chain variable domain antibody 41. Lyubchenko Y, Gall A, Shlyakhtenko L,
blocks Abeta-induced toxicity against human Harrington R, Jacobs B, Oden P, Lindsay S.
neuroblastoma cells. J Mol Biol 384:917928. 1992. Atomic force microscopy imaging of double
39. Kasturirangan S, Li L, Emadi S, Boddapati S, stranded DNA and RNA. J Biomol Struct Dyn 10:
Schulz P, Sierks M. 2012. Nanobody specific for 589606.
Radiolabeled Antibodies for
Therapy of Infectious Diseases

EKATERINA DADACHOVA1,2 and ARTURO CASADEVALL2,3


23
INTRODUCTION

There is a growing need for alternatives to conventional antibiotics for the


treatment of infectious diseases. The number of bacterial pathogens that are
resistant even to the most powerful antibiotics is growing each year. HIV remains
an incurable disease more than 30 years since its identification. Since 1979 there
has been a >200% increase in the annual number of cases of invasive fungal
infections in the United States. To exacerbate these problems, the number of
patients who cannot fight infections because of impaired immunity is growing and
includes HIV patients, patients who have been through cancer chemotherapy, and
organ transplant recipients.
Radioimmunotherapy (RIT) is based on the interaction between the pair of
antigen and antibody to carry cytocidal amounts of ionizing radiation to the
vicinity of specific cellular targets. Currently, RIT is clinically utilized in the
treatment of primary, refractory, and recurrent non-Hodgkin lymphoma with
the radiolabeled monoclonal antibodies (mAbs) Zevalin and Bexxar, and it offers
several significant advantages over naked antibody strategies: (i) RIT delivers
lethal radiation, such that it does not merely interfere with a single cellular
pathway but leads to physical destruction of targeted cells via radiation-induced

1
Department of Radiology; 2Department of Microbiology and Immunology; 3Department of Medicine,
Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0023-2014

399
400 DADACHOVA AND CASADEVALL

apoptosis/autophagy/necrosis; (ii) RIT is not C. neoformans results in life-threatening


subject to drug resistance mechanisms such as meningoencephalitis, which affects people
efflux through drug efflux pumps in malignant with a compromised immune system and is
cells; (iii) the effectiveness of RIT does not responsible for higher mortality among indi-
depend on the immunological status of the host; viduals with AIDS in sub-Saharan Africa than
(iv) RIT has the potential to reduce the number tuberculosis (2). The availability of good
of doses used to combat infections with stan- animal models and well-characterized mAbs
dard therapies from weeks or months to a single to C. neoformans antigens provided an impetus
or limited number of doses of RIT. to use this pathogen for investigating RIT for
A decade ago we suggested the use of RIT for infections. Importantly, immunotherapy in
the treatment of the fungal pathogen Crypto- patients with C. neoformans with the capsule
coccus neoformans (1). Since then we have polysaccharide-binding antibody 18B7 has
evaluated the suitability of this approach to already been evaluated clinically (3). Thera-
treating fungal infections for its efficacy and peutic studies employed AJ/Cr mice infected
safety as well as expanded it to treating systemically with C. neoformans. A/JCr mice
infections due to bacteria and viruses (Fig. 1). succumb to the systemic infection with C.
Here we provide a brief overview of the precli- neoformans, most likely because of the partial
nical development of RIT for infectious diseases. complement deficiency (4). The survival of
mice treated with radiolabeled C. neoformans
specific mAb 18B7 was significantly longer
FUNGAL INFECTIONS than the survival of mice treated with irrele-
vant labeled IgG1 or phosphate-buffered
We started with the investigation of RIT po- saline. We utilized a radiolabeled irrelevant
tency against C. neoformans fungal infection (1). mAb (213Bi- or 188Re-IgG1 MOPC21) to take

FIGURE 1 Mechanisms of RIT efcacy against infections. (a) Direct targeting of microbial cells with the
radiolabeled organism-specic antibodies. (b) Killing of virally infected host cells by targeting viral
antigens expressed on the surface of infected cells. doi:10.1128/microbiolspec.AID-0023-2014.f1
CHAPTER 23 Radiolabeled Antibodies for Therapy of ID 401

into account the possibility of the radiolabeled infection resulted in one log reduction in the
IgG binding to Fc receptors on phagocytic lung CFUs (P = 0.04), while no reduction in
cells present at the infected site, which might CFUs was observed in the brains (P = 0.07)
lead to nonspecific killing of some C. neo- (Fig. 2b). Treatment with 213Bi-18B7 mAb 24 h
formans cells. Interestingly, treatment with postinfection completely killed fungal cells in
100 Ci 213Bi-18B7 resulted in 60% of mice in the lungs and the brains (the plating assay
the 213Bi group surviving on day 75 post- sensitivity was 50 CFUs) (Fig. 2b), which agreed
therapy (P < 0.05). In the 188Re group 40% with the results of Gomon methenamine silver
and 20% of animals were alive after treatment (GMS) staining. Interestingly, for established
with 100 (P < 0.005) and 50 (P < 0.05) Ci infection at 48 h, 188Re-18B7 mAb efficiently
188
Re-18B7, respectively. In contrast, mice in decreased CFUs in the lungs (P = 0.03) and also
control groups died from C. neoformans notably decreased the number of fungal cells in
infection on days 35 to 40 (Fig. 2a). Adminis- the brains of treated mice (P = 0.02) (Fig. 2b).
tration of RIT resulted in significant reduction The latter observation might stem from the
of fungal burden in lungs and brains of mice increased permeability of the blood-brain bar-
with C. neoformans 48 h after RIT adminis- rier caused by the presence of infection, which
tration when compared to control groups. No results in facilitating the entrance of the
difference in the percentage decrease of the radiolabeled antibody into the brain (6). We
fungal burden in the lungs was observed concluded from this study that RIT demon-
between the groups that received 50 and 100 strated efficacy in both early and established
Ci 188Re-18B7. In contrast, the administration infection in immunocompetent C57BL6 mice.
of 200 Ci 188Re-18B7 led to pronounced reduc- In our previous work we showed RIT efficacy in
tion in lung CFUs when compared to the lower complement-deficient AJ/Cr mice. The results
activities (P < 0.05). We concluded that giving are consistent with and supportive of the pre-
a radiolabeled antibody to fungal polysaccha- sumed RIT independence of the status of the
ride to infected mice led to the prolongation in hosts immune system. Such a finding is encour-
survival and reduction in organ fungal burden. aging for the treatment of opportunistic infec-
Subsequently we investigated whether tions in cancer and organ transplant patients.
RIT was effective in a different mouse As a part of our effort to demonstrate the
modelimmunocompetent C57BL6 mice value of RIT as an anti-infective strategy, we
and whether it would be effective in the setting compared its efficacy with amphotericin B,
of more established cryptococcal infection. which is one of the major antifungal drugs (7).
RIT kills the microorganisms primarily by AJ/Cr mice were infected i.v. with 3 105
targeted cytocidal radiation, so it was para- melanized or nonmelanized C. neoformans
mount to establish its presumed independence cells. Twenty-four hours after infection the
from the immune status of the host. Addition- mice were given i.p. either 100 Ci 213Bi-18B7
ally, showing that RIT can be efficacious in or amphotericin in its deoxycholate form at
the setting of the established infections ac- 1 g/g body weight at 24, 48, and 72 h, were
companied by the high fungal load would be given both treatments, or were left untreated.
useful for its future translation into the clinic. Mouse survival was observed for 60 days. The
For this purpose C57BL6 mice were infected lungs and brains were analyzed at 60 days
intravenously (i.v.) via the tail vein with postinfection. This analysis demonstrated that
106C. neoformans cells and were left either treatment with amphotericin did not signifi-
untreated or treated intraperitoneally (i.p.) cantly decrease lung and brain CFUs in either
with 100 Ci 213Bi-18B7 24 h postinfection, nonmelanized or melanized C. neoformans
100 Ci 188Re-18B7 24 h postinfection, or groups (Fig. 2c) (P > 0.05). RIT administration
100 Ci 188Re-18B7 48 h postinfection (5). resulted in noted decreases in fungal burden in
Administration of 188Re-18B7 mAb 24 h post- comparison with the untreated controls and
402 DADACHOVA AND CASADEVALL

FIGURE 2 RIT of experimental fungal infections with 213Bi- and 188Re-labeled mAbs. (a) Kaplan-Meier
survival curves for A/JCr mice infected i.v. with 105 C. neoformans cells 24 h prior to treatment with 50 to
200 Ci 188Re-labeled mAbs. Animals injected with phosphate-buffered saline or 50 g cold 18B7 served
as controls. (b) RIT of C57BL6 mice infected i.v. with 106 C. neoformans cells: CFUs in the brains and
the lungs of RIT-treated and control mice. Mice were treated i.p. with either 100 Ci 213Bi-18B7 24 h
postinfection, 100 Ci 188Re-18B7 24 h postinfection, or 100 Ci 188Re-18B7 48 h postinfection or were
left untreated and sacriced 75 days posttreatment. The detection limit of the method was 50 CFUs. No
CFUs were found in the brains and lungs of mice treated with 100 Ci 213Bi-18B7, which are presented in
the graph as 40 CFUs/organ. The asterisks show the groups in which the CFUs were signicantly different
from the untreated controls. (c) Comparison of RIT and amphotericin B efcacy toward melanized
C. neoformans in vivo. CFUs in the lungs and brains of mice infected with melanized C. neoformans. AJ/Cr
mice were infected i.v. with 3 105 C. neoformans cells and were given either 100 Ci 213Bi-18B7 RIT or
amphotericin B at 1 g/g body weight on days 1, 2, and 3 postinfection or combined treatment or left
untreated. The detection limit of the method was 50 CFUs. No CFUs were found in the brains and lungs of
mice infected with melanized C. neoformans cells and treated with RIT, which are presented in the graph
as 40 CFUs/organ. (d) Median survival of AJ/Cr mice infected i.v. with 5 104 C. neoformans and treated
24 h later with 150 Ci 188Re-18B7 or 125 Ci 213Bi-18B7 mAb. CNnave, cells from ATCC; CNRe RIT, cells
recovered from mice treated with 188Re-18B7 mAb; CNBi RIT, cells recovered from mice treated with 213Bi-
18B7 mAb; Re RIT/CNnave, mice infected with CNnave and treated with 188Re-18B7; Bi RIT/CNnave, mice
infected with CNnave and treated with 213Bi-18B7; Re RIT/CNRe RIT, mice infected with CNRe RIT and treated
with 188Re-18B7; Bi RIT/CNBi RIT, mice infected with CNBi RIT and treated with 213Bi-18B7. (e, f) RIT with
213
Bi-4E12 antibody to Hsp60: (e) RIT of C. albicans; (f) RIT of C. neoformans. Each experiment was
performed three times, and the results shown are from one typical experiment. The CFUs for each
antibody dose were plated in triplicate. doi:10.1128/microbiolspec.AID-0023-2014.f2
CHAPTER 23 Radiolabeled Antibodies for Therapy of ID 403

the amphotericin-treated mice (P << 0.05). Of with 213Bi-18B7 mAb (CNBi-RIT), or with the
note, the fungus was practically cleared from RIT-nave C. neoformans cells (CNnave) (11).
the brains of the RIT-treated nonmelanized We treated C. neoformans-infected mice with
C. neoformans group (sensitivity of the detec- 150 Ci 188Re-18B7 or 125 Ci 213Bi-18B7 24 h
tion was 50 CFUs), while in the melanized after i.v. infection and then observed the mice
C. neoformans group RIT was capable of for survival and weight loss. The number of
practically clearing the infection from both deaths in mice infected with CNRe-RIT or CNBi-
brain and lungs. Our most important result was RIT was similar to that in mice infected with
the observation of the RIT efficacy in reducing CNnave (P > 0.05) (Fig. 2d). Mice given 213Bi-
fungal burden in lungs and brains when 18B7 mAb survived longer (P = 0.04) than those
compared to the high 1-g/g dose of ampho- given 188Re-18B7 (Fig. 2d), most likely due to the
tericin, when the majority of mice treated with higher killing power of 213Bi-emitted alpha
RIT were able to almost completely clear the particles when compared to 188Re-emitted beta
infection. The explanation of the inability of particles. In general, the interaction of fungal
amphotericin to decrease the fungal burden in cells with particulate radiation resulted in the
the organs of partially complement-deficient loss of the cells ability to divide (1, 12), which
AJ/Cr mice 3 days posttreatment was provided could provide an explanation for the
by the subsequent amphotericin study show- nonemergence of radiation-resistant pheno-
ing a trend toward a decrease in CFUs in brains types post-RIT. The residual cells that replicat-
and lungs that manifested itself only on day 14 ed post-RIT were probably shielded from the
of treatment with amphotericin B (7). antibodies that received radiation from a
Our results are in accordance with the biofilm, an abscess, or a host cell.
published data showing that amphotericin Finally, in an effort to develop broader anti-
was able to produce only a 1 to 1.5 log reduction fungal therapy that did not rely on pathogen-
in CFUs in immunocompetent mice such as specific mAbs, we investigated the targeting of
CD-1 and BALB/c, and all mice succumbed to antigens shared by major invasive fungal
C. neoformans infection around day 24 (8, 9). infection-causing fungi (pan-fungal antigens)
Our results also agree with the clinical reports to deliver RIT without the need for specific
that show that a short course of amphotericin mycological diagnosis or concerns about drug
was not able to sterilize cerebrospinal fluid resistance. We explored the possibility of
or blood of patients and that correlated the targeting common cell-wall-associated anti-
rate of sterilization with patient survival (10). gens, which also happen to be the dominate
Our observations underline the advantages of virulence factors for these fungal pathogens.
RIT, which has pronounced antimicrobial The majority of fungal cells, in both yeast and
effects in vivo after just one injection, com- hyphal forms, display beta-glucans on their
pared to long and often ineffective treatments cell surface. Cassone and colleagues generated
with amphotericin, which has long-lasting a mAb to beta-glucans in Candida albicans,
side effects. C. neoformans, and Aspergillus fumigatus in
Despite radiation being a weak mutagen, animal models (1315). Heat shock protein
the belief that even low doses of ionizing 60 (Hsp60) is a major regulator of virulence in
radiation are able to create potentially danger- Histoplasma capsulatum, and mAbs directed
ous cellular mutants persists within the scien- to this protein are protective in murine histo-
tific community and the lay public alike. To plasmosis (16). We established that a mAb to
evaluate the possibility that RIT might select H. capsulatum Hsp60 also bound other path-
for the radiation resistance in C. neoformans ogenic fungal species but did not react with
cells in vivo, AJ/Cr mice were infected with human Hsp60 (16). Melanin is present in the
C. neoformans cells recovered from mice treated cell wall of diverse human fungal pathogens,
previously with 188Re-18B7 mAb (CNRe-RIT), and a mAb 6D2 to fungal melanin was shown
404 DADACHOVA AND CASADEVALL

to bind C. neoformans, H. capsulatum, Asper-


gillus spp., C. albicans, Scytalidium dimidiatum,
Sporothrix schenckii, Paracoccidioides brasiliensis,
Coccidioides posadassi, and Blastomyces derma-
titidis (17). To explore the feasibility of using
RIT to target these pan-antigens we utilized
mAbs 4E12, an IgG2a to fungal HSP60; 2G8, an
IgG 2b to beta-(1,3)-glucan; and 6D2, an IgM
to melanin, and radiolabeled them with 213Bi
(18). C. neoformans and C. albicans were used
to evaluate the cytocidal effects of these
radiolabeled mAbs. 213Bi-labeled mAbs to
HSP60 (Fig. 2e,f) and to the beta-(1,3)-glucan
each decreased the viability of both fungi in
the 80 to 100% range. The 213Bi-6D2 mAb to
melanin eliminated 50% of C. neoformans cells
but did not kill C. albicans. Treatment with
unlabeled mAbs and with radiolabeled
isotype-matching control mAbs resulted in
no killing. These results point to the possibility
of developing RIT against fungal pathogens FIGURE 3 RIT of bacterial infections. (a) S. pneumo-
by targeting shared fungal antigens. This niae, 213Bi-labeled mAbs in C57BL/6 mice. Mice were
approach could be utilized against fungal infected i.p. with 1,000 organisms 1 h before treat-
ment with mAbs. (b) RIT of B. anthracis Sterne
infections for which current therapies are infection with 213Bi-labeled mAbs. Mice were infected
not working well. 1 h prior to labeled-mAb treatment. Survival exper-
iment was repeated three times with similar results.
Controls include unlabeled mAbs given in the same
BACTERIAL INFECTIONS amounts (15 g) as radiolabeled mAbs.
doi:10.1128/microbiolspec.AID-0023-2014.f3

Streptococcus pneumoniae, an important cause


of community-acquired pneumonia, meningi- explanation could be the absence of a target
tis, and bacteremia, was selected for evaluating for the irrelevant radiolabeled mAb to bind to,
the feasibility of RIT against bacterial diseases which resulted in an excessive dose of radia-
(19). A human mAb D11 that binds to pneumo- tion to the blood-rich dose-limiting organs
coccal capsular polysaccharide 8 (PPS 8) was such as bone marrow. Mice in control groups
chosen as a delivery vehicle for RIT and was died from bacteremia on days 1 to 3, while 87 to
radiolabeled with a short-range alpha-emitter, 100% of mice given 80 Ci 213Bi-D11 survived.
213
Bi. The RIT administration resulted in a Measuring CFUs in the blood of the RIT-
higher percentage of mice surviving in the treated mice demonstrated that they were not
213
Bi-D11-treated group compared to the un- bacteremic at 3, 6, and 10 h posttreatment or on
treated group (P < 0.01) (Fig. 3a). In contrast, days 3 and 14. RIT with radiolabeled D11 did
giving mice 5 g unlabeled D11 mAb did not not cause any weight loss in treated animals. In
lead to the prolongation in survival in compar- summary, this proof of principle study was the
ison to untreated mice (P > 0.05). Radiolabeled first to demonstrate the ability of RIT to treat
irrelevant IgM also did not show any thera- experimental bacterial infection.
peutic results (P > 0.05) and, in contrast, fewer Later we investigated RIT for experimental
mice survived its administration when com- Bacillus anthracis infection. B. anthracis is a
pared to the untreated group. A possible potential agent for bioterrorism and biological
CHAPTER 23 Radiolabeled Antibodies for Therapy of ID 405

weapons, which underscores the necessity for experiments demonstrated that dormant
additional, different mode of action therapies spores were not killed by 213Bi -EA2-1 mAb
for anthrax (20). The surface expression of either. Only when the dose of 213Bi-EA2-1 mAb
toxins on bacterial cells was demonstrated by reached 300 Ci was the significant spore
indirect immunofluorescence experiments killing observed. However, this killing was not
with mAbs to protective antigen (7.5G g2b mAb-specific, as the isotype control mAb
and 10F4 g1) and lethal factor (mAb 14FA g2b). labeled with 300 Ci 213Bi showed the same
Scatchard analysis of mAbs binding to the results. Our next step was to examine the
bacterial surface demonstrated high binding effects of RIT on the germinating versus
constants and multiple binding sites on the dormant spores. The reasoning for that was
surface of bacteria, which provided the impe- that the spores become pathogenic in a host
tus for RIT studies. The mAbs to the toxins when they start germinating and dividing,
were radiolabeled with either 188Re or 213Bi for which leads to the development of anthrax
investigating the microbicidal potential of RIT. disease. In addition, it is known from classical
213
Bi-labeled mAbs were more efficient in vitro radiobiology that dividing cells are much more
than 188Re-labeled mAbs in killing B. anthracis susceptible to ionizing radiation damage than
Sterne bacterial cells. Giving i.p. 213Bi-labeled cells that are not dividing. Thus, the germinat-
mAbs 10F4 and 14FA to A/JCr mice lethally ing spores might present a better target for
infected with B. anthracis cells significantly RIT. The experiments showed that 75 and
prolonged their survival (Fig. 3b). Our results 150 Ci 213Bi-EA2-1 killed significant numbers
point to RITs utility in treating experi- of germinating spores, while the matching
mental anthrax infection with mAbs targeting activities of the isotype matching control
B. anthracis tri-partite toxin components and mAb did not. We concluded from this study
suggest that toxigenic bacteria may be targeted that while dormant spores are resistant to both
with radiolabeled mAbs to its toxins. external radiation and RIT, the germinating
Finally, we recently investigated the poten- spores are RIT-susceptible, and this line of
tial of RIT against germinating B. anthracis study should be investigated further, possibly
spores (21). B. anthracis spores are covered by in animal models.
an impenetrable two-layered exosporium,
which is composed of a basal layer and an
external hair-like nap. The nap consists of HIV
filaments, which in turn are composed of
trimers of a collagen-like glycoprotein BclA. The HIV epidemic remains a major worldwide
BclA is considered to be an immunodominant health problem. Highly active antiretroviral
antigen on the spore surface. The antibodies therapy (HAART), a combination of drugs that
to BclA are highly specific and can specific- inhibits enzymes essential for HIV replication,
ally identify B. anthracis spores among the can decrease the viremia to almost undetectable
spores produced by other Bacillus species. We levels and decrease the likelihood of opportu-
investigated whether EA2-1 mAb to BclA nistic infections in the majority of patients. As
armed with 213Bi would be capable of steriliz- a result, patients on HAART now survive for
ing B. anthracis spores. We chose an alpha- decades. However, HAART regimens are com-
emitter 213Bi for this study, as this radionu- plex and require lifelong use, and many have
clide was successfully used in our previous significant long-term side effects such as
research on RIT for bacterial pathogens such metabolic syndrome, cardiotoxicity, etc. A
as S. pneumoniae and B. anthracis. replication-competent virus that hides in
First, we confirmed the previous reports latently infected cells serves as a source of
that the spores were completely resistant to viremia that emerges rapidly after the discontin-
the external gamma radiation. Our initial RIT uation of HAART. A modality that specifically
406 DADACHOVA AND CASADEVALL

targets and eliminates HIV-infected cells in tration of 188Re-armed 246-D mAb before or
patients on HAART could be a major contrib- after intrasplenic injection with HIV-infected
utor toward the eradication of persistent HIV hPBMCs significantly decreased the numbers
cellular reservoirs. We hypothesized that RIT of HIV-infected cells in mice (Fig. 4a). Similar
could be able to kill virally infected cells. RIT for results were obtained with 213Bi-246-D (Fig.
viral diseases would target viral antigens on 4a). In contrast, control SCID mice that
infected cells and consequently would provide a received equivalent amounts of cold mAb
completely different approach for treating HIV. 246-D or a radiolabeled isotype-matched con-
Initially, we studied the efficacy of RIT trol mAb showed no reduction in the average
against HIV infection in SCID mice using an number of infected cells detected in their
HIV envelope-specific human mAb 246-D to spleens. These results established the feasibil-
gp41 that we radiolabeled with 213Bi or 188Re ity of using RIT to specifically target and
(22). For these experiments human peripheral eliminate HIV-infected hPBMCs in vivo and
blood mononuclear cells (hPBMCs) were provided a first experimental proof for the
infected with HIV-1JR-CSF and injected intra- concept of fighting viral infections by targeting
splenically into SCID mice, and radiolabeled virally infected cells with the radioactively
mAbs were given i.p. 1 h later. The mice were armed mAbs to viral antigens. We anticipate
sacrificed 72 h after RIT, and the presence of that the same approach could be useful for
the residual HIV-infected cells was estab- treatment of other chronic viral infections, e.g.,
lished by quantitative coculture (23). This hepatitis C (24).
time interval was chosen to provide enough It should be noted that the antibodies used
time for the 188Re-radiolabel (its physical half- in RIT do not neutralize the virus and conse-
life is 16.9 h) on the mAb to deliver a lethal quently are not expected to exert selective
cytotoxic dose to the infected cells. Adminis- pressure on the virus. Only the epitopes on the

FIGURE 4 RIT of SCID mice injected intrasplenically with JR-CSF HIV-infected human PBMCs and treated with
188
Re- and 213Bi-labeled human anti-gp41 mAbs 246-D (a) or 2556 (b). (a) Limiting coculture results for 246-D
mAb. Mice received either 20 g cold anti-gp41 mAb 246-D, 100 Ci (20 g) 213Bi-1418 or 80 Ci (20 g) 188Re-
1418 as isotype-matching controls, 80 Ci (20 g) 188Re-246-D, or 100 Ci (20 g) 213Bi-246-D i.p. 1 h after
injection of PBMCs. In some experiments mice were given 80 Ci (20 g) 188Re-246-D i.p. 1 h prior to injection of
HIV-infected PBMCs. (b) PCR data for RIT with 50, 100, and 200 Ci 213Bi-2556 mAb. The cold 2556, untreated
mice, and matching activities of the irrelevant 1418 mAb were used as controls.
doi:10.1128/microbiolspec.AID-0023-2014.f4
CHAPTER 23 Radiolabeled Antibodies for Therapy of ID 407

viral proteins that are conserved throughout all investigate the potential bone marrow toxicity
HIV strains and clades, which is consistent of RIT, the peripheral blood of treated mice
with their role in the maintenance of envelope was analyzed for platelet counts. A drop in
protein structure, are chosen as targets for RIT. platelet numbers would be evidence of an
As a result, even in case of a mutation, such undesirable effect of the radiolabeled mAb on
epitopes will probably be present on mutated the bone marrow, indicating hematologic tox-
viral particles and, as a result, on HIV-infected icity (28). There was no difference in platelet
cells. In this regard RIT has certain advantages counts between RIT-treated and control mice,
over immunotoxins to eradicate infected cells. consistent with no significant acute hemato-
In immunotoxins a mAb is conjugated to logic toxicity.
immunogenic toxin and thus can elicit an Subsequently 213Bi-2556 was further evalu-
immune response, while in RIT no responses ated in the SCID mouse model described by
to radiolabeled human mAbs have been ob- Mosier (26). The Mosier model involves i.p.
served. RIT is a highly versatile modality due to implantation of hPBMCs into SCID mice,
the availability of the radionuclides with resulting in activation of T cells, thus providing
various emissions and decay schemes. Recently a cellular population easily infected by HIV
we identified a human mAb 2556 as a superb and mimicking the widespread lymphocyte
reagent for the development of an RIT-based activation observed in chronic HIV infection.
HIV elimination strategy (25). mAb 2556 is a HIV infection of hu-PBL SCID mice led to the
human mAb to a conserved domain of HIV loss of CD4+ T cells, as also seen as a result of
gp41 glycoprotein and was able to outperform HIV infection in human hosts. The Mosier
the endogenous antibodies in HIV-positive model has been broadly utilized in evaluating
serum for binding to gp41. This latter quality the efficacy of antiviral drugs (29, 30). Similar
is very important because it means that the to the splenic model, RIT led to a several log
antibody target site is not likely to be obscured reduction in viral load in groups treated with
by endogenous antibodies produced by in- 25, 50, and 100 Ci 213Bi-2556 relative to
fected individuals. controls (P < 0.05). These results are encour-
To investigate the feasibility of killing HIV- aging for further development of RIT as a
infected cells with radiolabeled 2556 mAb in backbone strategy for HIV eradication. We are
vivo we used two HIV mouse modelsa currently planning a pilot clinical trial of RIT
splenic model (22) and the Mosier model in HIV-infected patients on HAART.
(26). As in the previous study (22), human
mAb 1418 (IgG1) to parvovirus B19 (27) was
used as an irrelevant isotype-matched control. CONCLUSIONS
As in our previous study, HIV-infected
hPBMCs were given intrasplenically to SCID The foreign nature of microbial cells results in
mice. After 1.5 h, these mice were given a single their display of antigens that are not found
injection of 213Bi-2556 or control mAbs. The anywhere in the human body, making RIT for
radioactivity doses were 50, 100, and 200 Ci infectious diseases more specific than RIT for
per animal (1 mg of mAb 2556/kg body weight). cancer, since tumor-associated antigens are
Three days following treatment, the mice were also sometimes found on normal tissues. As a
sacrificed, their spleens were harvested, and consequence, the specificity of RIT for infec-
the viral load was determined by real-time PCR tions at least theoretically should be much
for HIV-1 DNA. The results demonstrated that more pronounced than in cancer, given higher
213
Bi-2556 killed HIV-infected hPBMCs much selectivity and specificity for target cells. This
more effectively than isotype-matching control exquisite specificity will lead to precise tar-
mAb armed with the same amounts of radio- geting, which in turn should translate into a
activity or cold mAb 2556 (Fig. 4b). To highly efficacious treatment not accompanied
408 DADACHOVA AND CASADEVALL

by side effects. Additionally, the technology for safety and pharmacokinetics of murine-derived
linking radionuclides to mAbs is well estab- anticryptococcal antibody 18B7 in subjects with
treated cryptococcal meningitis. Antimicrob
lished, so the lessons learned in the develop- Agents Chemother 49:952958.
ment of RIT in oncology could be readily 4. Rhodes JC, Wicker LS, Urba WJ. 1980. Genetic
applied to infectious diseases. Also, U.S. control of susceptibility to Cryptococcus neo-
hospitals that are now regularly using RIT to formans in mice. Infect Immun 29:494499.
treat cancer patients are fully equipped for 5. Jiang Z, Bryan RA, Morgenstern A,
Bruchertseifer F, Casadevall A, Dadachova E.
initiating infectious disease RIT since the two
2012. Treatment of early and established Crypto-
use the same approach and differ only in the coccus neoformans infection with radiolabeled
type of antibody used. An added bonus that antibodies in immunocompetent mice. Antimicrob
would occur with some isotopes is the possi- Agents Chemother 56:552554.
bility for imaging patients receiving RIT to 6. Pai MP, Sakoglu U, Peterson SL, Lyons CR,
Sood R. 2009. Characterization of BBB perme-
ascertain the targeting of radiolabeled mAbs to
ability in a preclinical model of cryptococcal
tissue and the anatomical extent of infection. meningoencephalitis using magnetic resonance
We believe a combination of need together imaging. J Cereb Blood Flow Metab 29:545553.
with the presence of a mature technology 7. Bryan RA, Jiang Z, Howell RC, Morgenstern A,
means that the time is ripe for deploying RIT Bruchertseifer F, Casadevall A, Dadachova E.
into the clinic to combat infectious diseases. 2010. Radioimmunotherapy is more effective than
antifungal treatment in experimental cryptococ-
cal infection. J Infect Dis 202:633637.
ACKNOWLEDGMENTS 8. Clemons KV, Stevens DA. 1998. Comparison of
fungizone, Amphotec, AmBisome, and Abelcet for
E. Dadachova was supported by the National treatment of systemic murine cryptococcosis.
Institute of Allergy and Infectious Disease Antimicrob Agents Chemother 42:899902.
9. Kakeya H, Miyazaki Y, Senda H, Senda H,
(NIAID) grant AI60507 and by the Bill and
Kobayashi T, Seki M, Izumikawa K, Yamamoto
Melinda Gates Foundation Grand Exploration Y, Yanagihara K, Tashiro T, Kohno S. 2008.
Grant. A. Casadevall was supported by NIAID Efficacy of SPK-843, a novel polyene antifungal, in
grants AI033142 and AI033774. a murine model of systemic cryptococcosis.
Conflicts of interest: We declare no conflicts. Antimicrob Agents Chemother 52:18711872.
10. Bicanic T, Muzoora C, Brouwer AE, Brouwer
AE, Meintjes G, Longley N, Taseera K, Rebe K,
CITATION Loyse A, Jarvis J, Bekker LG, Wood R,
Limmathurotsakul D, Chierakul W, Stepniewska
Dadachova E, Casadevall A. 2014. Radio- K, White NJ, Jaffar S, Harrison TS. 2009.
labeled antibodies for therapy of infectious Independent association between rate of clearance
diseases. Microbiol Spectrum 2(6):AID-0023- of infection and clinical outcome of HIV-associated
cryptococcal meningitis: analysis of a combined
2014. cohort of 262 patients. Clin Infect Dis 49:702
709.
11. Bryan RA, Jiang Z, Huang X, Morgenstern A,
REFERENCES
Bruchertseifer F, Sellers R, Casadevall A,
1. Dadachova E, Nakouzi A, Bryan R, Casadevall A. Dadachova E. 2009. Radioimmunotherapy is
2003. Ionizing radiation delivered by specific effective against a high infection burden of
antibody is therapeutic against a fungal infection. Cryptococcus neoformans in mice and does not
Proc Natl Acad Sci USA 100:1094210947. select for radiation-resistant phenotypes in cryp-
2. Park BJ, Wannemuehler KA, Marston BJ, tococcal cells. Antimicrob Agents Chemother 53:
Govender N, Pappas PG, Chiller TM. 2009. 16791682.
Estimation of the current global burden of crypto- 12. Bryan RA, Huang X, Morgenstern A,
coccal meningitis among persons living with Bruchertseifer F, Casadevall A, Dadachova E.
HIV/AIDS. AIDS 23:525530. 2008. Radio-fungicidal effects of external gamma
3. Larsen RA, Pappas PG, Perfect J, Aberg JA, radiation and antibody-targeted beta and alpha
Casadevall A, Cloud GA, James R, Filler S, radiation on Cryptococcus neoformans. Antimicrob
Dismukes WE. 2005. Phase I evaluation of the Agents Chemother 52:22322235.
CHAPTER 23 Radiolabeled Antibodies for Therapy of ID 409

13. Torosantucci A, Chiani P, Bromuro C, De 21. Rivera J, Morgenstern A, Bruchertseifer F,


Bernardis F, Palma AS, Liu Y, Mignogna G, Kearney JF, Turnbough CL Jr, Dadachova E,
Maras B, Colone M, Stringaro A, Zamboni S, Casadevall A. 2014. Microbicidal power of alpha
Feizi T, Cassone A. 2009. Protection by anti-beta- radiation in sterilizing germinating Bacillus
glucan antibodies is associated with restricted anthracis spores. Antimicrob Agents Chemother
beta-1,3 glucan binding specificity and inhibition 58:18131815.
of fungal growth and adherence. PLoS One 4: 22. Dadachova E, Patel MC, Toussi S, Apostolidis C,
e5392. doi:10.1371/journal.pone.0005392. Morgenstern A, Brechbiel MW, Gorny MK,
14. Rachini A, Pietrella D, Lupo P, Torosantucci A, Zolla-Pazner S, Casadevall A, Goldstein H.
Chiani P, Bromuro C, Proietti C, Bistoni F, 2006. Targeted killing of virally infected cells by
Cassone A, Vecchiarelli A. 2007. An anti-beta- radiolabeled antibodies to viral proteins. PLoS
glucan monoclonal antibody inhibits growth and Med 3:e427. doi:10.1371/journal.pmed.0030427.
capsule formation of Cryptococcus neoformans in 23. Ho DD, Moudgil T, Alam M. 1989. Quantitation of
vitro and exerts therapeutic, anticryptococcal human immunodeficiency virus type 1 in the blood
activity in vivo. Infect Immun 75:50855094. of infected persons. N Engl J Med 321:16211625.
15. Torosantucci A, Bromuro C, Chiani P, De 24. Casadevall A, Goldstein H, Dadachova E. 2007.
Bernardis F, Berti F, Galli C, Norelli F, Bellucci Targeting viruses-harboring host cells with
C, Polonelli L, Costantino P, Rappuoli R, radiolabeled antibodies. Expert Opin Biol Ther
Cassone A. 2005. A novel glyco-conjugate vaccine 7:595597.
against fungal pathogens. J Exp Med 202:597606. 25. Dadachova E, Kitchen SG, Bristol G, Baldwin
16. Guimaraes AJ, Frases S, Gomez FJ, Zancope- GC, Revskaya E, Empig C, Thornton GB, Gorny
Oliveira RM, Nosanchuk JD. 2009. Monoclonal MK, Zolla-Pazner S, Casadevall A. 2012. Pre-
antibodies to heat shock protein 60 alter the clinical evaluation of a 213Bi-labeled 2556 anti-
pathogenesis of Histoplasma capsulatum. Infect body to HIV-1 gp41 glycoprotein in HIV-1 mouse
Immun 77:13571367. models as a reagent for HIV eradication. PLoS One
17. Nosanchuk JD, Casadevall A. 2006. Impact of 7:e31866. doi:10.1371/journal.pone.0031866.
melanin on microbial virulence and clinical 26. Mosier DE. 1996. Viral pathogenesis in hu-PBL-
resistance to antimicrobial compounds. Anti- SCID mice. Sem Immunol 8:255262.
microb Agents Chemother 50:35193528. 27. Gigler A, Dorsch S, Hemauer A, Williams C, Kim
18. Bryan RA, Guimaraes AJ, Hopcraft S, Jiang Z, S. 1999. Generation of neutralizing human mono-
Bonilla K, Morgenstern A, Bruchertseifer F, Del clonal antibodies against parvovirus B19 proteins.
Poeta M, Torosantucci A, Cassone A, J Virol 73:19741979.
Nosanchuk JD, Casadevall A, Dadachova E. 28. Milenic DE, Brady ED, Brechbiel MW. 2004.
2012. Towards developing a universal treatment Antibody-targeted radiation cancer therapy. Nat
for fungal disease using radioimmunotherapy Rev Drug Discov 3:488499.
targeting common fungal antigens. Myco- 29. Uckun FM, Rajamohan F, Pendergrass S, Ozer
pathologia 73:463471. Z, Waurzyniak B, Mao C. 2003. Structure-based
19. Dadachova E, Burns T, Bryan RA, Apostolidis C, design and engineering of a nontoxic recombinant
Brechbiel MW, Nosanchuk JD, Casadevall A, pokeweed antiviral protein with potent-anti-
Pirofski L. 2004. Feasibility of radioimmuno- human immunodeficiency virus activity. Anti-
therapy of experimental pneumococcal infection. microb Agents Chemother 47:10521061.
Antimicrob Agents Chemother 48:16241629. 30. Uckun FM, Qazi S, Pendergrass S, Lisowski
20. Rivera J, Nakouzi AS, Morgenstern A, E, Waurzyniak B, Chen CL, Venkatachalam
Bruchertseifer F, Dadachova E, Casadevall A. TK. 2002. In vivo toxicity, pharmacokinetics,
2009. Radiolabeled antibodies to Bacillus and anti-human immunodeficiency virus activity
anthracis toxins are bactericidal and partially of stavudine-5-(p-bromophenyl methoxyalaninyl
therapeutic in experimental murine anthrax. phosphate) stampidine in mice. Antimicrob
Antimicrob Agents Chemother 53:48604868. Agents Chemother 46:34283436.
ALTERNATE SYSTEMS
FOR EXPRESSION
Plant-Derived Monoclonal
Antibodies for Prevention and
Treatment of Infectious Disease

ANDREW HIATT,1 KEVIN J. WHALEY,1 and LARRY ZEITLIN1


24
INTRODUCTION

Production and evaluation of monoclonal antibodies (MAbs) produced in plants


to combat infectious diseases (IDs) has been ongoing for almost 20 years (1). With
the recent FDA approval of the first plant-derived biologic (2), development of
rapid manufacturing technology (3), and the capability of producing MAbs with
homogenous mammalian glycosylation (4), a wave of infectious disease (and
other) MAbs that are plant-derived are expected to enter clinical trials in the
next several years. This review is intended to summarize the results of research
on plant-derived MAbs to infectious pathogens that have completed animal or
clinical studies.

PRODUCTION OF MAbs IN PLANTS

Rationale for Manufacturing ID MAbs in Plants


There are a number of advantageous characteristics of plant systems for
production of anti-infective antibodies. One in particular has to do with the ease
with which complex, multicomponent isotypes can be assembled (1, 5). The

1
Mapp Biopharmaceutical, Inc., San Diego, CA 92121.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0004-2012

413
414 HIATT ET AL.

most structurally complex type of antibody, a plant transformation technology that was
secretory immunoglobulin A (sIgA) consist- almost entirely based on regeneration of
ing of four different chains, can be assembled plants from infected plant cells carrying the
in plants to produce functional, protease- antibody-encoding cDNAs (5). Infection was
resistant antibody molecules appropriate for accomplished using engineered Agrobac-
use in environments where other types of terium that could selectively deliver the
antibodies are not found (e.g., the gastroin- immunoglobulin-encoding DNAs to the plant
testinal tract) (1). Since mucosal surfaces are cell nucleus (9). This process of creating
often the site of entry of infectious pathogens fertile transgenic plants suffered from the
(6), the technology that can provide an significant period of time required for regen-
appropriate type of antibody isotype for that eration (1 year) but did benefit from the
environment could be uniquely suited for ability to cross-pollinate the resulting plants to
combating infectious diseases. Moreover, the derive progeny that produced a desired
use of antibodies at mucosal surfaces empha- combination of immunoglobulin chains (1, 5).
sizes preventive strategies that would deny In the first set of transgenic experiments,
entry to pathogens, rather than postexposure plants expressing IgG() or Ig() chains were
or therapeutic strategies. Preventive applica- cross-pollinated to produce progeny ex-
tions of antibodies inherently require a much pressing assembled antibody (5). These IgGs
larger scale of production due to a require- possessed the same specificity when com-
ment for repeated dosing. Because preventive pared to the original mammalian cell-derived
strategies ordinarily involve larger popula- MAb. In subsequent experiments, the IgG
tions, an appropriate unit cost for a preventa- constant region was replaced with a hybrid
tive antibody can be achieved at a sufficiently constant region consisting of both IgG and IgA
large production scale (7, 8). The ability to components (1). When these hybrid heavy
scale up production without incurring exces- chain plants were crossed with plants ex-
sive capital requirements or a change in pro- pressing kappa, J chain, and secretory com-
duction methods is a hallmark of production ponent, an assembled sIgA was produced.
in plants (7). Microscopic examination of antibody accu-
mulation showed that specific cell types,
predominantly bundle sheath cells, were
responsible for the relatively high level of
Transgenic Technologies for
sIgA accumulation in these plants.
Plant-Based MAb Production
Overall, the results demonstrated that a
The first technology that was employed to variety of immunoglobulin isotypes could be
express an antibody in a plant involved the expressed in plants to yield complex, func-
transfer of recombinant selectable vectors tional antibodies and that multiple immuno-
into small pieces of leaf (leaf discs) (9) by globulin chains could be assembled into
use of Agrobacterium tumefaciens infection as antibodies by stepwise cross-pollination of
the transformation vehicle (1, 5). The early plants containing immunoglobulin genes.
results of plant expression of a MAb to an Moreover, the use of a hybrid constant region
infectious pathogen (1) demonstrated a num- could result in assembly and high-level pro-
ber of principles about the variety of MAb duction of a class of antibody that is not
isotypes that can be produced in plants as ordinarily observed from mammalian cell
well as the characteristics and localization of culture. Perhaps due to their inherent bio-
those antibodies within the plant leaf (1). chemical stability, the hybrid sIgA accumulat-
Early experiments exploring the capabilities ed in these plants to levels that are not
of plants for expressing, assembling, and ordinarily observed in transgenics (5% of
secreting complex immunoglobulins used a extractable protein).
CHAPTER 24 Plant-Derived MAbs for Prevention and Treatment of ID 415

Transient Technologies for Plant-Based tions are introduced into candidate molecules
MAb Production and evaluated for changes in efficacy, the
shortened time frame to multi-gram-level
Recent plant-based manufacturing techniques
production may accelerate this process greatly.
have been aimed at deriving a higher level of
expression and, importantly, a short time frame
to generate MAb. One commercially viable Technologies for Controlling MAb
technique, referred to as magnifection, N Glycosylation
involves transient expression of the MAb
Recently, transgenic technology has been used
wherein the infection of the cell is accom-
to generate plants that yield MAbs with
plished after introducing several pro-vectors
homogenous N glycosylation patterns (4)
into Agrobacterium tumefaciens that can de-
(Fig. 2). Specifically, using RNA interference
liver noncompeting viral components (derived
technology, Nicotiana benthamiana plants
from tobacco mosaic virus and potato virus X)
have been generated that have dramatically
as well as the MAb genes to plant cells (3, 10). In
reduced xylosyl or fucosyl transferase enzy-
this sense, Agrobacterium is the vehicle for
matic activities (DXF) (11). Antibodies ex-
primary infection of the plant, whereas the
pressed in this transgenic plant contain a
ultimately recombined, functional viral repli-
single dominant N-glycan species consisting
con provides cell-to-cell spread, amplification,
of biantennary N-glycans with terminal N-
and high expression (Fig. 1). The technology
acetylglucosamine on both branches (GnGn)
appears to be the most rapid path from genes to
that shows significantly enhanced binding to
full-length, assembled MAb, i.e., 14 days from
FcRIIIa, as has been observed with other
gene delivery to purified MAb (10). The
nonfucosylated MAbs derived from other
manufacturing technology can easily accom-
sources (11). A plant-derived anti-human im-
modate hundreds of grams per month for
munodeficiency virus (anti-HIV) antibody
preclinical and clinical studies as well as
produced in these plants was the first example
commercial scale (7, 8). Because antibody
of using glycoengineering to improve the
drug development has evolved into a highly
antiviral activity of a MAb (11).
iterative process where sequential modifica-
Introduction of the -1,4-galactosyl trans-
ferase gene into this DXF plant background
resulted in a fully galactosylated and relatively
homogeneous N-glycosylation. These MAbs
(11) consisted of two broadly neutralizing anti-
HIV MAbs (2G12, 4E10) and displayed im-
proved virus neutralization potency when
compared with other glycoforms produced in
plants and in CHO cells.
The final sugar addition in many mamma-
lian glycans is sialylation (12). Terminal -2,6-
sialic acid is an important mediator of diverse
biological activities. In particular, sialylation
appears to play a crucial role in the efficacy of
intravenous immunoglobulin for autoimmune
diseases (13, 14, 15). Plants do not make sialic
FIGURE 1 Fluorescent immunocytochemistry of a N.
acid; however, the entire biosynthetic pathway
benthamiana leaf 7 days after magnifection. Tissue
was stained with Cy3-labeled anti-human IgG. Image for antibody sialylation has been incorporated
courtesy of Jeffrey Pudney, Boston University Medical into DXF Nicotiana plants (12). IgG antibodies
College. doi:10.1128/microbiolspec.AID-0004-2012.f1 expressed in the sialo-engineered plants have a
416 HIATT ET AL.

FIGURE 2 N glycosylation of a humanized IgG1 produced in XF N. benthamiana and mammalian cell


culture (NS0 cells) as determined by 2-AA glycan analysis. doi:10.1128/microbiolspec.AID-0004-2012.f2

significant percentage of molecules with -2,6- an initial trial, patients received 9 days of
sialic acid termini that can exceed the percent- chlorhexidine treatment to eliminate Strepto-
age observed in intravenous immunoglobulin. coccus mutans from their oral cavities followed
In sum, the glycoengineering that has been by two treatments per week for 3 weeks with
accomplished to date has provided a variety of Guys 13 sIgA/G, saline, or control antibody.
glycoforms with either enhanced biological Patients treated with the plant-derived Guys
activity (e.g., antiviral) or fully human glycans. 13 had no recolonization with S. mutans (n = 4),
whereas patients treated with saline (n = 4) or
control antibody (n = 3) were all recolonized.
PLANT-DERIVED MAbs AGAINST All treatments in this study were performed in
ID TARGETS a dental clinic. In a subsequent trial testing
home treatment, 56 eligible adults had 9 days
To date, a variety of plant-produced MAbs of treatment with chlorhexidine and were
have been described. The focus here is on randomized equally to a group receiving 0, 2,
MAbs that have been tested for efficacy in 4, or 6 topical applications of plant-produced
animals or humans (Table 1). Guys 13 followed by 6, 4, 2, or 0 applications of
placebo, respectively, over a 3-week period.
After 6 months, there were no significant
Dental Caries MAbs
differences in S. mutans levels by number of
Clinical evaluation of the plant-produced applications, relative to placebo. No adverse
Guys 13 sIgA/G has been performed (16). In effects were observed during the study, and it
CHAPTER 24 Plant-Derived MAbs for Prevention and Treatment of ID 417

TABLE 1 In vivo studies with plant-derived MAbs against ID targets


Pathogen MAb descriptor Expression host Test model Efcacy P Comments (reference)a

Anthrax Receptor- N. benthamiana Rabbit 100% <0.05 2 mg/kg delivered 1 h after


human Fc 100 LD50 inhalation challenge
fusion (69)
Aglycosylated N. benthamiana Mouse 100% <0.01 8 mg/kg delivered to mice
human IgG1 Macaque 100% <0.05 prior to i.p. challenge with
20 LD50; 5 mg/kg delivered to
macaques prior to 500 LD50
aerosol challenge (68)
C. perfringens Chimeric IgG1 N. benthamiana Mouse 100% <0.001 0.8 mg/kg delivered prior to
ETX i.v. challenge (Uzal and Zeitlin,
unpublished)
Ebola virus Humanized N. benthamiana Mouse 100% <0.01 2 mg/kg delivered prior to i.p.
IgG1 challenge with 30,000 LD50
(21)
3-MAb IgG1 N. benthamiana Macaque 67 <0.05 50 mg/kg delivered 1 h to
cocktail 100% 2 days after 1,000 LD50 i.m.
challenge (26)
HSV Humanized Soybean Mouse 100% <0.001 0.5 mg/ml delivered vaginally
IgG1 prior to 10 LD50 vaginal
challenge (27)
Rabies Human IgG N. tabacum Hamster 100% <0.01 3 IU of MAb 4 h after
intracerebral challenge with
3 105 LD50 (36)
RSV Humanized N. benthamiana Cotton rat >102b <0.001 5 mg/kg delivered prior to i.n.
IgG1 challenge of 2 104 PFU
(Zeitlin et al., submitted)
SEB Chimeric IgG1 N. benthamiana Mouse 100% <0.001 0.4 mg/kg delivered prior to
i.p. challenge with 5 LD50
(Roy and Zeitlin, unpublished)
S. mutans Murine SIgA/G N. tabacum Human 100% <0.05 25 mg/kg prevented natural
recolonization over the 3-mo
course of study (18)
WNV Humanized N. benthamiana Mouse 8090% <0.001 15 mg/kg delivered 2 days
IgG1 post-footpad challenge with
510 LD50 (54)
a
LD50, 50% lethal dose; i.p., intraperitoneal; i.v., intravenous; i.m., intramuscular; i.n., intranasal.
b
Reduction in viral lung titer.

was concluded that plant-produced Guys 13 is clinical trial (www.planetbiotechnology.com/


safe but not effective at the frequency, con- products.html) (20).
centration, and number of applications used in
that study. However, subsequent clinical eval-
Ebola Virus MAbs
uation did define the effective parameters of
administration. CaroRx, the commercial name Recent outbreaks in central Africa and con-
for the plant-produced Guys 13 anti-caries cerns about weaponization highlight the lack
MAb product, is intended for regular oral of any vaccine or treatment for Ebola virus
topical preventative administration. New clin- infection. A recent study focused on the role of
ical trials have indicated that this treatment antibody glycosylation in the development of
can effectively eliminate decay-causing bacte- an immunoprotectant for Ebola virus revealed
ria for up to 2 years (16, 17, 18, 19). CaroRx has potential advantages of plant production of
demonstrated efficacy and safety in a phase 2 MAbs against infectious pathogens (21). An
418 HIATT ET AL.

anti-Ebola glycoprotein (GP) MAb was pro- plex virus (anti-HSV) glycoprotein B antibody
duced in XF Nicotiana plants, resulting in made in soybeans compared to the same MAb
MAb with a highly homogenous distribution expressed in mammalian cell culture of Sp2/0
of N-glycoforms (90% GnGn) and lacking cells (27). The comparative assays included
potentially immunogenic plant-specific 1,2 diffusion and stability in mucus as well as
xylose and core 1,3 fucose. The MAb, h-13F6, prevention of vaginal transmission of genital
recognizes the heavily glycosylated mucin-like herpes in the mouse. No significant difference
domain of GP. h-13F6 does not neutralize in was observed in these assays, and vaginal
vitro, as assessed by inhibition of plaque delivery of 10 g of either MAb provided
formation, even in the presence of comple- 100% protection against vaginal challenge
ment (21), suggesting an important role for with HSV type 2 (27). A microbicide con-
Fc-mediated effector functions in in vivo pro- taining a cocktail of HSV and HIV MAbs is
tection. The plant-derived h-13F6 was com- currently being evaluated for efficacy in the
pared with h-13F6 produced in CHO cells in a simian HIV nonhuman primate model and is
lethal mouse Ebola virus challenge model. The expected to enter a clinical safety trial in 2014
plant-derived h-13F6 carrying N-glycans (K. Whaley and F. Villinger, personal commu-
lacking fucose showed threefold-superior nication).
potency to h-13F6 with typical CHO glyco- A number of broadly neutralizing HIV
sylation patterns (21). Elimination of core MAbs have been produced in plants and
fucose has been shown to dramatically improve evaluated on the basis of binding ability as
antibody-dependent cellular cytotoxicity (ADCC) well as structure (28, 29, 30, 31). For example,
activity in vitro and in vivo through improved one of the broadly neutralizing MAbs, 2G12,
affinity for FcRIII (22, 23, 24, 25). Indeed, has been produced in transgenic maize plants
affinity analyses of the plant- and CHO- (29), and the HIV neutralization capability of
derived h-13F6 confirmed that elimination the antibody is equal to or superior to that of
of core fucose from h-13F6 resulted in an the same antibody produced in CHO cells.
improved affinity for FcgRIII. In total, these 2G12 has also been transiently produced in the
results suggested that ADCC is an important DXF Nicotiana line and was found to contain
mechanism of protection by this MAb. Further a relatively homogeneous N-glycan species
studies have now been performed showing without detectable xylose or -1,3-fucose
that a cocktail of 3 Nicotiana-derived anti- residues (4). In general, plant-derived 2G12
Ebola GP MAbs (including h-13F6) can protect was indistinguishable from CHO-derived 2G12
rhesus macaques from lethal challenge: 100% with respect to electrophoretic properties as
survival was observed when the MAbs were well as functional properties (i.e., antigen
administered an hour after challenge and binding and HIV neutralization activity). In
67% survival when delivered 1 or 2 days some cases, plant-produced HIV MAbs appear
postchallenge (26). This study demonstrated to have enhanced potency compared to CHO-
a greater therapeutic window as well as produced MAbs. Fully galactosylated 4E10
reduced viral burden and level of disease (another broadly neutralizing MAb) and 2G12
compared to other therapeutics in develop- were reported to be severalfold higher in
ment (26). neutralization potency than CHO-produced
MAbs (11). In addition, sialylated 2G12 exhibits
in vitro HIV neutralization potency similar to
HSV and HIV MAbs: Preventing Sexual
that of other glycoforms derived from plants
Transmission
and CHO cells (12). Other plant expression
The earliest in vivo comparison study of a systems have been used to produce these
plant-produced and a cell culture-produced MAbs as well: 4E10 has been produced in a
MAb involved a humanized anti-herpes sim- transgenic tobacco rhizosection system (31),
CHAPTER 24 Plant-Derived MAbs for Prevention and Treatment of ID 419

and a fusion protein of the broadly neutral- virus in hamsters, indicating that differences
izing HIV MAb b12 and cyanovirin produced in N-glycosylation do not affect the efficacy
in transgenic tobacco increased HIV potency of the antibody in this model (38).
compared to b12 or cyanovirin alone (32).
RSV MAbs
Rabies Virus MAbs
Synagis is the only MAb approved by the FDA
Since more than 10 million people annually for an ID indication and currently generates
receive rabies virus postexposure prophylaxis sales of greater than $1 billion/year. Although
in the form of equine antirabies immunoglob- Synagis is an effective drug for respiratory
ulin or human antirabies immunoglobulin syncytial virus (RSV) immunoprophylaxis in
(HRIG), together with rabies vaccine (33), at-risk neonates, ongoing analyses on the
there is a worldwide shortage of these immu- pharmacoeconomics suggest that Synagis
noglobulins. The limited supply and the addi- may not be cost-effective in actual use, nor
tional risk of adverse reactions associated with even when its use is restricted very tightly to its
equine antirabies immunoglobulin have labeled population (39, 40, 41, 42, 43, 44). A
spurred the search for new MAbs. In addition, less expensive RSV MAb could improve the
the high cost of HRIG has been a significant pharmacoeconomics in neonates and open up
impediment in providing postexposure pro- additional markets; RSV is a significant cause
phylaxis against rabies (34). To address these of morbidity and mortality in the elderly as
limitations, transgenic plants have been used well as in hematopoietic stem cell transplant
as an efficient production system for the patients. Several RSV-neutralizing MAbs have
expression of functional therapeutic antirabies been produced in Nicotiana using magni-
MAbs (35). In the first report of systemic fection and have been tested in the cotton rat
administration of a plant-derived MAb to model of protective efficacy (80). Synagis and
provide immunoprotection, a human anti- the identical amino acid sequence expressed in
rabies MAb was purified from transgenic Nicotiana were superior to other candidate
tobacco plants and characterized structurally MAbs and were essentially indistinguishable
and for its effectiveness in vivo (36). The plant- in terms of affinity, neutralization, and in vivo
derived MAb was compared with an equivalent protection, providing greater than 100-fold
human MAb expressed in murine-human reduction in viral lung titer when dosed at
hybridoma cell lines (37, 38) and/or commer- 5 mg/kg of body weight. The data also
cial HRIG for neutralization activity, protein suggested that the Nicotiana-derived MAb
stability, N-glycan processing, and the efficacy was more potent therapeutically than Synagis,
of rabies virus postexposure prophylaxis in raising the possibility that enhanced ADCC
exposed animals. The plant-derived MAb was conferred by the DXF Nicotiana N-glycans
as effective at neutralizing the activity of the may be responsible for improved efficacy in an
rabies virus as the mammalian-derived anti- established RSV infection (80).
body or HRIG. Due to the incorporation of an
endoplasmic retention signal on the MAb
WNV MAbs
heavy chain (to increase accumulation), the
plant-derived MAb contained mainly oligo- West Nile virus (WNV) has in the past been
mannose type N-glycans (90%) and had no responsible for outbreaks of mild illness
potentially antigenic (1,3)-linked fucose in regions of Africa, the Middle East, Asia,
residues. The plant-derived MAb had a and Australia. In the 1990s, however, the
shorter serum half-life than mammalian cell- epidemiology of infection changed and new
derived MAb but was as efficient as HRIG outbreaks in Eastern Europe were associ-
for postexposure prophylaxis against rabies ated with higher rates of severe neurological
420 HIATT ET AL.

disease (45). In 1999, WNV entered North anthrax lethal toxin activity in vitro and
America, and since then, WNV has spread to protected mice from lethal intraperitoneal
all 48 of the lower United States as well as to challenge with spores. The nonglycosylated
parts of Canada, Mexico, the Caribbean, and MAb possessed superior efficacy (100% pro-
South America. Because of the increased tection) against an aerosol challenge compared
range, the number of human cases has contin- with the glycosylated form (40%) after a single
ued to rise: in the United States between 1999 5-mg/kg of body weight intravenous admin-
and 2008, 28,961 cases that reached clinical istration in nonhuman primates. This was
attention were confirmed and associated with attributed to the shortened half-life observed
1,131 deaths (46). The lack of effective and with the glycosylated MAb. In general, the data
specific antiviral treatment for infection by demonstrate that the absence of N-linked
WNV or other flaviviruses (47) has prompted glycans does not affect the ability of this
the search for highly specific therapies, includ- MAb to protect animals against a lethal
ing MAbs. Although small-molecule antiviral spore challenge. The results also demonstrate
agents have been described with activity that plant production is an appropriate tech-
against WNV in vitro, efficacy in vivo has nology for generating nonglycosylated MAbs
been limited (48). A humanized murine MAb and could be useful in meeting the produc-
(HuE16) that binds to a highly conserved tion challenge involved in the treatment of
epitope on domain III (DIII) of the WNV inhalation anthrax in humans. An additional
envelope (E) protein has been described (49). plant-derived immunotherapeutic utilized a
This MAb blocks viral fusion at low concen- recombinant fusion protein comprised of a
trations (50, 51) and has therapeutic activity in fusion of a human receptor for anthrax toxins
rodents even after WNV has entered the (capillary morphogenesis protein 2 [CMG2])
central nervous system (52, 53). Magnifec- and the Fc of human IgG1, for long-circulating
tion has been used to produce high levels of the half-life and immune effector cell interaction
Hu-E16 MAb, which retained neutralizing (69). CMG2-Fc, purified from tobacco plants,
activity and significant pre- and postexposure fully protected rabbits against a lethal chal-
therapeutic activity in mice (54). The thera- lenge with Bacillus anthracis spores at a dose of
peutic activity was virtually equivalent to 2 mg/kg of body weight administered at the
that observed with mammalian cell culture- time of challenge. Treatment with CMG2-Fc
derived Hu-E16 with 80 to 90% survival when did not interfere with the development of the
delivered 2 or 4 days postchallenge (54). animals own immunity to anthrax, as treated
animals that survived an initial challenge also
survived a rechallenge 30 days later (69).
MAbs to Bacterial Toxins
Anthrax MAbs Clostridium Perfringens ETX
Anthrax toxin-specific antibodies have previ- Potential aggressors have shown interest in
ously been shown to provide passive protection the biological weapons potential of C. perfringens,
against an anthrax challenge (55, 56, 57, 58). and the UN Special Commission identified
These antibodies, generally against protective it as having been developed and potentially
antigen, protect rats, guinea pigs, rabbits, and weaponized in Iraq (70, 71). Epsilon toxin
nonhuman primates when administered with- (ETX) is the most potent of the C. perfringens
in a time frame of several hours before or after toxins, with a mouse intravenous lethal dose
the exposure to inhalation anthrax (59, 60, 61, of 100 ng per kg of body weight (72). A
62, 63, 64, 65, 66, 67, 68). A transgenic previously identified mouse MAb, 4D7, with
Nicotiana-derived nonglycosylated MAb di- potent neutralizing activity (73) was chim-
rected against protective antigen has been erized with human constant regions and
evaluated (68). This antibody neutralized produced in the magnifection system. This
CHAPTER 24 Plant-Derived MAbs for Prevention and Treatment of ID 421

MAb demonstrated 100% prophylactic and displays complement-dependent cytotoxicity in


postexposure efficacy against challenge (F. Uzal a manner that was similar to anti-HBs human
and L. Zeitlin, unpublished data). immunoglobulins used clinically. This pro-
duct is now being used in Cuba in the manu-
SEB facturing of HBsAg for active immunization.
Staphylococcal enterotoxin B (SEB) is an
extremely potent enterotoxin involved in a
large proportion of cases of toxic shock syn- CONCLUSION
drome as well as a very significant mediator of
staphylococcal food poisoning (74, 75, 76). Since the early days of plant expression of
Because of its extreme potency, current interest antibodies (5), research into the application of
in SEB relates to its potential for use as a the technology for preventing or treating
biowarfare or bioterrorism agent. There are infectious pathogens has resulted in established
currently no therapeutic options available for clinical efficacy in one case and fundamental
SEB exposure. SEB MAb production in the proof of concept in others. In addition, the
magnifection system resulted in very high levels ability of plants to readily assemble antibodies
of MAb accumulation (77). A human-mouse that are appropriate for applications involving
chimeric MAb against SEB has now been mucosal and topical surfaces derives from the
shown to provide 100% protection in mouse various ways that different antibody compo-
lethal aerosol and systemic challenge models nents can be assembled as well as the fidelity of
(C. Roy, L. Zeitlin, et al., unpublished data). the endomembrane system of plants in effi-
ciently assembling and secreting antibody mol-
ecules. Further, the ability to manufacture MAb
Plant-Produced MAbs for Purication of
with specific mammalian N-glycosylation
ID Antigens
patterns allows for customization of biological
MAbs specific for particular ID antigens have activity. Now that a plant-derived biologic has
also been produced in plants to facilitate large- found regulatory acceptance, the path to ap-
scale purification of the antigen for use as a proval of novel antibody anti-infectives derived
vaccine. For example, a recombinant antibody from plants should be straightforward. Where-
specific for the HBsAg was purified from as it remains to be seen the extent to which
transgenic Nicotiana tabacum plants to test its plant-derived antibody products will enjoy
ability to immunopurify a yeast rHBsAg to be benefit of scale, the promise of a production
putatively used as a vaccine in humans. The system that can provide novel anti-infectives at
purification system was mainly based on re- low cost to a large market continues to drive
combinant protein A expanded bed adsorption research in the field.
chromatography, which has been widely used
for MAb purification (78, 79). In this chroma-
ACKNOWLEDGMENT
tography, a strong interaction among the
protein A and Fc fragments of the antibodies We declare a conflict of interest: L.Z. and K.J.W.
is normally produced, allowing high recovery are co-owners of Mapp Biopharmaceutical,
and purity. Additionally, its main advantage Inc.
is the capacity to be introduced as a primary
recovery operation to handle unclarified
CITATION
materials, yielding a combination of clarifica-
tion, concentration, and adsorptive purifica- Hiatt A, Whaley KJ, Zeitlin L. 2014. Plant-
tion in a single step. Further preclinical derived monoclonal antibodies for prevention
evaluation has demonstrated that the MAb and treatment of infectious disease. Microbiol
readily interacts with cell surface HBsAgs and Spectrum 2(1):AID-0004-2012.
422 HIATT ET AL.

REFERENCES 12. Castilho A, Strasser R, Stadlmann J, Grass J, Jez


J, Gattinger P, Kunert R, Quendler H, Pabst M,
1. Ma JK, Hiatt A, Hein M, Vine ND, Wang F, Leonard R, Altmann F, Steinkellner H. 2010. In
Stabila P, van Dolleweerd C, Mostov K, Lehner planta protein sialylation through overexpression
T. 1995. Generation and assembly of secretory of the respective mammalian pathway. J Biol
antibodies in plants. Science 268:716719. Chem 285:1592315930.
2. Aviezer D, Brill-Almon E, Shaaltiel Y, 13. Jefferis R. 2005. Glycosylation of recombinant
Hashmueli S, Bartfeld D, Mizrachi S, Liberman antibody therapeutics. Biotechnol Prog 21:1116.
Y, Freeman A, Zimran A, Galun E. 2009. A plant- 14. Nimmerjahn F, Ravetch JV. 2008. Anti-
derived recombinant human glucocerebrosidase inflammatory actions of intravenous immuno-
enzymea preclinical and phase I investigation. globulin. Annu Rev Immunol 26:513533.
PLoS One 4:e4792e4796. 15. Anthony RM, Wermeling F, Ravetch JV. 2012.
3. Giritch A, Marillonnet S, Engler C, van Eldik G, Novel roles for the IgG Fc glycan. Ann NY Acad Sci
Botterman J, Klimyuk V, Gleba Y. 2006. Rapid 1253:170180.
high-yield expression of full-size IgG antibodies
16. Ma JK, Hikmat BY, Wycoff K, Vine ND,
in plants co-infected with noncompeting viral
Chargelegue D, Yu L, Hein MB, Lehner T.
vectors. Proc Natl Acad Sci USA 103:1470114706.
1998. Characterization of a recombinant plant
4. Strasser R, Stadlmann J, Schahs M, Stiegler G,
monoclonal secretory antibody and preventive
Quendler H, Mach L. 2008. Generation of
immunotherapy in humans. Nat Med 4:601606.
glycoengineered Nicotiana benthamiana for the
17. Ma JK, and Lehner T. 1990. Prevention of
production of monoclonal antibodies with a
colonization of Streptococcus mutans by topical
homogeneous human-like N-glycan structure.
application of monoclonal antibodies in human
Plant Biotech J 6:392402.
subjects. Arch Oral Biol 35:115122.
5. Hiatt A, Cafferkey R, Bowdish K. 1989. Produc-
18. Ma JK, Hunjan M, Smith R, Lehner T. 1989.
tion of antibodies in transgenic plants. Nature 342:
Specificity of monoclonal antibodies in local
7678.
passive immunization against Streptococcus
6. Bemark M, Boysen P, Lycke NY.2012. Induction
mutans. Clin Exp Immunol 77:331337.
of gut IgA production through T cell-dependent
and T cell-independent pathways. Ann N Y Acad 19. Weintraub JA, Hilton JF, White JM, Hoover CI,
Sci 1247:97116. Wycoff KL, Yu L, Larrick JW, Featherstone JD.
7. Pogue GP, Vojdani F, Palmer KE, Hiatt E, Hume 2005. Clinical trial of a plant-derived antibody on
S, Phelps J, Long L, Bohorova N, Kim D, Pauly re-colonization of mutans streptococci. Caries Res
M, Velasco J, Whaley K, Zeitlin L, Garger SJ, 39:241250.
White E, Bai Y, Haydon H, Bratcher B. 2010. 20. De Muynck B, Navarre C, Boutry M. 2010.
Production of pharmaceutical-grade recombinant Production of antibodies in plants: status after
aprotinin and a monoclonal antibody product twenty years. Plant Biotechnol J 8:529563.
using plant-based transient expression systems. 21. Zeitlin L, Pettitt J, Scully C, Bohorova N, Kim D,
Plant Biotechnol J 8:638654. Pauly M, Hiatt A, Ngo L, Steinkellner H, Whaley
8. Klimyuk V, Pogue G, Herz S, Butler J, Haydon KJ, Olinger GG. 2011. Enhanced potency of a
H. 2012. Production of recombinant antigens fucose-free monoclonal antibody being developed
and antibodies in Nicotiana benthamiana using as an Ebola virus immunoprotectant. Proc Natl
magnifection technology: GMP-compliant facil- Acad Sci USA 108:2069020694.
ities for small- and large-scale manufacturing. 22. Niwa R, Shoji-Hosaka E, Sakurada M, Shinkawa
Curr Top Microbiol Immunol doi:10.1007/ T, Uchida K, Nakamura K, Matsushima K, Ueda
82_2012_212. [Epub ahead of print.] R, Hanai N, Shitara K. 2004. Defucosylated
9. Horsch RB, Fraley RT, Rogers SG, Sanders PR, chimeric anti-CC chemokine receptor 4 IgG1
Lloyd A, Hoffmann N. 1984. Inheritance of with enhanced antibody-dependent cellular
functional foreign genes in plants. Science 223: cytotoxicity shows potent therapeutic activity to
496498. T-cell leukemia and lymphoma. Cancer Res 64:
10. Hiatt A, and Pauly M. 2006. Monoclonal anti- 21272133.
bodies from plants: a new speed record. Proc Natl 23. Cox KM, Sterling JD, Regan JT, Gasdaska JR,
Acad Sci USA 103:1464514646. Frantz KK, Peele CG, Black A, Passmore D,
11. Strasser R, Castilho A, Stadlmann J, Kunert R, Moldovan-Loomis C, Srinivasan M, Cuison S,
Quendler H, Gattinger P. 2009. Improved virus Cardarelli PM, Dickey LF. 2006. Glycan optimi-
neutralization by plant-produced anti-HIV anti- zation of a human monoclonal antibody in the
bodies with a homogeneous beta1,4-galactosylated aquatic plant Lemna minor. Nat Biotechnol 24:
N-glycan profile. J Biol Chem 284:2047920485. 15911597.
CHAPTER 24 Plant-Derived MAbs for Prevention and Treatment of ID 423

24. Umaa P, Jean-Mairet J, Moudry R, Amstutz bodies, biopharmaceuticals and edible vaccines in
H, Bailey JE. 1999. Engineered glycoforms of an plants. Trends Plant Sci 6:219226.
antineuroblastoma IgG1 with optimized antibody- 36. Ko K, Tekoah Y, Rudd PM, Harvey DJ, Dwek
dependent cellular cytotoxic activity. Nat Bio- RA, Spitsin S, Hanlon CA, Rupprecht C,
technol 17:176180. Dietzschold B, Golovkin M, Koprowski H.
25. Junttila TT, Parsons K, Olsson C, Lu Y, Xin Y, 2003. Function and glycosylation of plant-derived
Theriault J, Crocker L, Pabonan O, Baginski T, antiviral monoclonal antibody. Proc Natl Acad Sci
Meng G, Totpal K, Kelley RF, Sliwkowski MX. USA 100:80138018.
2010. Superior in vivo efficacy of afucosylated 37. Dietzschold B, Gore M, Casali P, Ueki Y,
trastuzumab in the treatment of HER2-amplified Rupprecht CE, Notkins AL, Koprowski H.
breast cancer. Cancer Res 70:44814489. 1990. Biological characterization of human mono-
26. Olinger GG, Pettitt JD, Kim DH, Working C, clonal antibodies to rabies virus. J Virol 6:3087
Bohorov O, Bratcher B, Hiatt E, Hume SD, 3090.
Johnson AK, Morton J, Pauly MH, Whaley KJ, 38. Prosniak M, Faber M, Hanlon CA, Rupprecht
Lear CM, Biggins JE, Scully C, Hensley LE, CE, Hooper DC, Dietzschold B. 2003. Develop-
Zeitlin L. 2012. Delayed treatment of Ebola virus ment of a cocktail of recombinant-expressed
infection with plant-derived monoclonal antibodies human rabies virus-neutralizing monoclonal anti-
provides protection in rhesus macaques. Proc Natl bodies for postexposure prophylaxis of rabies.
Acad Sci USA doi:10.1073/pnas.1213709109. J Infect Dis 188:5356.
27. Zeitlin L, Olmsted SS, Moench TR, Co MS, 39. Joffe S, Ray GT, Escobar GJ, Black SB, Lieu TA.
Martinell BJ, Paradkar VM, Russell DR, Queen 1999. Cost-effectiveness of respiratory syncytial
C, Cone RA, Whaley KJ. 1998. A humanized virus prophylaxis among preterm infants. Pediat-
monoclonal antibody produced in transgenic rics 104(3 Pt 1):419427.
plants for immunoprotection of the vagina against 40. Lofland JH, OConnor JP, Chatterton ML,
genital herpes. Nat Biotechnol 16:13611364. Moxey ED, Paddock LE, Nash DB, Desai
28. Forthal DN, Gach JS, Landucci G, Jez J, SA. 2000. Palivizumab for respiratory syncytial
Strasser R, Kunert R, Steinkellner H. 2010. virus prophylaxis in high-risk infants: a cost-
Fc-glycosylation influences Fc receptor binding effectiveness analysis. Clin Ther 22:13571369.
and cell-mediated anti-HIV activity of mono- 41. Numa A. 2000. Outcome of respiratory syncytial
clonal antibody 2G12. J Immunol 185:68766882. virus infection and a cost-benefit analysis of
29. Rademacher T, Sack M, Arcalis E, Stadlmann J, prophylaxis. J Paediatr Child Health 36:422427.
Balzer S, Altmann F. 2008. Recombinant antibody 42. Barton LL, Grant KL, Lemen RJ. 2001. Respira-
2G12 produced in maize endosperm efficiently tory syncytial virus immune globulin: decisions
neutralizes HIV-1 and contains predominantly and costs. Pediatr Pulmonol 32:2028.
single GlcNAc N-glycans. Plant Biotech J 6:189 43. Vogel A, McKinlay M, Ashton T, Lennon D,
201. Harding J, Pinnock R, Graham D, Grimwood K,
30. Ramessar K, Rademacher T, Sack M, Pattemore P, Schousboe M. 2002. Cost-effective-
Stadlmann J, Platis D, Stiegler G. 2008. Cost- ness of palivizumab in New Zealand. J Paediatr
effective production of a vaginal protein micro- Child Health 38:352357.
bicide to prevent HIV transmission. Proc Natl 44. Roeckl-Wiedmann I, Liese JG, Grill E, Fischer
Acad Sci USA 105:37273732. B, Carr D, Belohradsky BH. 2003. Economic
31. Drake PMW, Barbi T, Sexton A, McGowan E, evaluation of possible prevention of RSV-related
Stadlmann J, Navarre C. 2009. Development of hospitalizations in premature infants in Germany.
rhizosecretion as a production system for recom- Eur J Pediatr 162:237244.
binant proteins from hydroponic cultivated to- 45. Hubalek Z, Halouzka J. 1999. West Nile fevera
bacco. FASEB J 23:35813589. reemerging mosquito-borne viral disease in
32. Sexton A, Harman S, Shattock RJ, Ma JK. 2009. Europe. Emerg Infect Dis 5:643650.
Design, expression and characterization of a mul- 46. Diamond MS. 2009. Progress on the development
tivalent, combination HIV microbicide. FASEB of therapeutics against West Nile virus. Antivir
J 23:35903600. Res 83:214227.
33. Anonymous. 2002. Rabies vaccines. Wkly Epi- 47. Furuta Y, Takahashi K, Shiraki K, Sakamoto K,
demiol Rec 77:109120. Smee DF, Barnard DL, Gowen BB, Julander JG,
34. Wilde H, Tipkong P, Khawplod P. 1999. Eco- Morrey JD. 2009. T-705 (favipiravir) and related
nomic issues in postexposure rabies treatment. J compounds: novel broad-spectrum inhibitors of
Travel Med 6:238242. RNA viral infections. Antivir Res 82:95102.
35. Daniell H, Streatfield SJ, Wycoff K. 2001. 48. Morrey JD, Taro BS, Siddharthan V, Wang H,
Medical molecular farming: production of anti- Smee DF, Christensen AJ, Furuta Y. 2008.
424 HIATT ET AL.

Efficacy of orally administered T-705 pyrazine antibody to protective antigen in rabbits and
analog on lethal West Nile virus infection in cynomolgus monkeys, abstr. 3836. Progr Abstr
rodents. Antivir Res 80:377379. 43rd Intersci Conf Antimicrob Agents Chemother,
49. Oliphant T, Engle M, Nybakken GE, Doane C, Chicago, IL.
Johnson S, Huang L, Gorlatov S, Mehlhop E, 60. Beebe LE, Babin M, Barnewall R, Zhong J, Choi
Marri A, Chung KM, Ebel GD, Kramer LD, G. 2004. Post-exposure therapeutic potential of
Fremont DH, Diamond MS. 2005. Development PAMAb in an inhalation model of anthrax in New
of a humanized monoclonal antibody with thera- Zealand White rabbits (NZW), abstr. 167G. Abstr
peutic potential against West Nile virus. Nat Med 2004 Am Soc Microbiol Biodefense Res Meet,
11:522530. Baltimore, MD.
50. Pierson TC, Xu Q, Nelson S, Oliphant T, 61. Mabry R, Rani M, Geiger R, Hubbard GB,
Nybakken GE, Fremont DH, Diamond MS. Carrion R Jr, Brasky K, Patterson JL, Georgiou
2007. The stoichiometry of antibody-mediated G, Iverson BL. 2005. Passive protection against
neutralization and enhancement of West Nile anthrax by using a high-affinity antitoxin antibody
virus infection. Cell Host Microbe 1:135145. fragment lacking an Fc region. Infect Immun
51. Thompson BS, Moesker B, Smit JM, Wilschut J, 73:83628368.
Diamond MS, Fremont DH. 2009. A therapeutic 62. Mohamed N, Clagett M, Li J, Jones S, Pincus S,
antibody against West Nile virus neutralizes DAlia G, Nardone L, Babin M, Spitalny G, Casey
infection by blocking fusion within endosomes. L. 2005. A high-affinity monoclonal antibody to
PLoS Pathog 5:e1000453. anthrax protective antigen passively protects
52. Morrey JD, Siddharthan V, Olsen AL, Roper rabbits before and after aerosolized Bacillus
GY, Wang H, Baldwin TJ, Koenig S, Johnson S, anthracis spore challenge. Infect Immun 73:795
Nordstrom JL, Diamond MS. 2006. Humanized 802.
monoclonal antibody against West Nile virus 63. Peterson JW, Comer JE, Noffsinger DM,
envelope protein administered after neuronal Wenglikowski A, Walberg KG, Chatuev BM.
infection protects against lethal encephalitis in 2006. Human monoclonal anti-protective antigen
hamsters. J Infect Dis 194:13001308. antibody completely protects rabbits and is syn-
53. Samuel MA, Wang H, Siddharthan V, Morrey ergistic with ciprofloxacin in protecting mice and
JD, Diamond MS. 2007. Axonal transport guinea pigs against inhalation anthrax. Infect
mediates West Nile virus entry into the central Immun 74:10161024.
nervous system and induces acute flaccid paraly- 64. Peterson JW, Comer JE, Baze WB, Noffsinger
sis. Proc Natl Acad Sci USA 104:1714017145. DM, Wenglikowski A, Walberg KG. 2007.
54. Lai H, Engle M, Fuchs A, Keller T, Johnson S, Human monoclonal antibody AVP-21D9 to pro-
Gorlatov S, Diamond MS, Chen Q. 2010. Mono- tective antigen reduces dissemination of the
clonal antibody produced in plants efficiently Bacillus anthracis Ames strain from the lungs in
treats West Nile virus infection in mice. Proc a rabbit model. Infect Immun 75:34143424.
Natl Acad Sci USA 107:24192424. 65. Sawada-Hirai R, Jiang I, Wang F, Sun SM,
55. Beedham RJ, Turnbull PC, Williamson ED. Nedellec R, Ruther P. 2004. Human anti-anthrax
2001. Passive transfer of protection against Bacil- protective antigen neutralizing monoclonal anti-
lus anthracis infection in a murine model. Vaccine bodies derived from donors vaccinated with
19:44094416. anthrax vaccine adsorbed. J Immune Based Ther
56. Henderson J, Bauly JM, Ashford DA, Oliver SC, Vaccines 2:5.
Hawes CR, Lazarus CM, Venis MA, Napier RM. 66. Vitale L, Blanset D, Lowy I, ONeill T, Goldstein
1997. Retention of maize auxin-binding protein in J, Little SF. 2006. Prophylaxis and therapy of
the endoplasmic reticulum: quantifying escape inhalational anthrax by a novel monoclonal anti-
and the role of auxin. Planta 202:313323. body to protective antigen that mimics vaccine-
57. Little SF, Leppla SH, Cora E. 1988. Production induced immunity. Infect Immun 74:58405847.
and characterization of monoclonal antibodies to 67. Wild MA, Kumor K, Nolan MJ, Lockman H,
the protective antigen component of Bacillus Bowdish KS. 2007. A human antibody against
anthracis toxin. Infect Immun 56:18071813. anthrax protective antigen protects rabbits from
58. Pitt ML, Little SF, Ivins BE, Fellows P, Barth J, lethal infection with aerosolized spores. Hum
Hewetson J. 2001. In vitro correlate of immunity Antibodies 16:99105.
in a rabbit model of inhalational anthrax. Vaccine 68. Mett V, Chichester JA, Stewart ML, Musiychuk
19:47684773. K, Bi H, Reifsnyder CJ, Hull AK, Albrecht MT,
59. Beebe LE, Zhong J, Clagett M, Babin M, Ou Y, Goldman S, Baillie LW, Yusibov V. 2011. A non-
Roschke V. 2003. Protection against inhalation glycosylated, plant-produced human monoclonal
anthrax induced lethality by a human monoclonal antibody against anthrax protective antigen
CHAPTER 24 Plant-Derived MAbs for Prevention and Treatment of ID 425

protects mice and non-human primates from cantly associated with non-menstrual TSS. Lancet
B. anthracis spore challenge. Hum Vaccin 7(Suppl): i:11491150.
183190. 77. Karauzum H, Chen G, Abaandou L, Mahmoudieh
69. Wycoff KL, Belle A, Deppe D, Schaefer L, M, Boroun AR, Shulenin S, Devi VS, Stavale E,
Maclean JM, Haase S, Trilling AK, Liu S, Leppla Warfield KL, Zeitlin L, Roy CJ, Sidhu SS,
SH, Geren IN, Pawlik J, Peterson JW. 2011. Aman MJ. 2012. Synthetic human monoclonal
Recombinant anthrax toxin receptor-Fc fusion antibodies toward staphylococcal enterotoxin
proteins produced in plants protect rabbits against B (SEB) protective against toxic shock syndrome.
inhalational anthrax. Antimicrob Agents Che- J Biol Chem 287:2520325215.
mother 55:132139. 78. Valds R, Reyes B, Alvarez T, Garca J, Montero
70. Bowmann E. 1998. Iraqi Chemical and Biological JA, Figueroa A, Gmez L, Padilla S, Geada D,
Weapons Capabilities, p 15. Congressional Re- Abrahantes MC, Dorta L, Fernndez D,
search Service, Washington, DC. Mendoza O, Ramirez N, Rodriguez M, Pujol
71. Cor d e sm an RA. 19 9 8 . U N S C O M M a i n M, Borroto C, Brito J. 2003. Hepatitis B surface
Achievements, p 6771. United Nations Special antigen immunopurification using a plant-derived
Commission (UNSCOM), New York, NY. specific antibody produced in large scale. Biochem
72. Gill DM. 1982. Bacterial toxins: a table of lethal Biophys Res Commun 310:742747.
amounts. Microbiol Rev 46:8694. 79. Blank GS, Zapata G, Fahrner R, Milton M,
73. Hauer PJ, Clough NE. 1999. Development of Yedinak C, Knudsen H, Schmelzer C. 2001.
monoclonal antibodies suitable for use in antigen Expanded bed adsorption in the purification of
quantification potency tests for clostridial veter- monoclonal antibodies: a comparison of process
inary vaccines. Dev Biol Stand 101:8594. alternatives. Bioseparation 10:6571.
74. Burnett JC, Henchal EA, Schmaljohn AL, Bavari 80. Zeitlin L, Bohorov O, Bohorova N, Hiatt A, Kim
S. 2005. The evolving field of biodefence: thera- Do H, Pauly MH, Velasco J, Whaley KJ,
peutic developments and diagnostics. Nat Rev Barnard DL, Bates JT, Crowe JE Jr, Piedra
Drug Discov 4:281297. PA, Gilbert BE. 2013. Prophylactic and therapeu-
75. Schlievert PM. 1993. Role of superantigens in tic testing of Nicotiana-derived RSV-neutralizing
human disease. J Infect Dis 167:9971002. human monoclonal antibodies in the cotton rat
76. Schlievert PM. 1986. Staphylococcal enterotoxin model. mAbs 5(2):263269. doi:10.4161/mabs.
B and toxic-shock syndrome toxin-1 are signifi- 23281. Epub Feb 8 2013.
Vector-Mediated In Vivo
Antibody Expression

BRUCE C. SCHNEPP1 and PHILIP R. JOHNSON1


25
INTRODUCTION

The holy grail of human immunodeficiency virus (HIV) vaccine development is


an immunogen that elicits antibodies that neutralize field strains of the virus. In
recent years, we have gained tremendous insights into the structure and function
of the HIV envelope glycoprotein, but limited progress has been made in designing
such immunogens. These sobering observations underscore the tremendous
hurdles that must be overcome to develop an effective HIV vaccine (1, 2, 3, 4, 5).
Foremost among these hurdles is the inability to induce antibodies that neutralize
a wide array of HIV field isolates. Such antibodies are rare, and, until recently, only
a handful of these antibodies had been isolated (6, 7, 8, 9). Over the past few years, a
much larger number of HIV antibodies have been identified that have a much
broader range of neutralization and are orders of magnitude more potent than the
previously identified group (10, 11, 12, 13). These antibodies were isolated from the
high-throughput screening of sera from HIV-1-infected individuals and catego-
rized as elite neutralizers based on their neutralization breadth and potency (14).
Extensive sequence analysis of these potent, broadly neutralizing antibodies
revealed that high levels of somatic mutations were involved to generate the
mature antibody (11). Furthermore, the maturation may have involved repeated
rounds of antibody selection through HIV antigen interaction, a process that may

1
The Childrens Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104.
Antibodies for Infectious Diseases
Edited by James E. Crowe, Jr., Diana Boraschi, and Rino Rappuoli
2015 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.AID-0016-2014

427
428 SCHNEPP AND JOHNSON

not be possible to duplicate from a traditional fer can be applied to other difficult vaccine
HIV protein subunit or viral vector vaccine. targets like hepatitis C virus, malaria, respira-
Given the obstacles required to generate this tory syncytial virus, and tuberculosis.
class of broadly neutralizing antibodies by
current vaccine strategies, one option is to
deliver these antibodies by passive immuniza- IMPORTANCE OF HIV-NEUTRALIZING
tion. Passive immunization schemes using neu- ANTIBODIES
tralizing antibodies have protected monkeys
from simian-human immunodeficiency virus The need for a vaccine is still great, with ap-
(SHIV) challenge infections (15, 16, 17, 18, 19, proximately 35 million people currently in-
20). Unfortunately, an injection of antibodies fected with HIV, and more than 2 million new
every few weeks is neither practical nor cost- adult infections every year. Two earlier vac-
effective as a large-scale human vaccine ap- cine approaches, each targeting a different
proach. The vector-mediated gene transfer arm of the immune response, were evaluated
vaccine strategy eliminates the problems with in large efficacy trials. Both approaches failed
passive antibody transfer and uses a viral vector to protect vaccine recipients from infection,
to deliver the potent, broadly neutralizing and neither diminished viral replication after
antibodies directly to muscle by gene transfer. infection (23, 24, 25, 26). The more recent
In this scheme, the antibody gene of choice is RV144 trial in Thailand using a canarypox
packaged into an adeno-associated virus (AAV) virus expressing HIV proteins in conjunction
vector, which is then delivered by direct intra- with a gp120 subunit showed moderate effi-
muscular injection. Thereafter, antibody mole- cacy (31%), indicating that protection may be
cules are endogenously synthesized in myofibers achievable with the right immunogen (27).
and passively distributed to the circulatory Detailed analyses of the RV144 study results
system (21). In a proof-of-concept experiment revealed two significant correlations with
in a large animal model, rhesus monkeys were infection among vaccine recipients. The pre-
injected with AAV vectors expressing monkey sence of IgG antibodies against V1V2 Env may
antibodies able to neutralize simian immunode- have contributed to protection against HIV-1
ficiency virus (SIV) (22). The neutralizing infection, whereas high levels of Env-specific
antibodies were detected in the serum for over IgA antibodies correlated inversely with in-
6 years following a single intramuscular injec- fection (28). Very recently, the HVTN 505 trial
tion. Furthermore, monkeys were protected was stopped for futility. The HVTN 505 trial
from infection and the development of AIDS showed no difference in HIV-1 infections be-
following challenge with virulent SIV. tween those recipients who received the vaccine
The concept of immunoprophylaxis by gene and those receiving placebo (29). Vaccine
transfer is significant in that it can rapidly move recipients did generate IgG antibodies to Env;
existing and newly discovered molecules that however, the majority were nonneutralizing
block HIV infection into the clinic. In fact, the with low reactivity to the V1V2 antigen (29).
development of molecules that inhibit all steps The observations from these clinical trials,
in HIV entry could create a multilayered that neutralizing antibodies may be essential
blockade against HIV infection and provide a for an effective vaccine, is reinforced by the
shortcut to an HIV vaccine. These molecules large number of new, highly potent broadly
could be directed at any or all points of HIV neutralizing antibodies (bNAbs) that have
entry including (i) gp120 binding to CD4, been identified by using improved screening
(ii) gp120 binding to CCR5, and (iii) mem- and sequencing techniques. These new anti-
brane fusion. This concept need not be bodies were isolated by high-throughput
limited to HIV. In fact, the general strategy of screening of sera from healthy HIV-1-infected
immunoprophylaxis by antibody gene trans- individuals categorized as elite neutralizers
CHAPTER 25 Vector-Mediated In Vivo Antibody Expression 429

based on their neutralization breadth and Passive immunization using neutralizing mono-
potency (10, 11, 12, 13, 14, 30, 31, 32, 33). clonal antibodies has protected monkeys from
Detailed analyses of these antibodies indicated SHIV challenge infections (15, 16, 17, 18, 19, 20,
they are approximately 10- to 100-fold more 39). In a recent study by Moldt et al. (39), they
potent and have an increased breadth com- showed that passively administered PGT121
pared with the original 4 isolates (6, 7, 8, 9). can mediate sterilizing immunity against
Furthermore, this new class of antibodies can SHIV in monkeys at serum concentrations
neutralize HIV-1 through binding to a variety that were significantly lower than those ob-
of envelope domains including the CD4 served in previous studies (as low as 1.8 g/ml).
binding site (VRC01, NIH45-46, and PGV04) While this study demonstrated the potential
(13, 31, 34), glycan-containing regions in the for passive immunization with the new class of
variable loops (PG9, PG16, PGT121, and bNAbs, unfortunately, an injection of antibodies
PGT128)(11, 12), and the membrane-proximal every few weeks is not practical or cost-
external region (MPER) on gp41 (10E8) (30). effective as a large-scale human prophylactic
Epitope mapping of these new, potent anti- vaccine approach. Given these difficulties, we
bodies has invigorated the vaccine field by developed a second option: isolate the repre-
providing precise regions to target when design- sentative antibody gene and use gene transfer
ing new protein or subunit vaccine antigens to technology to endow a target host with the
induce bNAbs (35). However, even with this gene. In this way, the antibody gene directs
new wealth of information at hand, generating endogenous expression of the antibody mole-
bNAbs with improved, redesigned antigens may cule, and the host (in theory) will now have the
still prove to be problematic. Extensive sequence antibody in its circulation. Thus, after a single
analysis of these potent broadly neutralizing injection, the muscle now serves as a depot to
antibodies reveal that high levels of somatic synthesize the bNAbs that are passively distrib-
mutations (as much as 30%) can occur in the uted to the circulatory system (Fig. 1). The host
generation of the mature antibody (11, 13, 31, 33, is now armed with a potent bNAb against HIV-
36). Furthermore, the maturation may have 1 that effectively bypasses the adaptive immune
involved repeated rounds of antibody selection system. This is in contrast to the traditional idea
through HIV antigen interaction. In light of this, of passive immunization whereby the purified
several groups have developed novel immuno- antibodies are injected intravenously into the
gens, such as glycopeptides or computation- host to provide protection from infection.
derived multimerized nanoparticles, that are However, because of the antibody half-life,
designed to induce bNAbs (37, 38). These the levels decline, requiring repeated injections.
immunogens can bind to both mature bNAbs as The obvious advantage is that antibody gene
well as the receptors of their germline (nave) transfer engenders the host with long-term
B cells, which can trigger the activation and antibody persistence from a single injection due
maturation process required to produce a bNAb. to endogenous antibody expression.

HIV-NEUTRALIZING ANTIBODIES VECTOR EXPRESSION SYSTEMS FOR


AS A VACCINE ANTIBODY GENE TRANSFER

While induction of bNAbs by various next- Our chosen vector to deliver the antibody gene
generation immunization strategies holds is the recombinant adeno-associated virus
promise, the question remains as to the best (rAAV) vector, which is derived from wild-
use of the human monoclonal antibodies that type AAV. AAV is a Dependovirus with a 4.7-kb
have already been isolated and characterized. single-strand DNA genome that contains only
One obvious option is passive immunization. two genes (rep and cap) flanked by inverted
430 SCHNEPP AND JOHNSON

FIGURE 1 Immunoprophylaxis by antibody gene transfer. Passive immunization involves intravenous


delivery of puried antibodies to engender the host with short-lived immunity in serum and mucosa. In
contrast, vector-mediated antibody gene transfer uses a viral vector to deliver the antibody gene to the
host via intramuscular injection. The antibody is produced endogenously in the muscle and secreted into
the circulatory system and mucosa providing long-term protection from infection. (Reprinted from
reference 72 [Schnepp BC, Johnson PR. 2014. Adeno-associated virus delivery of broadly neutralizing
antibodies. Curr Opin HIV AIDS 9:205256] with permission.) doi:10.1128/microbiolspec.AID-0016-2014.f1

terminal repeats (ITRs). AAV natural infection rAAV gene transfer vectors are devoid of
is common and has not been associated with the endogenous rep and cap genes, and consist
any disease. Multiple AAV serotypes have been of the antibody gene expression cassette
identified with different transduction efficien- flanked by the AAV ITRs (Fig. 2). The ITRs
cies in different tissues, offering flexibility for (145 bp each), which are necessary for rAAV
gene transfer targets such as muscle or liver vector genome replication and packaging, are
(40). rAAV vectors have an established record the only part of the AAV genome present in
of high-efficiency gene transfer in a variety of the rAAV vectors. One method for antibody
model systems (41, 42). Following injection, expression utilizes a two-promoter system
the rAAV vector genome can form stable whereby the heavy- and light-chain genes
nonintegrating circular episomes that can per- are transcribed independently by using two
sist in nondividing cells (43, 44, 45). Because of different promoters and polyadenylation
these features, rAAV vectors have become signals within the same rAAV vector genome
popular gene delivery vehicles for use in clinical (Fig. 2) (47). Another method uses a single
studies for the treatment of diseases such as promoter for expression of both the heavy and
alpha1-antitrypsin deficiency, cystic fibrosis, light chains, which are separated by the foot-
hemophilia B, Lebers congenital amaurosis, and-mouth-disease virus (FMDV) 2A peptide,
lipoprotein lipase (LPL) deficiency, Parkinsons which undergoes self-cleavage to produce
disease, and muscular dystrophy (46). separate heavy- and light-chain proteins
CHAPTER 25 Vector-Mediated In Vivo Antibody Expression 431

FIGURE 2 rAAV vectors for antibody gene transfer. (A) The wild-type AAV (wtAAV) genome consists of the rep
and cap genes anked by inverted terminal repeats (ITRs). For rAAV vectors, the rep and cap genes are removed
and replaced by an antibody expression cassette anked by ITRs, which are necessary for rAAV vector genome
replication and packaging. (B) Immunoadhesins contain the antibody variable domains (VL, variable light; VH,
variable heavy) usually joined by a exible protein linker. The variable domains are connected to the hinge and
constant heavy-chain domains (CH2 and CH3). The immunoadhesins can form dimers through disulde bonding
in the hinge region. (C) Full antibodies can be expressed by using either a dual promoter or single-promoter
system. For dual promoter expression, the antibody heavy and light chains are each expressed separately from
their own promoter. For the single promoter system, the heavy and light chains are expressed as a single
polypeptide separated by the foot-and-mouth-disease virus 2A peptide (FMDV-2A). The FMDV-2A peptide can
undergo self-cleavage to give rise to separate heavy and light chains. doi:10.1128/microbiolspec.AID-0016-2014.f2

(Fig. 2) (48). The advantage of this system is constant light-chain domain and the constant
that the heavy and light chains can potentially heavy-chain domain 1 (CH1); however, it can be
be expressed in a 1:1 ratio using a single expressed as a single polypeptide from a single
promoter, which may translate to more effi- promoter, and forms dimers through disulfide
cient expression. However, a potential disad- bonding in the hinge region. While immuno-
vantage is that the FMDV-2A peptide is adhesins have many attractive features such as
derived from a viral sequence and may be efficient expression/secretion in vivo, they also
immunogenic in the host causing immune have some drawbacks. Immunoadhesins may
clearance of cells expressing the antibody. not exhibit the same neutralization breadth and
Another option is to create an immuno- potency as the native antibody. While we have
adhesin version of the neutralizing antibody. seen cases where a specific immunoadhesin
Immunoadhesins are chimeric, antibody-like functions identically to its native antibody
molecules that combine the functional domain counterpart, we have also seen an immuno-
of a binding protein like a scFv or CD4 adhesin become 10-fold less potent at neutral-
extracellular domains 1 and 2 (D1D2) with an izing HIV-1 (unpublished observation). Thus,
immunoglobulin constant domain (49) (Fig. 2), immunoadhesins must be fully characterized
and have been shown to be effective in dis- and compared with the native antibody from
ease models including HIV, SIV, and influenza which they were derived before consideration
(22, 50, 51). A typical immunoadhesin lacks the as a vaccine. Another drawback to using
432 SCHNEPP AND JOHNSON

immunoadhesins is possible immunogenicity. active humoral response, it was difficult to


Immunoadhesins are not naturally occurring determine if this activity would translate to
proteins and may contain amino acid linkers protection from a challenge infection. There-
connecting the variable domains (Fig. 2), fore, our next goal was to test our hypothesis in
which could trigger an immune response a macaque challenge model, but we were faced
leading to the loss of expression. However, it with the problem that the human antibody
should be noted that Enbrel (etanercept), an IgGb12 was viewed as foreign in a macaque,
immunoadhesin consisting of the tumor necro- triggering an immune response that lead to
sis factor receptor fused to IgG1-Fc, was well the elimination of IgG1b12 expression. We
tolerated in patients for long-term treatment then turned to using rhesus-derived antibodies
(10 years) of rheumatoid arthritis (52). by taking advantage of native macaque SIV
gp120-specific Fab molecular clones that
had been derived directly from SIV-infected
PROOF-OF-CONCEPT STUDIES IN macaques (53). When designing the antibody
ANIMAL MODELS gene transfer vectors, we chose to express the
Fabs as immunoadhesins, which in pilot
We first developed the concept of using experiments in mice were superior to single-
antibody gene transfer in 2002 (47). Because chain (scFv) or whole-antibody (IgG) mole-
of the significant obstacles that confronted both cules with respect to steady-state serum
active and passive immunization strategies, concentrations (unpublished data).
we began to explore an alternate strategy to For the macaque experiments, we con-
generate serum antibodies that neutralize pri- structed immunoadhesins derived from two
mary isolates of HIV-1. At that time, only a few different SIV Fab fragments (4L6 and 5L7), as
monoclonal antibodies existed (6, 7, 8, 9). We well as a third immunoadhesin containing the
chose to express IgG1b12 to test the feasibility of rhesus CD4 D1D2, which was modeled after
antibody gene transfer using rAAV vectors. To CD4-Ig fusion proteins (54). All of the con-
generate the IgG1b12 expression construct, the structs neutralized in vitro the proposed SIV
IgG1b12 heavy and light chains were expressed challenge stock (SIVmac316), indicating that
independently in the same rAAV vector using the immunoadhesins were functioning like the
the dual-promoter system. The resulting vector original Fab clones (22). The 3 immuno-
was injected into the quadriceps muscles of adhesins were injected into 3 monkeys each
immunodeficient mice (to avoid immune re- (for 9 total), followed by an intravenous SIV
sponses to human IgG). IgG1b12 was expressed challenge 4 weeks later, including 6 nave
in mouse muscle (confirmed by histochemical controls. Immunoadhesin expression levels
staining), and biologically active antibody was were as high as 190 g/ml at the time of
found in sera for over 6 months (47). Charac- challenge (4 weeks postinjection) and peaked
teristic biologic activity was determined by HIV around 6 months with levels reaching 400 g/
neutralization assays against IgG1b12 sensitive/ ml in some animals (22). Overall, 6 of the 9
resistant viruses. This study provided the first monkeys receiving the immunoadhesins were
evidence that: (i) rAAV vectors transferred completely protected after challenge, while all
antibody genes to muscle; (ii) myofibers pro- 6 nave controls became infected. Analysis of
duced antibodies; (iii) antibodies were distrib- the 3 monkeys from the immunoadhesin group
uted to the circulation; and (iv) such antibodies that became infected revealed that these
were biologically active. specific animals had developed an immune
While these initial findings supported the response to the immunoadhesin by 3 weeks
hypothesis that antibody gene transfer to the postinjection, suggesting a correlation between
muscle can produce systemic levels of HIV- an immune response to the immunoadhesin
neutralizing activity without the need for an and failure to protect from infection. We have
CHAPTER 25 Vector-Mediated In Vivo Antibody Expression 433

performed longitudinal studies of the protected vector expressing bNAb 10-1074 (56), which
monkeys, which are now over 6 years postin- targets the base of the V3 stem of gp120 (57).
jection. Immunoadhesin levels dropped to a These mice maintained a high level of antibody
stable level of approximately 20 g/ml, which 10-1074 expression of around 200 g/ml for the
has persisted for over the past 4 years. The entire length of the 67-day observation period.
monkeys have remained negative for SIV During this time, 6 of the 7 mice in the group
infection and have not developed an immune were able to control HIV plasma viral loads,
response to the immunoadhesins (unpublished whereas 1 mouse exhibited viral escape. As seen
observation). Thus, this crucial study was with escape mutants from passive immuniza-
instrumental in proving the concept of vector- tion studies (56, 58), sequence analysis of the
mediated gene transfer as a viable HIV vaccine. gp120 of these escape viruses revealed muta-
More recently, other investigators per- tions in the 10-1074 binding site that conferred
formed rAAV vector-mediated gene transfer resistance to the antibody. It remains to be seen
expression/challenge studies, which they called if simultaneously administering rAAV vectors
vectored immunoprophylaxis (VIP) (55). They expressing multiple bNAbs could dramatically
expressed the native, full antibodies of 2G12, reduce or even possibly eliminate the genera-
IgG1b12, 2F5, 4E10, and VRC01 using the single- tion of escape mutants. Furthermore, long-term
promoter FMDV-2A system. Following intra- studies will be required to see if escape mutants
muscular rAAV injection in mice, antibody could arise over time, even in the presence of
expression levels greater than 100 g/ml were multiple antibodies. These results in human-
observed for at least 12 months. Using a ized mice suggest that using vector-mediated
humanized mouse model, they further showed gene transfer to deliver bNAbs to HIV-infected
that these rAAV vectors provided protection individuals could be a viable option, possibly
following HIV challenge, with antibody serum even used in conjunction with standard
levels as low as 8.3 g/ml (antibody VRC01). antiretroviral therapy (ART). An overriding
These encouraging results reinforce the effi- theme is that multiple bNAbs would be required
cacy of the antibody gene transfer approach, to provide the selective pressure to avoid viral
especially when potent antibodies such as escape mutants. Multiple antibodies could
VRC01 are used. Taken together, these murine target different gp120 domains such as the
and primate studies show that vector-mediated exterior loops, CD4 binding site, and MPER.
antibody gene transfer can bypass the adaptive Furthermore, multiple antibodies could be used
immune response and engender the host with that target different stages of viral entry
antibodies that provide protection from infec- including CD4 binding, CCR5 binding, and
tion. Furthermore, antibody expression can membrane fusion. Of course, this strategy of
persist several years following a single injection, the simultaneous use of multiple antibodies
suggesting that long-term protection is possible. against multiple viral targets or stages of entry
could also be applicable in a prophylactic
vaccine approach for maximum efficacy.
ANTIBODY GENE TRANSFER FOR
HIV-POSITIVE INDIVIDUALS
ANTIBODY GENE TRANSFER FOR
While antibody gene transfer shows great RESPIRATORY TRACT INFECTIONS
promise for providing protection from HIV
infection, one obvious question is whether this The use of vector-mediated antibody gene
strategy can also be used for antibody therapy in transfer has not been limited to just HIV
HIV-positive individuals. To answer the ques- (Table 1). Respiratory syncytial virus (RSV) is
tion, HIV-infected humanized mice received an a major cause of severe respiratory infection
intravenous injection of a rAAV (serotype 8) in high-risk populations (such as infants) for
434 SCHNEPP AND JOHNSON

TABLE 1 Vector-mediated antibody gene transfer are quickly cleared by the immune system,
studies resulting in the rapid loss of transgene expres-
Application Antibody Reference sion. In contrast, rAAV vectors have very low
HIV SIV Lewis 2002 (47), immunogenicity and can give rise to long-
vaccine immunoadhesins, Johnson 2009 (22), term gene (antibody) expression for potentially
4E10, 2G12, 2F5, Balazs 2012 (55) the life of the individual. The Ad-palivizumab
b12, VRC01, PG9a
HIV 10-1074 Horwitz 2013 (56) vector was administered intravenously, with
therapy palivizumab detected in the lungs by day 3
RSV Palivizumab Skaricic 2008 (59) postadministration. Following an intranasal
vaccine RSV challenge 7 days postadministration, the
Inuenza FI6, F10, CR6261 Limberis 2013 (61), mice showed >5-fold decrease in RSV titers in
vaccine Balazs 2013 (62)
Drug NIC9D9, Hicks 2012 (65),
the lung compared with control animals. Long-
addiction GNC92H2 Rosenberg 2012 term antibody expression and challenge studies
(66) were done using the rAAVrh.10-palivizumab
Cancer DC101 Fang 2005 (48) vector via intrapleural administration. Palivi-
therapy
zumab was detected in the serum of these
a
The rst clinical trial using rAAV vector-mediated PG9 antibody animals by 8 weeks postadministration that
gene transfer began in 2014 as a result of collaboration between
The Childrens Hospital of Philadelphia, The International AIDS started to peak by week 20. These rAAVrh.10-
Vaccine Initiative, and the Division of AIDS (DAIDS) in the National palivizumab mice were intranasally challenged
Institute of Allergy and Infectious Diseases. with RSV at 7 and 21 weeks postadministra-
tion. They showed 14.3-fold and 10.6-fold lower
which a vaccine is not yet available. Currently, numbers of RSV PFU in the lungs, indicating
the only way to prevent infection is through the that protection against RSV infection can be
passive administration of anti-RSV antibodies, sustained at least 21 weeks postdelivery of an
such as palivizumab (also known as Synagis, rAAV vector.
manufactured by MedImmune). This antibody Antibody gene transfer studies using rAAV
can be administered intramuscularly once each vectors have also been done to prevent influ-
month during the RSV season (winter and enza. Although traditional vaccination strate-
spring) to prevent RSV infection. While this gies for influenza are quite effective, they may
treatment is effective, it is costly and limited to not be adequate for a possible zoonotic strain
high-risk individuals, which are attributes that that could lead to a pandemic (such as the 2009
make it a prime candidate for antibody gene H1N1). In this case, the time needed to develop
transfer. Instead of repeated monthly injections a traditional vaccine may not be rapid enough.
of the purified antibody, the antibody could be The rationale is that vector-mediated antibody
endogenously expressed from a single injection gene transfer could quickly deliver a bNAb that
using antibody gene transfer and provide a is effective against multiple strains of influenza
constant level of protective anti-RSV antibodies that would provide protection against a pan-
in the host. demic. One study looked at delivering the
The study used different vector systems to bNAb antibody FI6 (60) as an immunoadhesin
deliver antibodies against RSV infection (59). using rAAV serotype 9 via intranasal delivery
They compared the expression and efficacy of a in mice and ferrets (51). FI6 immunoadhesin
mouse version of palivizumab in a mouse model expression was detected in the nasal and lung
system when delivered by either a rAAV vector lavage fluids of mice 14 days after vector
(serotype rh.10) or adenovirus (Ad) vector. administration at concentrations ranging from
Adenovirus vectors have the capacity for high- 0.5 to 2.0 g/ml. Animals challenged as early as
level gene transfer with rapid and robust 3 days after rAAV9-FI6 administration could be
transgene expression. However, Ad vectors protected. Furthermore, this strategy was able
are highly immunogenic, and transduced cells to protect both mice and ferrets from the
CHAPTER 25 Vector-Mediated In Vivo Antibody Expression 435

exposure of lethal doses of various clinical 83% of the nicotine bound to the NIC9D9
isolates of H5N1 and H1N1. An additional antibody in serum, which drastically reduced
study (61) also demonstrated that rAAV9-FI6 the amount of serum delivered to the brain.
administration showed partial efficacy in mice Furthermore, these mice had reduced cardio-
challenged with the newly emergent avian vascular effects compared with control animals.
H7N9, which is believed to be transmitted These results indicate that this strategy may
from poultry to humans. hold promise as an effective preventative
A separate study used a similar strategy but therapy for nicotine addiction.
with intramuscular administration of the rAAV Along the same line, GNC92H2 is a mouse
antibody vector in mice (62). They expressed monoclonal antibody with high affinity for
antibodies F10 (63) and CR6261 (64) in an cocaine. This antibody was also delivered to
rAAV serotype 8 vector by using the FMDV-2A mice by using the rAAVrh.10 vector via intra-
expression system. Antibody expression levels venous injection (66). GNC92H2 was detected
in the serum reached 200 g/ml at 5 weeks in the serum for the entire duration of the 24-
after intramuscular injection, with levels still week study. The GNC92H2 antibody was able
around 10 g/ml out to at least 11 months after a to sequester intravenously administered co-
single injection. These treated mice were caine in the blood, thereby protecting the brain
protected from diverse strains of H1N1 influ- from the effects of cocaine. Furthermore, these
enza when challenged at either of these time mice showed suppressed cocaine-induced hy-
points (5 weeks and 11 months), demonstrating peractivity derived from weekly cocaine expo-
once again the incredible potential for this sure (12 to 17 weeks after rAAVrh.10 vector
strategy as a vaccine. The results from both the administration). These findings offer an alter-
intranasal (51) and intramuscular (62) routes of native intervention to cocaine addiction ther-
vector administration reinforce the flexibility apy. High-affinity cocaine antibodies could be
of vector-mediated gene transfer and provide maintained long term in the serum following a
important proof-of-concept studies that could single administration. This strategy could be
lead to translation into humans. coupled with traditional behavioral therapies
for a combined approach for the treatment of
cocaine addiction.
ANTIBODY GENE TRANSFER FOR
DRUG ADDICTION
ANTIBODY GENE TRANSFER
Perhaps a less conventional use of vector- FOR CANCER
mediated antibody gene transfer is a potential
role in the treatment of drug/substance addic- The vector-mediated antibody gene transfer
tion. Antibodies exist that can bind to these strategy can be expanded for use in noninfec-
substances in the blood and prevent their tious disease applications where antibodies still
transfer to the brain, which leads to their play a critical role, such as cancer treatment. In
addictive properties. Antibody therapy for a study by Fang et al. (48), they examined the
addiction would require routine, costly in- efficacy of an antitumor antibody to reduce
jections, which once again makes this a prime tumor growth in a mouse model system. They
candidate for antibody gene transfer. NIC9D9 is used an rAAV8 vector that expressed antibody
an antinicotine antibody that was delivered DC101 by the FMDV-2A system. Antibody
intravenously (targeting the liver) to mice by DC101 is an antiangiogenic monoclonal anti-
using a rAAVrh.10 vector (65). NIC9D9 anti- body that targets vascular endothelial cell
body was detected in the serum for the length of growth factor receptor-2 (VEGFR2). Mice
the 18-week study. Following intravenous nic- given an intravenous injection of rAAV8-
otine delivery, the rAAV-NIC9D9 mice had DC101 could express high levels (>1 mg/ml) of
436 SCHNEPP AND JOHNSON

the antibody in the serum for the length of the Perhaps of greater concern is the risk
5-month monitoring period. Mice receiving that the antibody will bind off-target caus-
this rAAV vector exhibited shrinkage of tumors ing an unanticipated adverse event. Preclin-
and prolonged survival time compared with ical testing, such as passive administration
untreated control animals. These encouraging and Good Laboratory Practice (GLP) human
results set the stage for combining antibody tissue binding studies, can help avert most
gene transfer technology with the ever increas- of these issues. However, if off-target
ing number of antibody-based therapies for effects occur in vivo, there is currently no
cancers that include such antibodies as efficient method to stop antibody gene
Herceptin and Avastin (Genentech), to name expression. As the data shows from animal
a few. models, antibodies are expressed for poten-
tially the life of the host following a single
intramuscular administration. A few studies
DRAWBACKS OF rAAV ANTIBODY have attempted to regulate gene expression
GENE TRANSFER from rAAV gene transfer vectors in mice
and monkeys (69, 70, 71), but these schemes
Essentially any therapeutic or immunopro- are transient and require continuous exog-
phylactic protein can be expressed by using enous drug administration to maintain a
rAAV vector gene transfer, as long as it fits constant level of gene expression. Clearly,
within the vector packaging limit. However, identifying an efficient method to perma-
one must be careful that the expressed protein nently eliminate antibody gene expression
is not immunogenic in the host. However, this is in the host is a top priority if rAAV vector-
also the same concern for all exogenously mediated antibody gene transfer is to be-
(passively) administered proteins, including come applicable for wide-scale use.
monoclonal antibodies and other biologics.
Most, if not all, of the 25 monoclonal antibodies ACKNOWLEDGMENT
that have been approved as therapeutics have
exhibited some level of immunogenicity (67, Conflicts of interest: We declare no conflicts.
68). Several factors may contribute to immuno-
genicity including antibody structure, dosing CITATION
regimen, and the recipients genetic back-
Schnepp BC, Johnson PR. 2014. Vector-
ground. Also, it remains to be determined if an
mediated in vivo antibody expression. Micro-
antibody that was endogenously expressed in
biol Spectrum 2(4):AID-0016-2014.
the host via gene transfer will be more or less
immunogenic than when passively adminis-
tered as an exogenously produced protein. The REFERENCES
big question is what effect would an immune 1. Desrosiers R. 2004. Prospects for an AIDS
response to the transgene have in the host? In vaccine. Nat Med 10:221223.
the simplest scenario, as was seen in the non- 2. Fauci AS, Johnston MI, Dieffenbach CW,
Burton DR, Hammer SM, Hoxie JA, Martin M,
human primate studies (22), the appearance of
Overbaugh J, Watkins DI, Mahmoud A, Greene
anti-antibody responses would limit the vaccine WC. 2008. HIV vaccine research: the way for-
efficiency through loss of transgene expression, ward. Science 321:530532.
with no adverse events observed. Ultimately, at 3. Morgan C, Marthas M, Miller C, Duerr A, Cheng-
this stage, it is difficult to predict with any Mayer C, Desrosiers R, Flores J, Haigwood N, Hu
SL, Johnson RP, Lifson J, Montefiori D, Moore J,
certainty which, if any, of the candidates would
Robert-Guroff M, Robinson H, Self S, Corey L.
be immunogenic, and what the consequences 2008. The use of nonhuman primate models in
would be. Human clinical trials will be the best HIV vaccine development. PLoS Med 5:e173.
predictor. doi:10.1371/journal.pmed.0050173.
CHAPTER 25 Vector-Mediated In Vivo Antibody Expression 437

4. Walker B, Burton D. 2008. Toward an AIDS 14. Simek MD, Rida W, Priddy FH, Pung P, Carrow
vaccine. Science 320:760764. E, Laufer DS, Lehrman JK, Boaz M, Tarragona-
5. Watkins DI. 2008. Basic HIV Vaccine develop- Fiol T, Miiro G, Birungi J, Pozniak A, McPhee
ment. Top HIV Med 16:78. DA, Manigart O, Karita E, Inwoley A, Jaoko W,
6. Burton DR, Pyati J, Koduri R, Sharp SJ, Dehovitz J, Bekker LG, Pitisuttithum P, Paris R,
Thornton GB, Parren PW, Sawyer LS, Hendry Walker LM, Poignard P, Wrin T, Fast PE,
RM, Dunlop N, Nara PL, et al. 1994. Efficient Burton DR, Koff WC. 2009. Human immunode-
neutralization of primary isolates of HIV-1 by a ficiency virus type 1 elite neutralizers: individuals
recombinant human monoclonal antibody. Sci- with broad and potent neutralizing activity iden-
ence 266:10241027. tified by using a high-throughput neutralization
7. Muster T, Steindl F, Purtscher M, Trkola A, assay together with an analytical selection algo-
Klima A, Himmler G, Ruker F, Katinger H. 1993. rithm. J Virol 83:73377348.
A conserved neutralizing epitope on gp41 of human 15. Baba TW, Liska V, Hofmann-Lehmann R,
immunodeficiency virus type 1. J Virol 67:6642 Vlasak J, Xu W, Ayehunie S, Cavacini LA,
6647. Posner MR, Katinger H, Stiegler G, Bernacky
8. Trkola A, Purtscher M, Muster T, Ballaun C, BJ, Rizvi TA, Schmidt R, Hill LR, Keeling ME,
Buchacher A, Sullivan N, Srinivasan K, Sodroski Lu Y, Wright JE, Chou TC, Ruprecht RM. 2000.
J, Moore JP, Katinger H. 1996. Human monoclonal Human neutralizing monoclonal antibodies of the
antibody 2G12 defines a distinctive neutralization IgG1 subtype protect against mucosal simian-
epitope on the gp120 glycoprotein of human imm- human immunodeficiency virus infection. Nat
unodeficiency virus type 1. J Virol 70:11001108. Med 6:200206.
9. Zwick MB, Labrijn AF, Wang M, Spenlehauer C, 16. Mascola JR, Stiegler G, VanCott TC, Katinger H,
Saphire EO, Binley JM, Moore JP, Stiegler G, Carpenter CB, Hanson CE, Beary H, Hayes D,
Katinger H, Burton DR, Parren PW. 2001. Frankel SS, Birx DL, Lewis MG. 2000. Protection
Broadly neutralizing antibodies targeted to the of macaques against vaginal transmission of a
membrane-proximal external region of human pathogenic HIV-1/SIV chimeric virus by passive
immunodeficiency virus type 1 glycoprotein gp41. infusion of neutralizing antibodies. Nat Med
J Virol 75:1089210905. 6:207210.
10. Burton DR, Poignard P, Stanfield RL, Wilson IA. 17. Parren PW, Marx PA, Hessell AJ, Luckay A,
2012. Broadly neutralizing antibodies present new Harouse J, Cheng-Mayer C, Moore J, Burton
prospects to counter highly antigenically diverse DR. 2001. Antibody protects macaques against
viruses. Science 337:183186. vaginal challenge with a pathogenic R5 simian/
11. Walker LM, Huber M, Doores KJ, Falkowska E, human immunodeficiency virus at serum levels
Pejchal R, Julien JP, Wang SK, Ramos A, Chan- giving complete neutralization in vitro. J Virol
Hui PY, Moyle M, Mitcham JL, Hammond PW, 75:83408347.
Olsen OA, Phung P, Fling S, Wong CH, Phogat S, 18. Hessell AJ, Poignard P, Hunter M, Hangartner
Wrin T, Simek MD, Koff WC, Wilson IA, Burton L, Tehrani DM, Bleeker WK, Parren PW, Marx
DR, Poignard P. 2011. Broad neutralization PA, Burton DR. 2009. Effective, low-titer antibody
coverage of HIV by multiple highly potent anti- protection against low-dose repeated mucosal
bodies. Nature 477:466470. SHIV challenge in macaques. Nat Med 15:951
12. Walker LM, Phogat SK, Chan-Hui PY, Wagner 954.
D, Phung P, Goss JL, Wrin T, Simek MD, Fling S, 19. Hessell AJ, Rakasz EG, Poignard P, Hangartner
Mitcham JL, Lehrman JK, Priddy FH, Olsen OA, L, Landucci G, Forthal DN, Koff WC, Watkins
Frey SM, Hammond PW, Kaminsky S, Zamb T, DI, Burton DR. 2009. Broadly neutralizing human
Moyle M, Koff WC, Poignard P, Burton DR. anti-HIV antibody 2G12 is effective in protection
2009. Broad and potent neutralizing antibodies against mucosal SHIV challenge even at low serum
from an African donor reveal a new HIV-1 vaccine neutralizing titers. PLoS Pathog 5:e1000433.
target. Science 326:285289. doi:10.1371/journal.ppat.1000433.
13. Wu X, Yang ZY, Li Y, Hogerkorp CM, Schief WR, 20. Willey R, Nason MC, Nishimura Y, Follmann
Seaman MS, Zhou T, Schmidt SD, Wu L, Xu L, DA, Martin MA. 2010. Neutralizing antibody
Longo NS, McKee K, ODell S, Louder MK, titers conferring protection to macaques from a
Wycuff DL, Feng Y, Nason M, Doria-Rose N, simian/human immunodeficiency virus challenge
Connors M, Kwong PD, Roederer M, Wyatt RT, using the TZM-bl assay. AIDS Res Hum Retro-
Nabel GJ, Mascola JR. 2010. Rational design of viruses 26:8998.
envelope identifies broadly neutralizing human 21. Berkhout B, Sanders RW. 2012. Gene therapy as a
monoclonal antibodies to HIV-1. Science 329:856 vaccine for HIV-1. Expert Opin Biol Ther 12:1315
861. 1321.
438 SCHNEPP AND JOHNSON

22. Johnson PR, Schnepp BC, Zhang J, Connell MJ, HIV-1 vaccine efficacy trial. N Engl J Med 366:
Greene SM, Yuste E, Desrosiers RC, Clark KR. 12751286.
2009. Vector-mediated gene transfer engenders 29. Hammer SM, Sobieszczyk ME, Janes H, Karuna
long-lived neutralizing activity and protection ST, Mulligan MJ, Grove D, Koblin BA,
against SIV infection in monkeys. Nat Med 15: Buchbinder SP, Keefer MC, Tomaras GD,
901906. Frahm N, Hural J, Anude C, Graham BS,
23. Buchbinder SP, Mehrotra DV, Duerr A, Enama ME, Adams E, DeJesus E, Novak RM,
Fitzgerald DW, Mogg R, Li D, Gilbert PB, Lama Frank I, Bentley C, Ramirez S, Fu R, Koup RA,
JR, Marmor M, Del Rio C, McElrath MJ, Mascola JR, Nabel GJ, Montefiori DC, Kublin J,
Casimiro DR, Gottesdiener KM, Chodakewitz McElrath MJ, Corey L, Gilbert PB. 2013. Efficacy
JA, Corey L, Robertson MN. 2008. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N
assessment of a cell-mediated immunity HIV-1 Engl J Med 369:20832092.
vaccine (the Step Study): a double-blind, ran- 30. Huang J, Ofek G, Laub L, Louder MK, Doria-
domised, placebo-controlled, test-of-concept Rose NA, Longo NS, Imamichi H, Bailer RT,
trial. Lancet 372:18811893. Chakrabarti B, Sharma SK, Alam SM, Wang T,
24. Flynn NM, Forthal DN, Harro CD, Judson FN, Yang Y, Zhang B, Migueles SA, Wyatt R, Haynes
Mayer KH, Para MF. 2005. Placebo-controlled BF, Kwong PD, Mascola JR, Connors M. 2012.
phase 3 trial of a recombinant glycoprotein 120 Broad and potent neutralization of HIV-1 by
vaccine to prevent HIV-1 infection. J Infect Dis a gp41-specific human antibody. Nature 491:
191:654665. 406412.
25. McElrath MJ, De Rosa SC, Moodie Z, Dubey 31. Scheid JF, Mouquet H, Ueberheide B, Diskin R,
S, Kierstead L, Janes H, Defawe OD, Carter Klein F, Oliveira TY, Pietzsch J, Fenyo D,
DK, Hural J, Akondy R, Buchbinder SP, Abadir A, Velinzon K, Hurley A, Myung S,
Robertson MN, Mehrotra DV, Self SG, Boulad F, Poignard P, Burton DR, Pereyra F,
Corey L, Shiver JW, Casimiro DR. 2008. Ho DD, Walker BD, Seaman MS, Bjorkman PJ,
HIV-1 vaccine-induced immunity in the test-of- Chait BT, Nussenzweig MC. 2011. Sequence and
concept Step Study: a case-cohort analysis. Lancet structural convergence of broad and potent HIV
372:18941905. antibodies that mimic CD4 binding. Science
26. Pitisuttithum P, Gilbert P, Gurwith M, Heyward 333:16331637.
W, Martin M, van Griensven F, Hu D, Tappero 32. Zhou T, Georgiev I, Wu X, Yang ZY, Dai K, Finzi
JW, Choopanya K. 2006. Randomized, double- A, Kwon YD, Scheid JF, Shi W, Xu L, Yang Y,
blind, placebo-controlled efficacy trial of a bivalent Zhu J, Nussenzweig MC, Sodroski J, Shapiro L,
recombinant glycoprotein 120 HIV-1 vaccine Nabel GJ, Mascola JR, Kwong PD. 2010.
among injection drug users in Bangkok, Thailand. Structural basis for broad and potent neutra-
J Infect Dis 194:16611671. lization of HIV-1 by antibody VRC01. Science
27. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, 329:811817.
Kaewkungwal J, Chiu J, Paris R, Premsri N, 33. Bonsignori M, Hwang KK, Chen X, Tsao CY,
Namwat C, de Souza M, Adams E, Benenson M, Morris L, Gray E, Marshall DJ, Crump JA,
Gurunathan S, Tartaglia J, McNeil JG, Francis Kapiga SH, Sam NE, Sinangil F, Pancera M,
DP, Stablein D, Birx DL, Chunsuttiwat S, Yongping Y, Zhang B, Zhu J, Kwong PD, ODell
Khamboonruang C, Thongcharoen P, Robb S, Mascola JR, Wu L, Nabel GJ, Phogat S,
ML, Michael NL, Kunasol P, Kim JH. 2009. Seaman MS, Whitesides JF, Moody MA, Kelsoe
Vaccination with ALVAC and AIDSVAX to pre- G, Yang X, Sodroski J, Shaw GM, Montefiori DC,
vent HIV-1 infection in Thailand. N Engl J Med Kepler TB, Tomaras GD, Alam SM, Liao HX,
361:22092220. Haynes BF. 2011. Analysis of a clonal lineage of
28. Haynes BF, Gilbert PB, McElrath MJ, Zolla- HIV-1 envelope V2/V3 conformational epitope-
Pazner S, Tomaras GD, Alam SM, Evans DT, specific broadly neutralizing antibodies and their
Montefiori DC, Karnasuta C, Sutthent R, Liao inferred unmutated common ancestors. J Virol
HX, DeVico AL, Lewis GK, Williams C, Pinter A, 85:999810009.
Fong Y, Janes H, DeCamp A, Huang Y, Rao M, 34. Falkowska E, Ramos A, Feng Y, Zhou T,
Billings E, Karasavvas N, Robb ML, Ngauy V, de Moquin S, Walker LM, Wu X, Seaman MS,
Souza MS, Paris R, Ferrari G, Bailer RT, Wrin T, Kwong PD, Wyatt RT, Mascola JR,
Soderberg KA, Andrews C, Berman PW, Frahm Poignard P, Burton DR. 2012. PGV04, an HIV-1
N, De Rosa SC, Alpert MD, Yates NL, Shen X, gp120 CD4 binding site antibody, is broad and
Koup RA, Pitisuttithum P, Kaewkungwal J, potent in neutralization but does not induce
Nitayaphan S, Rerks-Ngarm S, Michael NL, conformational changes characteristic of CD4.
Kim JH. 2012. Immune-correlates analysis of an J Virol 86:43944403.
CHAPTER 25 Vector-Mediated In Vivo Antibody Expression 439

35. Kwong PD, Mascola JR, Nabel GJ. 2013. persist as episomal chromatin in primate muscle.
Broadly neutralizing antibodies and the search J Virol 82:78757885.
for an HIV-1 vaccine: the end of the beginning. 45. Schnepp BC, Clark KR, Klemanski DL, Pacak
Nat Rev Immunol 13:693701. CA, Johnson PR. 2003. Genetic fate of recom-
36. Zhou T, Zhu J, Wu X, Moquin S, Zhang B, binant adeno-associated virus vector genomes
Acharya P, Georgiev IS, Altae-Tran HR, Chuang in muscle. J Virol 77:34953504.
GY, Joyce MG, Do Kwon Y, Longo NS, Louder 46. Aalbers CJ, Tak PP, Vervoordeldonk MJ. 2011.
MK, Luongo T, McKee K, Schramm CA, Skinner Advancements in adeno-associated viral gene
J, Yang Y, Yang Z, Zhang Z, Zheng A, Bonsignori therapy approaches: exploring a new horizon.
M, Haynes BF, Scheid JF, Nussenzweig MC, F1000 Med Rep 3:17. doi:10.3410/M3-17.
Simek M, Burton DR, Koff WC, Mullikin JC, 47. Lewis AD, Chen R, Montefiori DC, Johnson
Connors M, Shapiro L, Nabel GJ, Mascola JR, PR, Clark KR. 2002. Generation of neutraliz-
Kwong PD. 2013. Multidonor analysis reveals ing activity against human immunodeficiency
structural elements, genetic determinants, and virus type 1 in serum by antibody gene transfer.
maturation pathway for HIV-1 neutralization by J Virol 76:87698775.
VRC01-class antibodies. Immunity 39:245258. 48. Fang JM, Qian JJ, Yi SY, Harding TC, Tu GH,
37. Alam SM, Dennison SM, Aussedat B, Vohra Y, VanRoey M, Jooss K. 2005. Stable antibody
Park PK, Fernandez-Tejada A, Stewart S, Jaeger expression at therapeutic levels using the 2A
FH, Anasti K, Blinn JH, Kepler TB, Bonsignori peptide. Nat Biotechnol 23:584590.
M, Liao HX, Sodroski JG, Danishefsky SJ, 49. Ashkenazi A, Chamow SM. 1997. Immuno-
Haynes BF. 2013. Recognition of synthetic adhesins as research tools and therapeutic agents.
glycopeptides by HIV-1 broadly neutralizing anti- Curr Opin Immunol 9:195200.
bodies and their unmutated ancestors. Proc Natl 50. Capon DJ, Chamow SM, Mordenti J, Marsters
Acad Sci USA 110:1821418219. SA, Gregory T, Mitsuya H, Byrn RA, Lucas C,
38. Jardine J, Julien JP, Menis S, Ota T, Kalyuzhniy Wurm FM, Groopman JE, Broder S, Smith DH.
O, McGuire A, Sok D, Huang PS, MacPherson S, 1989. Designing CD4 immunoadhesins for AIDS
Jones M, Nieusma T, Mathison J, Baker D, Ward therapy. Nature 337:525531.
AB, Burton DR, Stamatatos L, Nemazee D, 51. Limberis MP, Adam VS, Wong G, Gren J, Kobasa
Wilson IA, Schief WR. 2013. Rational HIV D, Ross TM, Kobinger GP, Tretiakova A, Wilson
immunogen design to target specific germline B JM. 2013. Intranasal antibody gene transfer in
cell receptors. Science 340:711716. mice and ferrets elicits broad protection against
39. Moldt B, Rakasz EG, Schultz N, Chan-Hui pandemic influenza. Sci Transl Med 5:187ra172.
PY, Swiderek K, Weisgrau KL, Piaskowski doi:10.1126/scitranslmed.3006299
SM, Bergman Z, Watkins DI, Poignard P, 52. Weinblatt ME, Bathon JM, Kremer JM,
Burton DR. 2012. Highly potent HIV-specific Fleischmann RM, Schiff MH, Martin RW,
antibody neutralization in vitro translates into Baumgartner SW, Park GS, Mancini EL,
effective protection against mucosal SHIV chal- Genovese MC. 2011. Safety and efficacy of
lenge in vivo. Proc Natl Acad Sci USA 109:18921 etanercept beyond 10 years of therapy in North
18925. American patients with early and longstanding
40. Asokan A. 2010. Reengineered AAV vectors: old rheumatoid arthritis. Arthritis Care Res (Hoboken)
dog, new tricks. Discov Med 9:399403. 63:373382.
41. Coura Rdos S, Nardi NB. 2007. The state of the art 53. Johnson WE, Sanford H, Schwall L, Burton DR,
of adeno-associated virus-based vectors in gene Parren PW, Robinson JE, Desrosiers RC. 2003.
therapy. Virol J 4:99. doi:10.1186/1743-422X-4-99 Assorted mutations in the envelope gene of simian
42. Daya S, Berns KI. 2008. Gene therapy using immunodeficiency virus lead to loss of neutraliza-
adeno-associated virus vectors. Clin Microbiol tion resistance against antibodies representing a
Rev 21:583593. broad spectrum of specificities. J Virol 77:9993
43. Nowrouzi A, Penaud-Budloo M, Kaeppel C, 10003.
Appelt U, Le Guiner C, Moullier P, von Kalle C, 54. Allaway GP, Ryder AM, Beaudry GA, Maddon
Snyder RO, Schmidt M. 2012. Integration fre- PJ. 1993. Synergistic inhibition of HIV-1 envelope-
quency and intermolecular recombination of mediated cell fusion by CD4-based molecules in
rAAV vectors in non-human primate skeletal combination with antibodies to gp120 or gp41.
muscle and liver. Mol Ther 20:11771186. AIDS Res Hum Retroviruses 9:581587.
44. Penaud-Budloo M, Le Guiner C, Nowrouzi A, 55. Balazs AB, Chen J, Hong CM, Rao DS, Yang L,
Toromanoff A, Cherel Y, Chenuaud P, Schmidt Baltimore D. 2012. Antibody-based protection
M, von Kalle C, Rolling F, Moullier P, Snyder RO. against HIV infection by vectored immuno-
2008. Adeno-associated virus vector genomes prophylaxis. Nature 481:8184.
440 SCHNEPP AND JOHNSON

56. Horwitz JA, Halper-Stromberg A, Mouquet H, Marasco WA. 2009. Structural and functional
Gitlin AD, Tretiakova A, Eisenreich TR, Malbec bases for broad-spectrum neutralization of avian
M, Gravemann S, Billerbeck E, Dorner M, and human influenza A viruses. Nat Struct Mol Biol
Buning H, Schwartz O, Knops E, Kaiser R, 16:265273.
Seaman MS, Wilson JM, Rice CM, Ploss A, 64. Throsby M, van den Brink E, Jongeneelen M,
Bjorkman PJ, Klein F, Nussenzweig MC. 2013. Poon LL, Alard P, Cornelissen L, Bakker A,
HIV-1 suppression and durable control by com- Cox F, van Deventer E, Guan Y, Cinatl J, ter
bining single broadly neutralizing antibodies and Meulen J, Lasters I, Carsetti R, Peiris M, de
antiretroviral drugs in humanized mice. Proc Natl Kruif J, Goudsmit J. 2008. Heterosubtypic
Acad Sci USA 110:1653816543. neutralizing monoclonal antibodies cross-pro-
57. Mouquet H, Scharf L, Euler Z, Liu Y, Eden C, tective against H5N1 and H1N1 recovered from
Scheid JF, Halper-Stromberg A, Gnanapragasam human IgM+ memory B cells. PLoS One 3:
PN, Spencer DI, Seaman MS, Schuitemaker H, e3942. doi:10.1371/journal.pone.0003942
Feizi T, Nussenzweig MC, Bjorkman PJ. 2012. 65. Hicks MJ, Rosenberg JB, De BP, Pagovich OE,
Complex-type N-glycan recognition by potent Young CN, Qiu JP, Kaminsky SM, Hackett NR,
broadly neutralizing HIV antibodies. Proc Natl Worgall S, Janda KD, Davisson RL, Crystal RG.
Acad Sci USA 109:E3268E3277. 2012. AAV-directed persistent expression of a gene
58. Klein F, Halper-Stromberg A, Horwitz JA, encoding anti-nicotine antibody for smoking cessa-
Gruell H, Scheid JF, Bournazos S, Mouquet H, tion. Sci Transl Med 4:140ra187. doi:10.1126/
Spatz LA, Diskin R, Abadir A, Zang T, Dorner M, scitranslmed.3003611
Billerbeck E, Labitt RN, Gaebler C, 66. Rosenberg JB, Hicks MJ, De BP, Pagovich O,
Marcovecchio PM, Incesu RB, Eisenreich TR, Frenk E, Janda KD, Wee S, Koob GF, Hackett NR,
Bieniasz PD, Seaman MS, Bjorkman PJ, Ravetch Kaminsky SM, Worgall S, Tignor N, Mezey JG,
JV, Ploss A, Nussenzweig MC. 2012. HIV therapy Crystal RG. 2012. AAVrh.10-mediated expression
by a combination of broadly neutralizing anti- of an anti-cocaine antibody mediates persistent
bodies in humanized mice. Nature 492:118122. passive immunization that suppresses cocaine-
59. Skaricic D, Traube C, De B, Joh J, Boyer J, induced behavior. Hum Gene Ther 23:451459.
Crystal RG, Worgall S. 2008. Genetic delivery of 67. Keizer RJ, Huitema AD, Schellens JH, Beijnen
an anti-RSV antibody to protect against pulmonary JH. 2010. Clinical pharmacokinetics of therapeutic
infection with RSV. Virology 378:7985. monoclonal antibodies. Clin Pharmacokinet 49:493
60. Corti D, Voss J, Gamblin SJ, Codoni G, Macagno 507.
A, Jarrossay D, Vachieri SG, Pinna D, Minola A, 68. Kessler M, Goldsmith D, Schellekens H. 2006.
Vanzetta F, Silacci C, Fernandez-Rodriguez BM, Immunogenicity of biopharmaceuticals. Nephrol
Agatic G, Bianchi S, Giacchetto-Sasselli I, Calder Dial Transplant 21(Suppl 5):912.
L, Sallusto F, Collins P, Haire LF, Temperton N, 69. Fang J, Yi S, Simmons A, Tu GH, Nguyen M,
Langedijk JP, Skehel JJ, Lanzavecchia A. 2011. A Harding TC, VanRoey M, Jooss K. 2007. An
neutralizing antibody selected from plasma cells antibody delivery system for regulated expression
that binds to group 1 and group 2 influenza A of therapeutic levels of monoclonal antibodies in
hemagglutinins. Science 333:850856. vivo. Mol Ther 15:11531159.
61. Limberis MP, Racine T, Kobasa D, Li Y, Gao GF, 70. Rivera VM, Gao GP, Grant RL, Schnell MA,
Kobinger G, Wilson JM. 2013. Vectored expres- Zoltick PW, Rozamus LW, Clackson T, Wilson
sion of a broadly neutralizing antibody (FI6) in JM. 2005. Long-term pharmacologically regulated
mouse airway provides partial protection against a expression of erythropoietin in primates following
new avian influenza A (H7N9) virus. Clin Vaccine AAV-mediated gene transfer. Blood 105:1424
Immunol 20:18361837. 1430.
62. Balazs AB, Bloom JD, Hong CM, Rao DS, 71. Nguyen M, Huan-Tu G, Gonzalez-Edick M,
Baltimore D. 2013. Broad protection against Rivera VM, Clackson T, Jooss KU, Harding TC.
influenza infection by vectored immunopro- 2007. Rapamycin-regulated control of anti-
phylaxis in mice. Nat Biotechnol 31:647652. angiogenic tumor therapy following rAAV-mediat-
63. Sui J, Hwang WC, Perez S, Wei G, Aird D, Chen ed gene transfer. Mol Ther 15:912920.
LM, Santelli E, Stec B, Cadwell G, Ali M, Wan H, 72. Schnepp BC, Johnson PR. 2014. Adeno-associated
Murakami A, Yammanuru A, Han T, Cox NJ, virus delivery of broadly neutralizing antibodies.
Bankston LA, Donis RO, Liddington RC, Curr Opin HIV AIDS 9:205256.
Index

AAV (adeno-associated virus), 428436 IgE activation of immune response in presence


Abiotic surfaces, inhibition of bacterial attachment of, 76
to, 28 interactions with IgE and Fc triggering
Activation-induced cytidine deaminase (AID), 331, hypersensitivity responses, 79
332 neutralizing antibody response induced by active
Active systemic anaphylaxis (ASA), 7980 allergen immunotherapy, 8990
Adalimumab, 11, 89, 330, 333334 Allergic inflammation, 7595
ADC (antibody drug complex/conjugate), 9, 15, allergen-specific IgG antibody triggering
334335 hypersensitivity responses, 7980
ADCC (antibody-dependent cellular cytotoxicity), antibodies in treatment of allergies, 8292
3234, 65, 6970, 322326 cancer, 9294
Adcetris (brentuximab vedotin), 15, 334, 335 clinical perspective, 7576
ADCP (antibody-dependent cellular phagocytosis), future roles of antibodies in therapy, 9495
322323, 325 IgE activation of immune response, 76
ADCVI (antibody-dependent cell-mediated virus IgE antibodies in infectious diseases, 8082
inhibition), 3334 IgE effector cells, 7779
ADE. See Antibody-dependent enhancement APCs, 78
Adeno-associated virus (AAV), 428436 B cells, 78
Adhesins, 16, 27 mast cells, 7778
Adhesion of bacteria, interference with, 2728 monocytes and eosinophils, 7879
AD-HIES (autosomal dominant hyper-IgE IgE receptors, 7677
syndrome), 81 interactions between allergens, IgE, and Fc
Affinity matrix, artificial, 151152 triggering hypersensitivity responses, 79
Affinity maturation, 174, 175, 330334 AllergoOncology, 94
broad neutralization versus, 184 Allergy treatment, antibodies in, 8292
by mammalian display, 332333 antibodies targeting cytokines, chemokines, and
in silico, 184185 their receptors, 8889
by somatic hypermutation, 332 clinical studies in 2012, 8687
by somatic mutations and computational design, future roles, 9495
333 IgG4, allergen specific, 9092
in vitro, 331333 neutralizing antibody response induced by active
without display, 333334 allergen immunotherapy, 8990
in vivo, 331 recombinant antibodies targeting IgE interactions
AFM (atomic force microscopy), 382, 386389, with Fc receptors, 8288
391395 specific immunotherapy (SIT), 9092
Agglutination, 26 Alternative pathway of complement activation, 66
Agglutinins, yeast, 115 AMA (Antibody Modeling Assessment), 179180
Aggregation, 26 Aminopterin, 148
Agrobacterium tumefaciens, 414415 AMPV. See Avian pneumovirus/metapneumovirus
AID (activation-induced cytidine deaminase), 331, Analytic vaccinology, 136, 137
332 Anaphylatoxins, 6869
Alemtuzumab, 323, 324 Anaplasma phagocytophilum, 29
Algorithm, antibody docking, 180 Animal models
Allergen sensitization, 76 humanized mice, 158160
Allergens staphylococcal enterotoxin B, 307309

441
442 INDEX

Animal models (continued) for respiratory tract infections, 433435


vector-mediated in vivo antibody expression, Antibody modeling
432433 canonical structure of CDRs, 176178
Anthim, 17 challenges in, 174, 175176
Anthrax, 17, 114, 120, 420 comparative, 176180
Antibacterials, 1617 motivations for, 175
Antibiotics predicting conformation of HCDR3, 177178
introduction of, 5 programs, platforms, and servers dedicated to,
pros and cons of antibody-based therapy relative 179180
to, 14 Antibody Modeling Assessment (AMA), 179180
resistance to, 5, 13 Antibody selection
Antibody AFM, 391393
classes, 10 FACS, 390391
computer-aided prediction of structure and SPR, 389390
design of function, 173187 Antibody-based therapy. See also Immunotherapy;
effector functions, 1011 specific applications
functions, 2536 allergic diseases, 8292
generating, methods and platforms for, 1012 approved/pending therapies, 68, 65
informatics, 363377 complement role in, 6372
structure, 810, 4959, 346347 cost of therapy/production, 12, 16
3D, informatics and, 373376 examples, 1618
Antibody design, 183187 antibacterials, 1617
broad neutralization versus affinity maturation, antifungals, 18
184 antivirals, 1718
overview, 183184 future perspectives, 1819
in silico affinity maturation, 184185 history of, 319
Antibody docking pros and cons, 14
algorithms, 180181 strategic considerations in developing, 1216
epitope mapping and, 180183 antibody engineering, 1315
experimental data, 181183 antibody formats, 1516
modeling, 174, 175176 avoiding escape mutants, 13
Antibody drug complex/conjugate (ADC), 9, 15, magic bullet creation, 15
334335 Antibody-dependent cell-mediated virus inhibition
Antibody engineering, 1315, 321337 (ADCVI), 3334
affinity maturation, 330334 Antibody-dependent cellular cytotoxicity (ADCC),
antibody drug conjugates, 334335 3234, 65, 6970, 322326
antibody informatics and, 370 Antibody-dependent cellular phagocytosis (ADCP),
bispecific antibodies, 335337 322323, 325
choice of Ig isotypes and subclasses, 322324 Antibody-dependent enhancement (ADE),
IgA, 322323 249265
IgG1, 323 intrinsic ADE (iADE), 251, 254258, 263264
IgG2, 323324 Leishmania, 255, 258
IgG3, 324 overview, 249252
IgG4, 324 Antibody-dependent enhancement (ADE), dengue
Fc portion, 322327 virus, 249265
glycosylation, Fc, 324327 afferent phenomena, 252261, 263264
humanization, 327330 enhancing antibodies, 252258, 263264
next-generation antibodies, 334337 genetic host factors contributing to DVPS
Antibody gene rearrangements, 346348 susceptibility, 252, 263
Antibody gene transfer role of dengue viruses in ADE, 258261, 264
for cancer, 435436 DENV-2
drawbacks of rAAV, 436 enhanced growth in primary human monocytes,
for drug addiction, 435 258, 264
expression systems for, 429432 genetic differences after infection with DENV-1,
for HIV-positive individuals, 433 259261, 264
proof-of-concept studies in animal models, genetic differences after infection with DENV-3,
432433 260261, 264
INDEX 443

strain differences in neutralization by DENV-1 Aurograb, 16


antibodies, 258259, 264 Autosomal dominant hyper-IgE syndrome
efferent phenomena, 252, 253, 261, 264265 (AD-HIES), 81
enhancing antibodies, 252258 Autosomal recessive hyper-IgE syndrome
attributes of iADE, 254257, 263 (AR-HIES), 81
role of FcRs, 257258, 264 Avastin, 436
role of infection sequence, 257, 263264 Avian pneumovirus/metapneumovirus
role of myloid cells, 257, 264 (APV/AMPV), 238, 240
sensitizing infection, 252254, 263 Avidin-biotin bridging, 146
Antifungals, 18
Antigen B cell receptor (BCR), 132, 357
studies in humanized mice, 166 B cells
target-agonist approach for antigen discovery in allergic inflammation and, 76, 78
vaccine design, 136137 antibodies blocking CD23 functions, 8384, 88
Antigen design antibody gene rearrangements, 346348
epitope grafting, 185187 high-throughput DNA sequencing and, 346348,
neutralizing antibody elicited through, 185 355357
Antigen presenting cells (APCs), 72, 7678 human hybridoma technology, 141152
Antigen-antibody docking immortalized, 12, 130, 131133, 143, 283
algorithms, 180181 mRNA for phage library, 106
epitope mapping and, 180183 rotavirus and, 289295, 298299
experimental data, 181183 Bacillus anthracis, 17
modeling, 174, 175176 antianthrax monoclonal antibodies discovered by
Antigen-antibody interactions, probing, 381395 phage display, 114
antibody selection antianthrax monoclonal antibodies discovered by
AFM, 391393 yeast display, 120121
FACS, 390391 radioimmunotherapy (RIT), 404405
SPR, 389390 Bacteria. See also specific bacteria
atomic force microscopy (AFM), 382, 386389, antibacterial monoclonal antibodies
391395 discovered by phage display, 113115
fluorescence-activated cell sorting (FACS), 382, discovered by yeast display, 119121
384386, 390391, 394395 antibody-based therapy examples, 1617
overview, 381382 antibody-dependent cellular cytotoxicity
surface plasmon resonance (SPR), 382384, (ADCC), 33
389390, 394 complement activation, 3031, 64
Antilysosomal antibody, 184 history passive antibody therapy, 45
Antiretroviral therapy (ART), 433 hyper-IgE syndrome (HIES), 82
Antivirals, 1718 neutralization of infectivity, 2627
APCs (antigen presenting cells), 72, 7678 phagocytosis, 3435
APV. See Avian pneumovirus/metapneumovirus plant-derived monoclonal antibody for bacterial
APV/AMPV. See Avian pneumovirus/ toxins, 420421
metapneumovirus anthrax, 420
AR-HIES (autosomal recessive hyper-IgE Clostridium perfringens epsilon toxin, 420421
syndrome), 81 staphylococcal enterotoxin B, 421
ART (antiretroviral therapy), 433 radioimmunotherapy (RIT), 404405
ASA (active systemic anaphylaxis), 7980 sensitivity to infection, 82
Aspergillus fumigatus, 403 Bacterial opsonophagocytosis killing assay, 132
Asthma Bacteriophage. See Phage; Phage display
IgE and, 8285 BAFF, 144
omalizumab (Xolair), 8385 Barcode, sequence, 354
respiratory syncytial virus (RSV) and, 82 Bavituximab, 17
Atomic force microscopy (AFM), 382, 386389, Bcl-6, 130131, 164
391395 Bcl-xL, 130131, 164
Atopic dermatitis, staphylococcal enterotoxin B, Benralizumab, 89
309310 Bevacizumab, 184, 330
Attachment, interference with, 2728 Bexxar (tositumumab), 15, 334, 399
Aurexis (tefibazumab), 16 BIAcore SPR system, 383
444 INDEX

Binding affinity, 180 CD30, 334


Binding constant, antibody, 382, 385386, 389390 CD33, 334
Binding interactions, antigen-antibody, 381. See also CD34+ cells, 159162
Antigen-antibody interactions, probing CD40, 77, 132
Biological cloning methods, in human hybridoma CD40L (CD40 ligand), 130, 132, 144, 164
technology, 149152 CD45+ cells, 160
Biological weapon, as staphylococcal enterotoxin B, CD46, 71
304 CD47, 167
Biopanning CD59, 71
AFM-based, 391393 CD154
magnetic bead-based, 392 CDC (complement-dependent cytotoxicity), 65,
Bispecific antibodies, 335337 6871, 322324
Bite (bi-specific T cell engager), 9, 336 CDCC (complement-dependent cell cytotoxicity),
Blinatumomab, 336 65, 68, 70
BLT mice, 159, 160164, 166167 CDR. See Complementarity-determining region
BLyS, 144 Certolizumab pegol, 89, 334
Bordetella pertussis, 34 Cetuximab, 12, 184
Borrelia, 27 Chemical shift mapping, 181183
Botulism toxin, 113114, 120 Chemokines, 7778, 8889
Brentuximab vedotin (Adcetris), 15, 334, 335 Chemokines receptor 9 (CCR9), 294, 295
Brucella abortus, 33 Chimeric antibodies, 11, 16, 314
Brugia malayi, 33 Chronic lymphocytic leukemia (CLL), 70
Classes of immunoglobulins, 10
Calicheamicin, 334 Classical pathway of complement activation,
Camelids, 54 6566
Cancer Clonality score, 356
allergic response against, 9294 ClonePix, 152
AllergoOncology, 94 Clostridium botulinum
antibody gene transfer for, 435436 antibotulinum monoclonal antibodies discovered
magic bullet creation for, 15 by phage display, 113114
Candida, 27 antibotulinum monoclonal antibodies discovered
Candida albicans, 18, 28, 31, 403404 by yeast display, 120
Capillary morphogenesis protein 2, 420 Clostridium difficile, 17, 114115
Carbohydrates, recognition of self, 5557 Clostridium perfringens epsilon toxin, 420421
CaroRx, 417 Clostridium tetani
Catumaxomab, 336 antibacterial monoclonal antibodies discovered
CCR5 receptor, 17, 53, 111, 161, 194, 198, 428, 433 by phage display, 115
CCR9 (chemokines receptor 9), 294, 295 passive antibody therapy, 4
CD2, 306 CMV. See Cytomegalovirus
CD4/CD4+ cells, 17, 53, 76, 112, 130 Coagulation, complement and, 6364
enterotoxins and, 303 Cocaine, 435
human immunodeficiency virus (HIV) and, Cocktails of antibodies, 13, 17, 1819
194195, 199200, 428, 433 Coincidence index, 356
humanized mice and, 163 Colliers de Perles, 365, 366, 369370
CD5+ cells, 163 Colorectal cancer, Erbitux (cetuximab) therapy for,
CD8/CD8+ cells, 70 12
dengue virus infection and, 262 Combinatorial library approaches, for humanization,
humanized mice and, 163164 330
in respiratory syncytial virus (RSV) disease, 224 Common cold viruses, IgE antibodies induced by,
response to CMV, 276, 278 82
staphylococcal enterotoxin B, 310 Complement
CD19+ cells, 163 activation, 30, 64
CD20, 15 alternative pathway, 66
CD21, 144 antibody-mediated, 6668
CD22, 332 classical pathway, 6566
CD23, 7780, 8384, 88 lectin pathway, 66
CD28, 306 antibody functions dependent on, 3031
INDEX 445

antibody therapy and, 6372 Coronaviruses


antibody-mediated activation, 6668 antiviral monoclonal antibodies discovered by
IgA, 6768 phage display, 112113
IgG, 6567 Middle East respiratory syndrome coronavirus
IgM, 6567 (MERS-CoV), 111, 112113
coagulation and, 6364 severe acute respiratory syndrome coronavirus
dengue vascular permeability syndrome (DVPS) (SARS-CoV), 111, 112
and, 261262, 264 Corynebacterium diphtheriae, passive antibody
homeostasis, role in, 63, 6465 therapy for, 4
inflammatory responses, 69 Cost of therapy, 12
manipulating Ab-mediated responses, 6970 Coxiella burnetii, 33
increasing complement response, 6970 CpG, B cell immortalization using, 130, 131133,
reducing negative effects of complement, 70 166
membrane attack complex (MAC), 68 Crossmab, 336
opsonization, 6869 Cryptococcus neoformans, 27, 28, 31, 35, 67, 323
overview of system, 6365 labeled antibodies against, 15, 400404
roles of, 6365 monoclonal antibody therapy, 18
Complementarity-determining region (CDR), 173 Cryptosporidium parva, 28
affinity maturation, 184185 CTLs (cytotoxic T lymphocytes), 224
antibody docking, 180181 CXCR4 receptor, 17, 111, 161, 194, 198
antibody gene rearrangements, 346347 Cyanovirin, 419
CDR-IMGT lengths for antibody humanization Cytogam, 280
by grafting, 366 Cytokine inhibitors, for staphylococcal enterotoxin
cytotoxic drug delivery and, 15 B treatment, 311
high-throughput DNA sequencing and, 346347, Cytokines. See also Chemokines; specific cytokines
355356 to amplify B cells, 143144
humanization and CDR grafting, 327330 antibodies targeting cytokines and their receptors,
abbreviated CDRS containing SDRs (specificity- 8889
determining residues), 328329 in antibody-dependent enhancement (ADE),
framework adaptation, 330 255257, 264
framework shuffling, 329330 humanized mice and, 167
human string content optimization, 329 immunocytokine (ICK), 335
main decision-making points, 328 in immunodeficient mice strains, 158
standard technology, 327328 staphylococcal enterotoxin B and, 306307,
superhumanization, 329 311
veneering/resurfacing, 328 Cytomegalovirus (CMV), 273284
lengths and posttranslational modifications, 55 anti-CMV antibody structure, 55
modeling, 174, 176181, 184 future challenges, 283284
recombination and, 175 immune response, 276279
structure, 910, 5055, 5759 latency, 276278
canonical structure of, 176178 overview, 273275
disulfides in, 5455 structure and life cycle, 275276
H3 structure, 5153 studies in humanized mice, 166
predicting conformation of HCDR3, 177178 target-agonist approach for antigen discovery in
synthetic phage antibody libraries, 107108 vaccine design, 136137
Complement-coated Ab transfer, 68 testing therapies for, 6
Complement-dependent cell cytotoxicity (CDCC), therapeutic antibodies, 280283
65, 68, 70 monoclonal antibodies, 282283
Complement-dependent cytotoxicity (CDC), 65, polyclonal CMV-IVIG, 280282
6871, 322324 table of antibody-based therapeutics, 281
Computational design of antibodies, 183187 Townes's strains, 279, 280
Computer-aided prediction of structure and design treatment spectrum for high-risk demographic
of function, 173187. See also Antibody design; groups, 274
Antibody modeling vaccine development, 279280
Constant domains, 810, 174 Cytotect IVIG, 282, 283
Contact-mode AFM, 387 Cytotoxic drugs, antibody-delivered, 15
Convertases, 68 Cytotoxic T lymphocytes (CTLs), 224
446 INDEX

Daclizumab, 330 DNA sequencing. See High-throughput DNA


DC-SIGN, 326 sequencing
Dendritic cells, 7678, 166, 256, 326 DOCK8, 81
Dengue hemorrhagic fever/dengue shock syndrome Drug addiction, antibody gene transfer, 435
(DHF/DSS), 251253, 261262, 264 Duobody of Genmab, 324
Dengue vascular permeability syndrome (DVPS), DVPS. See Dengue vascular permeability syndrome
251253, 261, 263265
clinical features of, 251, 261 EBI (European Bioinformatics Institute), 364
genetic factors contributing to susceptibility, 252, Ebola, 56, 111, 113, 417418
263 EBV. See Epstein-Barr virus
sensitizing infection, 252254, 263 Effector functions of antibody, 1011
Dengue virus Effector silent antibodies, 325326
afferent phenomena, 252261, 263264 Efungumab, 18
enhancing antibodies, 252258, 263264 Ehrlichia risticii, 33
genetic host factors contributing to DVPS ELAM, 306
susceptibility, 252, 263 Electrofusion, in hybridoma production, 146148
role of dengue viruses in ADE, 258261, 264 Electron microscopy, antibody docking and, 181183
antibody-dependent enhancement (ADE), Electroporation, 146
249265 Enbrel (etanercept), 432
antiviral monoclonal antibodies Ensembl, 364
discovered by phage display, 111 Enterotoxins, staphylococcal, 303304. See also
discovered by yeast display, 118, 119 Staphylococcal enterotoxin B
dengue hemorrhagic fever/dengue shock Entrez Gene, 364
syndrome (DHF/DSS), 251253, 261262, Entry, inhibition of, 28
264 Enzyme-linked immunosorbent assay (ELISA), 382,
dengue vascular permeability syndrome (DVPS), 388
251253, 261, 263265 Eosinophilia, in allergic conditions, 8889
DENV-2 Eosinophils, 7879, 8889
enhanced growth in primary human monocytes, Epitope
258, 264 discontinuous, 186
genetic differences after infection with DENV-1, IMGT (International ImMunoGeneTics
259261, 264 information system), 374376
genetic differences after infection with DENV-3, Epitope drift, 330
260261, 264 Epitope grafting, 185187
strain differences in neutralization by DENV-1 Epitope mapping
antibodies, 258259, 264 antibody docking and, 180183
efferent phenomena, 252, 253, 261, 264265 HIV neutralizing antibodies, 429
enhancing antibodies, 252258 Epsilon toxin, Clostridium perfringens, 420421
attributes of iADE, 254257, 263 Epstein-Barr virus (EBV)
role of FcRs, 257258, 264 B cells immortalization using, 130, 131133,
role of infection sequence, 257, 263264 143145, 148, 283
role of myloid cells, 257, 264 high-throughput DNA sequencing applications
sensitizing infection, 252254, 263 for study of, 356
heterohybridomas, 149 studies in humanized mice, 161164
studies in humanized mice, 162 Erbitux (cetuximab), 12
Dental caries monoclonal antibodies, 416417 ERK, 255
Dependovirus, 429 Error correction strategies, for high-throughput
Design. See Antibody design DNA sequencing, 353
Destabilization of organisms, 26 Escherichia coli, 17, 90, 327
DHF/DSS. See Dengue hemorrhagic fever/dengue adhesion, interference with, 27
shock syndrome bacteriophages, 105
Dielectrophoresis, 146147 enteropathogenic, 27
Diphtheria, history of serum therapy for, 4 uropathogenic, 27
Directed evolution, 184 Esterification, in atomic force microscopy (AFM),
Dissociation constant, 386, 389391 388
Disulfide bonds, 8, 5455 Etanercept, 89
DJ recombination, 50 European Bioinformatics Institute (EBI), 364
INDEX 447

Evolution complement dependent, 3031


directed, 184 dependent on Fc-Fc receptor interactions, 3235
viral, 110 incentive for measuring, 25
independent of effector cells/molecules, 2629
Fab inflammation modulation, 3536
Fab-pIII phage display format, 109 neutralization, 2629
structure, 9, 50, 180 overview of, 2526
yeast display and, 115117 phagocytosis, 3435
FACS (fluorescence-activated cell sorting), 115117, Fungi
382, 384386, 390391, 394395 antibody-based therapy examples, 18
Factor VII, 64 radioimmunotherapy (RIT) for fungal infections,
Fc 400404
aglycosylated, 327 yeast display, 115121
antibody engineering, 1415, 322327 Fusion, inhibition of, 28
fusion proteins, 9, 16 Fusion efficiency, 142
glycosylation and effector functions, 324327 Fusion methods for hybridoma production, 145148
sialylation, 326327 electrical cytofusion, 146148
structure, 910, 50, 174 PEG, 145146
Fc, 7677, 79, 8288 viral, 145
Fc-Fc receptor interactions, antibody functions Fusion partners for hybridoma production,
dependent on, 3235 148149
FcRs Fusion proteins, 9, 16
allergic responses and, 7980 Fab-pIII phage display format, 109
antibody engineering, 322327 scFv-pIII phage display format, 108109
role in antibody-dependent enhancement (ADE), Fv structure, 50
249, 250, 254255, 257258, 264
FcRn (neonatal Fc receptor), 10, 15, 326 Galactose, yeast display and, 115
Feline infectious peritonitis virus, 250 Galectin-3, 77, 79
Filamentous phage, 105106 Ganciclovir, 277
Fimbriae, 27 Gel microdroplet encapsulation, 152
5' RACE, 350351 Gell-Coombs immunopathology, 249
Flaviviruses, 251 Gemtuzumab, 324
antiviral monoclonal antibodies discovered by Gemtuzumab ozogamicin, 334
phage display, 111 Gene rearrangements, antibody, 346348
antiviral monoclonal antibodies discovered by Genome Database, 364
yeast display, 118, 119 Giardia, 28
Flow cytometry Glycan shield, 56, 194
fluorescence-activated cell sorting (FACS), GlycoFi, 325
115117, 382, 384386, 390391, 394395 Glycosylation
hybridoma cell sorting and, 151 Fc, 324327
Fluorescence-activated cell sorting (FACS), 115117, of plant-derived monoclonal antibodies, 415416
382, 384386, 390391, 394395 Glycotope, 325
FMDV (foot-and-mouth disease virus), 26, 430431, Golimumab, 89, 334
435 Graft versus host disease (GVHD), 159
Food poisoning, staphylococcal enterotoxin B, Guided selection, 330
309 Guy's 13, 416417
Foot-and-mouth disease virus (FMDV), 26,
430431, 435 HAART (highly active antiretroviral therapy),
Foravirumab, 18 405406
Framework adaptation, 330 Haemophilus influenzae, 31
Framework shuffling, 329330 history of serum therapy for, 4
Fucosylation, 324325 hyper-IgE syndrome, 81
Functions of antibodies, 2536 vaccine, 357
agglutination, 26 Half-life, immunoglobulin, 10
aggregation, 26 Hamming distance, 354
antibody-dependent cellular cytotoxicity HCDR3, modeling, 177180
(ADCC), 3234 Heat shock proteins, 18, 27
448 INDEX

Heavy chains instruments, 348350


antibody gene rearrangements and, 346348 overview, 345346
domination of antibody-antigen interaction, strategies for Ig repertoires, 350354
5354 5' RACE, 350351
paired heavy and light chain sequencing, 354 cell populations, 350
in scFv-pIII phage display format, 108 error correction strategies, 353
structure, 810, 50, 5354, 174175 multiplexing and chimeric sequences, 352353
Helper phage, 108109 paired heavy and light chain sequencing, 354
Hemagglutinin replicate library preparation, 353
antibodies against, 28, 58, 111, 119, 132135, targeted PCR, 350351
210216 template choice, 351352
antibodies targeting residues mediating receptor HighV-QUEST, 355
specificity, 213214 Histoplasma capsulatum, 15, 403404
antibodies that mimic sialic acid, 214215 History of antibody-based therapy
antibody response in humanized mice, 162 monoclonal antibodies, 1112
characteristics of globular head versus stem HA passive antibody therapy, 45
antibodies, 210 HIV. See Human immunodeficiency virus
cross-reactivity of globular head HA antibodies, HLA
212213 CMV and, 278, 283
future areas for study, 215216 humanized mice and, 164, 166, 167
antigenic mapping, 211212 HMPV. See Human metapneumovirus; Human
structure, 110111, 209211 metapneumovirus (HMPV)
Hematopoietic stem cells Homeostasis, complement and, 63, 6465
Hu-HSC mice, 159, 161164, 166167 Hooke's law, 388
Hu-Liver-HSC mice, 160 HRIG (human antirabies immunoglobulin), 419
Hemolytic uremic syndrome, 17 HSV. See Herpes simplex virus
Hepatitis B virus HTLV-1, 166
combinations of antibodies against, 13 Hu-HSC mice, 159, 161164, 166167
studies in humanized mice, 162, 166 Hu-Liver-HSC mice, 160
Hepatitis C virus, 17 Human antirabies immunoglobulin (HRIG), 419
monoclonal antibodies against, 149 Human Genome Organization (HUGO), 364
studies in humanized mice, 166 Human Genome Organization Nomenclature
Herceptin (trastuzumab), 325, 330, 335, 336, 436 Committee (HGNC), 364
Herd immunity, respiratory syncytial virus (RSV) Human immunodeficiency virus (HIV)
and, 222 antibody response to, 193196
Herpes simplex virus (HSV) conventional antibodies, 195
antibody-dependent cellular cytotoxicity neutralizing antibodies, exceptional broadly,
(ADCC), 3233 195196
antiviral monoclonal antibodies discovered by pregnancy and, 196
phage display, 111 virus escape mechanisms, 194195
plant-derived monoclonal antibody, 418 antibody-dependent cellular cytotoxicity
studies in humanized mice, 166 (ADCC), 33
Heterohybridomas, 148149 anti-HIV antibody structure, 49, 53, 54, 57, 183,
HGNC (Human Genome Organization Nomenclature 186
Committee), 364 attachment, interference with, 27
Highly active antiretroviral therapy (HAART), combinations of antibodies against, 13
405406 complement and, 30
High-throughput DNA sequencing, 345357 conformational masking, 194195
antibody gene rearrangements, 346348 glycan shield, 56, 194
data analysis, 354357 HAART (highly active antiretroviral therapy),
alignment and parsing programs, 354355 405406
applications in infectious disease research, high-throughput DNA sequencing applications
355357 for study of, 356
primer trimming, 354 IgG3 and, 324
quality scores, 354 incidence and prevalence, 193
sequence barcode analysis, 354 monoclonal antibody against, 17, 130131
future directions, 357 discovered by phage display, 112
INDEX 449

discovered by yeast display, 118119 Humanized mice, 157168


mutation rate, 194 antigens studied in, 166
neutralization of infectivity, 2629, 58, 185186 future prospects, 167168
neutralizing antibodies HLA restriction, 163164, 164, 166, 167
exceptional broadly, 195196 human cell reconstitution and antibody response,
importance of, 428429 162163
as vaccine, 429 human immunodeficiency virus (HIV) studies,
plant-derived monoclonal antibody, 418419 158159, 161164, 166
radioimmunotherapy (RIT), 405407 immunodeficient mouse strains, 158, 160
self-carbohydrate recognition by HIV antibodies, monoclonal antibody generation in, 164166
5657 mouse models, 158160
somatic hypermutation in immune systems advantages and disadvantages, 166167
chronically exposed to, 348 BLT mice, 159, 160164, 166167
studies in humanized mice, 158159, 161164, Hu-HSC mice, 159, 161164, 166167
166 Hu-Liver-HSC mice, 160
target-agonist approach for antigen discovery in Hu-PBL mice, 158159
vaccine design, 136 SCID-Hu mice, 159, 163
vaccine development and immunogen design, pathogens studied in, 166
196200, 427429 preparation, infection, and immunization,
Abs targeting CD4 binding sites, 199200 160162
Abs targeting membrane-proximal external T-cell response in, 163164
region (MPER), 200 Humira (adalimumab), 11
Abs targeting variable loops 1 and 2, 197198 Hu-PBL mice, 158159
Abs targeting variable loops 3, 198199 Hybridoma, 12
Human metapneumovirus (HMPV), 132133, fusion methods, 145148
237244 electrical cytofusion, 146148
age-related antibody development, 239 PEG, 145146
antibody cross-protection, 238239 viral, 145
antibody response to infection, 239 heterohybridomas, 148149
antibody specificity, 238 history, 141142
antigenic proteins, 242243 human hybridoma technology, 141152
F protein, 239, 241244 advantages of, 141142
immunoglobulin classes after infection, 239 B cell source, 143145
induction of antibodies by immunization, biological cloning methods, 149152
241242 disadvantages of, 142
monoclonal antibodies, 243244 first human hybridomas, 142143
phylogeny, 240 fusion methods, 145148
seroprevalence of infection, 237238 fusion partners, 148149
Human papillomavirus, 29 trioma, 149
Humanization Hydrogen-deuterium exchange, 181183
antibody engineering, 327330 Hyper-IgE syndrome (HIES), 82
antibody informatics and, 370 Hypersensitivity
CDR grafting, 327330 allergic inflammation, 7595
abbreviated CDRS containing SDRs (specificity- serum therapy and, 4, 1011
determining residues), 328329 Hypoxanthine, 148
framework adaptation, 330
framework shuffling, 329330 iADE (intrinsic antibody-dependent enhancement),
human string content optimization, 329 251, 254258, 263264
main decision-making points, 328 Ibalizumab, 17
standard technology, 327328 Ibritumomab (Zevalin), 15, 334, 399
superhumanization, 329 Ichthyophthirius multifilis, 26
veneering/resurfacing, 328 ICK (immunocytokine), 335
CDR-IMGT lengths for antibody humanization IFN. See Interferon
by grafting, 366 IgA
combinatorial library approaches, 330 agglutination and, 26
guided selection, 330 antibody engineering, 322323
overview, 327 complement activation, 6768
450 INDEX

IgA (continued) Fc sialylation, 326327


half-life, 10 half-life, 10
human metapneumovirus infection, 239240 human metapneumovirus infection, 239240
plant-derived monoclonal antibody, 414, 416 IgG4, allergen specific, 9092
respiratory syncytial virus (RSV) monoclonal plant-derived monoclonal antibody, 414, 416
antibody, 227 respiratory syncytial virus (RSV) monoclonal
rotavirus and, 291299 antibody, 227228
IgBlast, 355 rotavirus, 290295
IgD, rotavirus and, 291 structure, 910, 173175
IgE, 7595 subtypes, 67
allergic inflammation, 7595 IgM
antibodies in treatment of allergies, 8292 antibody response humanized mice, 162163, 166
clinical perspective, 7576 complement activation, 6567
IgE activation of immune response, 96 half-life, 10
IgE antibodies in infectious diseases, 8082 rotavirus and, 291293, 299
IgE effector cells, 7779 iHMMunealign, 355
APCs, 78 IL-1, 306
B cells, 78 IL-2, 158, 164, 306307, 312, 314
mast cells, 7778 IL-4, 255
monocytes and eosinophils, 7879 IL-4, antibodies targeting, 88
IgE receptors, 7677 IL-5, antibodies targeting, 8889
interactions between allergens, IgE, and Fc IL-6, 134, 257, 307
trigger hypersensitivity responses, 79 IL-9, antibodies targeting, 89
allergic response against cancer, 9294 IL-10, 9092, 255257, 263
allergy treatment, antibodies in, 8292 IL-12, 255, 307
antibodies targeting cytokines, chemokines, and IL-13, antibodies targeting, 88
their receptors, 8889 IL-21, 130, 144
clinical studies in 2012, 8687 Illumina high-throughput DNA sequencing
future roles of antibodies in therapy, 9495 instruments, 349350
IgG4 triggered protective immunity with IMGT (International ImMunoGeneTics information
immunotherapy, 9092 system), 348, 363377
neutralizing antibody response induced by active antibody 3D structures, 373376
allergen immunotherapy, 8990 IMGT/2Dstructure-DB, 376
recombinant antibodies targeting IgE interactions IMGT/3Dstructure-DB card, 373
with Fc receptors, 8288 IMGT/3Dstructure-DB chain details, 373
enterotoxin induction of, 310 IMGT/3Dstructure-DB contact analysis, 374
half-life, 10 IMGT/DomainSuperimpose, 376
IgE antibodies in infectious diseases, 8082 IMGT/mAb-DB, 376
bacterial infections, 81 IMGT/StructureQuery, 376
parasitic infections, 8081 paratope and epitope, 374376
viral infections, 82 antibody engineering, importance of, 370
IgE receptors, 7677 availability and citation, 377
IgG Colliers de Perles, 365, 366, 369370
allergen-specific IgG antibody triggering humanization, importance of, 370
hypersensitivity responses, 7980 IMGT/DomainGapAlign, 370373
antibody engineering IMGT/LIGM-DB, 376
Fc glycosylation and effector functions, 324327 nomenclature, 364
IgG1, 323 overview, 363374
IgG2, 323324 paratope and epitope, 374376
IgG3, 324 standardized and integrated system, 366367
IgG4, 324 targeted and customized therapeutic antibodies,
antibody response humanized mice, 162163, 166 376377
antibody-dependent cellular cytotoxicity (ADCC) unique numbering, 364366
and, 322326 anchors for V and C domains, 365366
complement activation, 6567 CDR-IMGT lengths for antibody humanization
complement-dependent cytotoxicity (CDC) and, by grafting, 366
322324 highly conserved amino acids, 366
INDEX 451

V- and C-domain 2D representations, 369 hemagglutinin


V- and C-domain amino acid sequence analysis, antibodies against, 28, 58, 111, 119, 132135, 162,
370373 210216
V-domain nucleotide sequence analysis, 367369 structure, 110111, 209211
IMGT/HighV-QUEST for deep sequencing, heterohybridomas, 149
367369 inhibition of fusion/entry, 28
IMGT/V-QUEST, 367 inhibition of liberation, 29
IMGT-ONTOLOGY, 364, 366367 intracellular inhibition, 29
Immune complex formation, 4 monoclonal antibodies against, 149
Immune system discovered by phage display, 110112
allergic inflammation, 7595 discovered by yeast display, 118, 119
modulation by antibodies, 3536 screening monoclonal antibodies by neutralization,
transplantation of human into mice, 158162 132133
Immunization, for human metapneumovirus neutralization mechanism, 58
(HMPV), 241242 Informatics, antibody, 363377
Immunoadhesins, 431434 analysis of V-domain nucleotide sequences,
Immunocytokine (ICK), 335 367369
Immunoglobulin. See also Antibody; specific antibody 3D structures, 373376
immunoglobulin classes antibody engineering, importance of, 370
antibody gene rearrangements, 346348 humanization, importance of, 370
high-throughput DNA sequencing strategies for nomenclature, 364
Ig repertoires, 350354 overview, 363374
Immunotherapy standardized and integrated system, 366367
cytomegalovirus (CMV) targeted and customized therapeutic antibodies,
monoclonal antibodies, 282283 376377
polyclonal CMV-IVIG, 280282 unique numbering, 364366
vaccine, 279280 V- and C-domain 2D representations, 369
human metapneumovirus (HMPV) V- and C-domain amino acid sequence analysis,
induction of antibodies by immunization, 370373
241242 Innate immune system, complement functions in,
monoclonal antibodies, 243244 63
radioimmunotherapy (RIT), 399408 iNOS (inducible nitric oxide synthase), 256
respiratory syncytial virus (RSV), 225231 Interferon (IFN)
intravenous immunoglobulin (IVIG), 225226 antibody-dependent enhancement (ADE) and,
monoclonal antibody, 226231 250, 255257, 263
rotavirus, 296298 staphylococcal enterotoxin B and, 306307, 310,
staphylococcal enterotoxin B, 311315 314315
passive immunotherapy, 313315 Interleukins. See also specific interleukins
vaccines, 312313 allergic inflammation and, 7678
Indels, unusual, 57 to amplify B cells, 144
Inducible nitric oxide synthase (iNOS), 256 antibodies targeting, 8889
Infectivity, neutralization of, 26 humanized mice and, 167
Inflammation. See also Allergic inflammation hyper-IgE syndrome and, 81
complement, 69 in immunodeficient mice strains, 158
Fc sialylation and, 326327 staphylococcal enterotoxin B and, 306307, 312,
modulation by antibodies, 3536 314
in respiratory syncytial virus (RSV) disease, 224 Intermittent contact-mode AFM, 388
Infliximab, 89, 330, 334 Internalization of antibodies, 29
Influenza virus International ImMunoGeneTics information
antibody gene transfer, 433434 system. See IMGT
antibody-dependent cellular cytotoxicity Intrabodies, 29
(ADCC), 33 Intravenous immunoglobulin (IVIG), 5
anti-influenza antibody structure, 49, 54, 57 for cytomegalovirus, 280282
complement and, 30 for respiratory syncytial virus (RSV), 225226
H1N1, 210214, 434435 Intrinsic antibody-dependent enhancement (iADE),
H2N2, 210 251, 254258, 263264
H5N1, 215, 435 Ion Torrent platform, 349
452 INDEX

JAK/STAT signaling, 256 Membrane-proximal external region (MPER), HIV,


Japanese encephalitis virus, 254, 263264 200
Jawless vertebrates, 5354 Memory B cells
Junctional diversity, 175, 184 efficient methods to isolate monoclonal
antibodies from, 129137
Kadcyla (ado-trastuzumab emtansine), 15 frequency in circulation, 142
Kaposi's sarcoma-associated herpesvirus, 166 immortalization, 130, 131133
Knobs-into-holes technology, 336 Meningitis, history of serum therapy for, 5
Mepolizumab, 88
Lebrikizumab, 88 Metapneumovirus. See also Human
Lectin pathway of complement activation, 66 metapneumovirus (HMPV)
Legionella pneumophilia, 31 avian, 238, 240
Leishmania, 30 phylogeny, 240
intrinsic antibody-dependent enhancement MHC. See Major histocompatibility complex
(iADE), 255, 258 Mice
studies in humanized mice, 166 heterohybridomas, 148149
Lentiviruses, antibody-dependent cellular humanized, 157, 168. See also Humanized mice
cytotoxicity (ADCC) and, 33 immunodeficient strains, 158, 160
Light chains monoclonal antibody production in, 130
antibody gene rearrangements and, 346348 staphylococcal enterotoxin B
paired heavy and light chain sequencing, 354 monoclonal antibodies for treatment of, 313314
in scFv-pIII phage display format, 108 mouse model, 307308
structure, 8, 50, 174175 Microscopy
Limiting dilution cloning, 150 atomic force microscopy (AFM), 382, 386389,
Lineweaver-Burke plot, 385 391395
Listeria moncytogenes, 29 electron microscopy, antibody docking and,
LocusLink, 364 181183
Lovastatin, 314 Middle East respiratory syndrome coronavirus
Lymphoblastoid cells, 144 (MERS-CoV), 111, 112113
Lysozyme-antilysozyme binding, 389 Modeling. See Antibody modeling
MOE modeling server, 179
Magic bullet creation, 15 Monoclonal antibody
Magnetic assisted cell sorting (MACS), 115116 anti-infectious, 65
Magnetic bead-based biopanning, 392 approved/pending therapies, 68, 65
Major histocompatibility complex (MHC) complement and, 67, 6871
CMV and, 278 cost of therapy, 12
humanized mice and, 162163, 167 defined, 11
staphylococcal enterotoxin B and, 304308, 311, history, 1112, 141142
313 hybridoma and, 12
Mammalian display, affinity maturation of antibody immunotherapy
by, 332333 cytomegalovirus (CMV), 282283
Mannose-activating serine proteases (MASPs), 66 human metapneumovirus (HMPV), 243244
Mannose-binding lectin (MBL), 66 respiratory syncytial virus (RSV), 226231
MARMs (monoclonal antibody resistant mutants), neutralization of infectivity and, 2627
243244 phage display, 106, 109115
Mass spectromtery, staphylococcal enterotoxin B antibacterial mAbs discovered, 113115
and, 310 antiviral mAbs discovered, 110113
Mast cells, 7778 plant-derived, 413421
Mating type, yeast, 115 production, 12
Maytansinoids, 335 efficient methods to isolate from memory B cells
MCP-1 (monocyte chemoattractant protein 1), and plasma cells, 129137
307308 high-throughput cellular screens, 130135
Mean fluorescence intensity (MFI), 385386 human hybridoma technology, 141152
Measles virus in mice, 130, 157168
IgE antibodies induced by, 82 in plants, 413416
inhibition of, 29 single-cell reverse transcription (RT)-PCR, 130,
Membrane attack complex (MAC), 68 133, 135
INDEX 453

specificity of, 12, 13 in immunodeficient mice strains, 158


staphylococcal enterotoxin B, 313315 NMR spectroscopy, 181183
chicken, 314 NOG mice, 158160
chimeric, 314 Nonobese diabetic (NOD)-SCID mice, 158, 160, 161
human, 314315 NS1, dengue virus, 260262, 264265
murine, 313314 NSG mice, 158161
yeast display, 118121 Nude mice, 158
antiviral mAbs discovered, 118119 Numax (motavizumab), 17
bacterial mAbs discovered, 119121
Monoclonal antibody resistant mutants (MARMs), OKT3, 1112, 283
243244 Omalizumab (Xolair), 8388
Monocyte chemoattractant protein 1 (MCP-1), Opsonization, 6869
307308 OraVax, 227
Monocytes, 7879 Ouabain, 148
Motavizumab/motavizumab-YTE, 228229, 231
MPER (membrane-proximal external region), HIV, Pagibaximab, 16
200 Palivizumab, 6, 17, 18, 65, 137, 227231, 321, 434
Murray Valley encephalitis virus (MVEV), 250 Panobacumab, 16
Mutagenesis, affinity maturation and, 331333 Parainfluenza virus, 30
Mutations, escape in antibody therapy, 13 Paramyxoviruses, 30, 222
Mycobacterium tuberculosis, 27 Parasites
Myeloma, hybridoma production and, 148149 antibody-dependent cellular cytotoxicity
Myoltarg (gemtuzumab ozogamicin), 15 (ADCC), 33
complement and, 31
NALP3 inflammasome complex, 72 IgE mechanisms of protection in parasitic infections,
Nanobody, 231 8081
Nanoparticles, 71 interference with attachment, 28
Natalizumab, 324 phagocytosis, 35
National Center for Biotechnology Information replication inhibition by IgA, 29
(NCBI), 364 Passive antibody therapy. See also Monoclonal
Neisseria gonorrhoeae, 31 antibody
Neisseria meningitidis, 4, 31, 33, 35 antibody gene transfer, 429436
Neonatal Fc receptor (FcRn), 10, 15, 326 for cytomegalovirus, 280283
Neuraminidase, antibodies against, 29, 58 future roles in allergic disease treatment, 94
Neutralization, 2629 history of, 45
antibody design, 184 for HIV, 428429
complement and, 65 intravenous immunoglobulin (IVIG), 5, 225226,
defined, 26 280282
inhibition of fusion/entry, 28 for respiratory synctial virus, 225226
interference with attachment, 2728 for rotavirus, 296297
intracellular, 29 side effects, 4, 1011
mechanisms of, 5758 staphylococcal enterotoxin B, 313315
by monoclonal antibodies discovered by phage Pathogenicity islands, 304
display, 110 PCR, 350354
multiple hit phenomenon, 27 Pearl chain formation, 147
neutralizing antibody elicited through Penicillin, discovery of, 5
antigen design, 185 Pentoxifylline, 311
epitope grafting, 185187 Peptide antagonists, for staphylococcal enterotoxin
neutralizing antibody response induced by active B treatment, 311
allergen immunotherapy, 8990 Permeabilization, 146
postattachment, 2829 Phage. See also Phage display
preattachment, 2627 described, 105106
screening by, 132133 helper, 108109
Nicotiana, 415421 recovery using AFM, 394
NK cells Phage display, 105115, 129130, 184
antibody-dependent cellular cytotoxicity (ADCC) antibody display formats, 108109
and, 322, 323, 325, 326 Fab-pIII display, 109
454 INDEX

Phage display (continued) Pneumococcal vaccine, 357


scFv-pIII display, 108109 Pneumovirinae (subfamily), 222
discovery of antibodies for infectious diseases by, Poisson distribution, 150
109115 Poliovirus
antibacterial antibodies, 113115 aggregation of, 26
antiviral antibodies, 110113 inhibition of vesicle escape, 28
library selection procedure, 106107 Polyethylene glycol (PEG)-mediated cell fusion,
overview, 105106 142, 145146
purpose of, 106 Polymerase chain reaction (PCR), 350354
sources of antibody genes for display libraries, Posttranslational modification, 5053
106108 PREVENT study, 226
immune phage antibody libraries, 106107 PRO140, 17
nave phage antibody libraries, 106 Programs, antibody modeling, 179180
synthetic phage antibody libraries, 107108 Protein Data Bank, 49, 175176, 178180
yeast display compared, 116117 Pseudomonas aeruginosa, 1617, 26
Phagemid vectors, 108
Phagocytosis Quadroma technology, 336
antibody function and, 3435 Quality scores, DNA sequencing, 354
antibody-dependent cellular phagocytosis
(ADCP), 322323, 325 Rabies, 1718, 419
Pichia pastoris, 90 RACE (rapid amplification of cDNA ends), 350351
PIGS server, 179 Radioimmunoassay (RIA), 382
PIII phage antibody display Radioimmunotherapy (RIT), 399408
Fab-pIII, 109 bacterial infections, 404405
scFv-pIII, 108109 fungal infections, 400404
Pili, 27 HIV, 405407
Plant-derived monoclonal antibody, 413421 overview, 399400
bacterial toxins, 420421 Radioisotopes
anthrax, 420 antibody drug conjugates, 334
Clostridium perfringens epsilon toxin, 420421 labeled antibodies, 15, 399408
staphylococcal enterotoxin B, 421 Rapamycin, 311
infectious disease targets, 416421 Rapid amplification of cDNA ends (RACE), 350351
dental caries, 416417 Raxibacumab, 17, 114
Ebola virus, 417418 Recombinant adeno-associated virus (rAAV),
herpes simplex virus, 418 428436
human immunodeficiency virus (HIV), 418419 Recombination, 50, 174, 184
rabies, 419 Resilizumab, 89
respiratory synctial virus (RSV), 419 RespiGam, 226
in vivo studies, 417 Respiratory diseases. See also specific etiologic agents
West Nile virus, 419420 antibody gene transfer for, 433435
production, 413416 staphylococcal enterotoxin B and, 310
controlling Mab N glycosylation, 415416 Respiratory syncytial virus (RSV), 221232
rationale, 413414 antibody gene transfer, 433434
transgenic technologies for, 414 antiviral monoclonal antibodies discovered by
transient technologies for, 415 phage display, 111
for purification of infectious disease antigens, 421 clinical features of infection/disease, 221222
Plasma cells combinations of antibodies against, 13
efficient methods to isolate monoclonal epitope grafting, 186187
antibodies from, 129137 F protein, 222223, 225, 227, 239, 242244
long-term culture, 133134 future opportunities for treatment and prevention,
Plasmodium, 28 231232
Plasmodium falciparum, 28, 31 herd immunity, 222
ADCC, 33 IgE antibodies induced by, 82
phagocytosis, 35 immunoprophylaxis, 225231
studies in humanized mice, 166 intravenous immunoglobulin (IVIG), 225226
Platforms, antibody modeling, 179180 monoclonal antibody, 226231
Pneumococcal pneumonia, serum therapy for, 4 motavizumab/motavizumab-YTE, 228229, 231
INDEX 455

overview, 221224 SEED (strand-exchange engineered domain), 336


palivizumab for, 6, 17, 18, 65, 137, 227231, 321 Self-carbohydrate recognition, 5557
pathogenesis, 223 Semliki Forest virus, 30
plant-derived monoclonal antibody, 419 Sendai virus
ribavirin for, 224 for fusion in hybridoma production, 145
screening monoclonal antibodies by neutralization, inhibition of, 29
132 Sensitizing infection, in dengue vascular
structure and life cycle, 222223 permeability syndrome (DVPS), 252254, 263
target-agonist approach for antigen discovery in Serum sickness, 4
vaccine design, 137 Serum therapy
vaccine, 224225 history of, 45
Resurfacing, 328 pros and cons of antibody-based therapy relative
Reverse vaccinology, 198 to, 14
RG mice, 158160, 162, 164 for toxin-mediated diseases, 311
Rhinitis, staphylococcal enterotoxin B and, 310 Servers, antibody modeling, 179180
Rhinovirus Severe acute respiratory syndrome coronavirus
IgE antibodies induced by, 82 (SARS-CoV), 111, 112
interference with attachment, 27 Severe combined immunodeficiency (SCID) mice,
RIA (radioimmunoassay), 382 158161, 163, 406407
Ribavirin, 224 Sharks, 54
RIT. See Radioimmunotherapy Shiga toxin, 17
Rituximab (Rituxan), 11, 70, 325 Shigella flexneri, 33
RNA interference (RNAi), 231 Sialic acid, antibodies that mimic, 214215
Roche/454 platform, 349 Sialylation, Fc, 326327
Root mean square deviation (RMSD), 180181 Silanization, in atomic force microscopy (AFM), 388
Rosetta Design, 186 Simian immunodeficiency virus (SIV), 428
Rosetta Antibody, 179 Sindbis virus, 26
Rosetta Dock algorithm, 180181 Single-cell reverse transcription (RT)-PCR, 130, 133,
Ross River virus (RRV), 250, 257 135
Rotarix vaccine, 297 Single-chain variable fragments (scFv)
Rotavirus, 289299 scFv-pIII phage display format, 108109
antibody response against, 290296 yeast display and, 115117
compartmentalization of rotaviris-B cells, SIT (specific immunotherapy), 9092
293295 Site-directed mutagenesis, 181183, 312313
Ig gene usage of rotavirus-B cells, 292293 SnugDock algorithm, 181
mechanism of protection and specificity of SoDA2, 355
rotavirus-Ig, 295296 Somatic hypermutation, 175, 184
ontology of rotavirus-B cells and rotavirus-Ig, antibody gene rearrangements, 348
290292 in vitro affinity maturation by, 332
future challenges, 298299 Somatic recombination, 175
overview, 289290 Specific immunotherapy (SIT), 9092
therapeutic/prophylactic applications of rotavirus SPR (surface plasmon resonance), 382384,
antibodies, 296298 389390, 394
RSV. See Respiratory syncytial virus Spring constant, 388
Staphylococcal enterotoxin B, 303315
Saccharomyces cerevisiae, 115 animal models, 307309
Salmonella, 33 mouse, 307308
Salmonella enterica serovar Typhimurium, 26, 166 nonhuman primate, 308309
Scaffold, epitope grafting and, 186187 piglet, 308
scFv. See Single-chain variable fragments rabbit, 308
scFv-pIII display, 108109 rat, 308
Schistosoma haematobium, 80 biological and pathological activities, major, 307
Schistosoma mansoni, 33, 80 as biological weapon, 304
SCID-Hu mice, 159, 163 clinical manifestation and epidemiology, 309310
SEBILS (SEB-induced lethal shock), 307308, atopic dermatitis, 309310
311312, 314 food poisoning, 309
Secretory IgA, plant-derived monoclonal, 414 respiratory diseases, 310
456 INDEX

Staphylococcal enterotoxin B (continued) Surface biomembrane force probe, 389


toxic shock syndrome, 309 Surface plasmon resonance (SPR), 382384,
ulcerative colitis, 310 389390, 394
description of agent, 304306 Sym003, 231
diagnosis, 310 Synagis, 6, 12, 17, 18, 321, 419, 434. See also
future challenges, 315 Palivizumab
immune cell interaction with, 306307 Synthetic phage antibody libraries, 107108
infection, 304307
overview, 304 T cell receptor (TCR)
plant-derived monoclonal antibody, 421 CMV and, 278279, 283
SEBILS (SEB-induced lethal shock), 307308, diversity estimates, 357
311312, 314 informatics and, 363364, 370
treatment, 310315 staphylococcal enterotoxin B and, 304307, 313, 314
cytokine inhibitors, 311 T cells
immunotherapy, 311315 antibody-dependent enhancement (ADE) and,
peptide antagonists, 311 255256
V domains, 311 cytotoxic T lymphocytes (CTLs) in respiratory
Staphylococcus aureus. See also Staphylococcal syncytial virus (RSV) disease, 224
enterotoxin B response in humanized mice, 159, 163164
antibacterial monoclonal antibodies discovered staphylococcal enterotoxin B and, 306307
by phage display, 115 T helper (Th) cells, 76, 78, 224, 255256, 310
complement evasion, 71 Talizumab (TNX-901), 83
enterotoxins, 303315 Tapping mode AFM, 388
hyper-IgE syndrome, 81 T-DM1, 335
methicillin-resistant (MRSA), 5, 16, 65, 115 Tefibazumab, 16
single-chain variable fragments (ScFv) targeting, 16 Terminal deoxynucleotidyltransferase (TdT), 363
vancomycin-resistant, 5 TERT, 149
STAT3, 81 Tetanus toxoid, 162, 166
STAT5, 164 Th17 cells, 81
Strand-exchange engineered domain (SEED), 336 Thravixa, 17
Streptavidin-biotin interaction, 388389 Thymidine, 148
Streptococcus, history of serum therapy for, 4 Tissue remodeling, 7879
Streptococcus mutans, 16, 416417 TNF-. See Tumor necrosis factor alpha
Streptococcus pneumoniae, 33, 3435, 67 Toll-like receptors
history of serum therapy for, 4 complement and, 63
hyper-IgE syndrome, 81 monoclonal antibody production and, 132133,
labeled antibodies against, 15 143, 166
monoclonal antibodies against, 149 Tositumumab, 15, 334, 399
radioimmunotherapy (RIT), 404405 Total internal reflection (TIR), 382383
Structure of antibodies, 810, 4959 Toxic shock syndrome (TSS), 306, 309
antibody gene rearrangements and, 346348 Toxic shock syndrome toxin 1 (TSST-1), 166, 303,
basics of, 810, 5051 308, 309, 312, 313
CDR H3, 5153 Toxins
computer-aided prediction of structure and antibacterial monoclonal antibodies discovered
design of function, 173187 by phage display, 113115
disulfides in CDRs, 5455 passive antibody therapy, 4
glycan shield, 5557 plant-derived monoclonal antibody for bacterial
heavy chain domains, 5354 toxins, 420421
mechanisms of antibody neutralization and, 5758 staphylococcal enterotoxin B, 303315
posttranslational modification, 5053 Toxoplasma gondii, 29
recombination and, 50 Tralokinumab, 88
relation of sequence, structure, and function, 173175 Trastuzumab (Herceptin), 325, 330, 335, 336, 436
self-carbohydrate recognition, 5557 Trichinella spiralis
unusual indels, 57 ADCC, 33
Substance P, 309 IgE and, 8081
Superantigens, enterotoxins as, 303305, 315 interference with attachment, 28
Superhumanization, 329 Trioma, 149
INDEX 457

Trypanosoma brucei, 31 Virus. See also specific viruses


TSS (toxic shock syndrome), 306, 309 antibody-based therapy examples, 1718
TSST-1 (toxic shock syndrome toxin 1), 166, 303, antiviral monoclonal antibodies discovered by
308, 309, 312, 313 phage display, 110113
Tumor necrosis factor alpha (TNF-), 77 antiviral monoclonal antibodies discovered by
antibodies targeting, 89, 333334 yeast display, 118119
staphylococcal enterotoxin B and, 306307, 312, evolution, 110
313, 315 for fusion in hybridoma production, 145
IgE antibodies induced by, 82
Ulcerative colitis, staphylococcal enterotoxin B and, neutralization, 2629, 110
310 phagocytosis, 34
Ultrasound, electroacoustic fusion and, 146 plant-derived monoclonal antibody
Ebola virus, 417418
Vaccine herpes simplex virus, 418
against adhesins, 27 human immunodeficiency virus (HIV), 418419
cytomegalovirus (CMV), 279280 rabies, 419
high-throughput DNA sequencing applications respiratory synctial virus (RSV), 419
for study, 356357 West Nile virus, 419420
human immunodeficiency virus (HIV), 196200, Viruslike particles
427429 human metapneumovirus immunization with,
respiratory syncytial virus (RSV), 224225 239, 241, 243
reverse vaccinology, 198 rotavirus and, 290291, 293
rotavirus, 297298 V(D)J recombination, 50, 174, 184. See also
staphylococcal enterotoxin B, 312313 Antibody gene rearrangements
Vaccine design V-QUEST, 355
analytic vaccinology, 136, 137
target-agonist approach for antigen discovery, WAM modeling server, 179
136137 Wellcome Trust Sanger Institute, 364
Valortim, 17 West Nile virus, 28
Variable domains, 810, 174175 antibody-dependent enhancement (ADE), 250
antibody gene rearrangements and, 346348 antiviral monoclonal antibodies discovered by
rotavirus-B cells and, 292293 phage display, 111, 113
in scFv-pIII phage display format, 108 antiviral monoclonal antibodies discovered by
Vascular endothelial cell growth factor receptor-2 yeast display, 118, 119
(VEGFR2), 435 complement and, 30
V domains, for staphylococcal enterotoxin B dengue virus cocirculation with, 254
treatment, 311 plant-derived monoclonal antibody, 419420
VD recombination, 50 studies in humanized mice, 162, 163
Vector-mediated in vivo antibody expression, 427436 WHO-International Nonproprietary Names
antibody gene transfer (WHO-INN), 364
for cancer, 435436 World Health Organization-International Union of
drawbacks of rAAV, 436 Immunological Societies (WHO-IUIS), 364
for drug addiction, 435
expression systems for, 429432 Xolair (omalizumab), 8388
for HIV-positive individuals, 433 X-ray crystallography, 179
proof-of-concept studies in animal models,
432433 Yeast, 115
for respiratory tract infections, 433435 Yeast display, 115121
expression systems for antibody gene transfer, discovery of antibodies for infectious diseases by,
429432 117121
proof-of-concept studies in animal models, 432433 antibacterial antibodies, 119121
Veneering, 328 antiviral antibodies, 118119
Vertebrate Genome Annotation (Vega) Browser, 364 overview, 115116
Vesicular stomatitis virus, for fusion in hybridoma phage display compared, 116117
production, 145
Vibrio cholerae, 26 Zanamir, 58
Viral attachment, interference with, 2728 Zevalin (ibritumomab), 15, 334, 399

You might also like