Bookshelf NBK83689
Bookshelf NBK83689
html
We ship printed books within 1 business day; personal PDFs are available immediately.
Visit the National Academies Press online, the authoritative source for all books
from the National Academy of Sciences, the National Academy of Engineering,
the Institute of Medicine, and the National Research Council:
• Download hundreds of free books in PDF
• Read thousands of books online for free
• Explore our innovative research tools – try the “Research Dashboard” now!
• Sign up to be notified when new books are published
• Purchase printed books and selected PDF files
Thank you for downloading this PDF. If you have comments, questions or
just want more information about the books published by the National
Academies Press, you may contact our customer service department toll-
free at 888-624-8373, visit us online, or send an email to
[email protected].
THE INFECTIOUS
ETIOLOGY OF
CHRONIC DISEASES
Defining the Relationship,
Enhancing the Research, and
Mitigating the Effects
Workshop Summary
NOTICE: The project that is the subject of this report was approved by the Governing Board of
the National Research Council, whose members are drawn from the councils of the National
Academy of Sciences, the National Academy of Engineering, and the Institute of Medicine.
Support for this project was provided by the U.S. Department of Health and Human Services’
National Institutes of Health, Centers for Disease Control and Prevention, and Food and Drug
Administration; U.S. Agency for International Development; U.S. Department of Defense; U.S.
Department of State; U.S. Department of Veterans Affairs; U.S. Department of Agriculture;
American Society for Microbiology; Burroughs Wellcome Fund; Ellison Medical Foundation;
Pfizer; GlaxoSmithKline; and The Merck Company Foundation. The views presented in this
report are those of the editors and attributed authors and are not necessarily those of the funding
agencies.
This report is based on the proceedings of a workshop that was sponsored by the Forum on
Microbial Threats. It is prepared in the form of a workshop summary by and in the name of the
editors, with the assistance of staff and consultants, as an individually authored document. Sec-
tions of the workshop summary not specifically attributed to an individual reflect the views of
the editors and not those of the Forum on Microbial Threats. The content of those sections is
based on the presentations and the discussions that took place during the workshop.
International Standard Book Number 0-309-08994-8 (Book)
International Standard Book Number 0-309-52673-6 (PDF)
Library of Congress Control Number: 2004107939
Additional copies of this report are available from the National Academies Press, 500 Fifth
Street, NW, Lockbox 285, Washington, DC 20055; (800) 624-6242 or (202) 334-3313 (in the
Washington metropolitan area); Internet, http://www.nap.edu.
For more information about the Institute of Medicine, visit the IOM home page at:
www.iom.edu.
Copyright 2004 by the National Academy of Sciences. All rights reserved.
Printed in the United States of America.
The serpent has been a symbol of long life, healing, and knowledge among almost all cultures
and religions since the beginning of recorded history. The serpent adopted as a logotype by the
Institute of Medicine is a relief carving from ancient Greece, now held by the Staatliche Museen
in Berlin.
COVER: The background for the cover of this workshop summary is a photograph of a batik
designed and printed specifically for the Malaysian Society of Parasitology and Tropical Medi-
cine. The print contains drawings of various parasites and insects; it is used with the kind
permission of the Society.
The National Academy of Engineering was established in 1964, under the charter of
the National Academy of Sciences, as a parallel organization of outstanding engineers. It
is autonomous in its administration and in the selection of its members, sharing with the
National Academy of Sciences the responsibility for advising the federal government.
The National Academy of Engineering also sponsors engineering programs aimed at
meeting national needs, encourages education and research, and recognizes the superior
achievements of engineers. Dr. Wm. A. Wulf is president of the National Academy of
Engineering.
The Institute of Medicine was established in 1970 by the National Academy of Sci-
ences to secure the services of eminent members of appropriate professions in the exami-
nation of policy matters pertaining to the health of the public. The Institute acts under the
responsibility given to the National Academy of Sciences by its congressional charter to
be an adviser to the federal government and, upon its own initiative, to identify issues of
medical care, research, and education. Dr. Harvey V. Fineberg is president of the Insti-
tute of Medicine.
The National Research Council was organized by the National Academy of Sciences in
1916 to associate the broad community of science and technology with the Academy’s
purposes of furthering knowledge and advising the federal government. Functioning in
accordance with general policies determined by the Academy, the Council has become the
principal operating agency of both the National Academy of Sciences and the National
Academy of Engineering in providing services to the government, the public, and the
scientific and engineering communities. The Council is administered jointly by both Acad-
emies and the Institute of Medicine. Dr. Bruce M. Alberts and Dr. Wm. A. Wulf are chair
and vice chair, respectively, of the National Research Council.
www.national-academies.org
Liaisons
YVES BERGEVIN, Department of Child and Adolescent Health and
Development, World Health Organization, Geneva, Switzerland
ENRIQUETA BOND, President, Burroughs Wellcome Fund, Research
Triangle Park, North Carolina
EDWARD McSWEEGAN, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, Maryland
Staff
STACEY KNOBLER, Director, Forum on Microbial Threats
MARJAN NAJAFI, Research Associate
KATHERINE OBERHOLTZER, Research Assistant
vi
Staff
PATRICK KELLEY, Director
MONISHA ARYA, Policy Intern
HARRIET BANDA, Senior Project Assistant
ALLISON BERGER, Senior Project Assistant
STACEY KNOBLER, Senior Program Officer
MARJAN NAJAFI, Research Associate (through November 2003)
KATHERINE OBERHOLTZER, Research Assistant
LAURA SIVITZ, Research Associate
DIANNE STARE, Research Assistant/Administrative Assistant
vii
Reviewers
All presenters at the workshop have reviewed and approved their respective
sections of this report for accuracy. In addition, this workshop summary has been
reviewed in draft form by independent reviewers chosen for their diverse per-
spectives and technical expertise, in accordance with procedures approved by the
National Research Council’s Report Review Committee. The purpose of this in-
dependent review is to provide candid and critical comments that will assist the
Institute of Medicine (IOM) in making the published workshop summary as sound
as possible and to ensure that the workshop summary meets institutional stan-
dards. The review comments and draft manuscript remain confidential to protect
the integrity of the deliberative process.
The Forum and IOM thank the following individuals for their participation in
the review process:
Paul Eke, Centers for Disease Control and Prevention, Chamblee, Georgia
Charlotte Gaydos, Johns Hopkins University School of Medicine, Balti-
more, Maryland
Julie Parsonnet, Stanford University School of Medicine, Palo Alto, Cali-
fornia
David Relman, Veterans Administration Palo Alto Health Care System, Palo
Alto, California
Donald Silberberg, University of Pennsylvania School of Medicine, Phila-
delphia
ix
x REVIEWERS
The review of this report was overseen by Melvin Worth, M.D., Scholar-in-
Residence, National Academies, who was responsible for making certain that an
independent examination of this report was carried out in accordance with institu-
tional procedures and that all review comments were carefully considered. Re-
sponsibility for the final content of this report rests entirely with the editors and
individual authors.
Preface
xi
xii PREFACE
an infectious etiology (e.g., peptic ulcer disease with Helicobacter pylori, cervi-
cal cancer with several human papillomaviruses, Whipple’s disease with
Tropheryma whippeli, Lyme arthritis and neuroborreliosis with Borrelia
burgdorferi, AIDS with HIV). Evidence implicating microorganisms as etiologic
agents of chronic diseases with substantial mortality and morbidity impact, in-
cluding atherosclerosis and cardiovascular disease, diabetes mellitus, inflamma-
tory bowel disease, and a variety of neurological and neuropsychiatric diseases,
continues to mount.
Emerging infectious diseases are conceptualized as either newly identified or
appreciated illnesses, conditions, or well-recognized diseases that are newly at-
tributed to infection. Now, scientists are beginning to believe that a substantial
portion of chronic diseases may actually be associated with infection.
In an effort to identify cross-disciplinary aspects of the challenge of infec-
tious etiologies of chronic diseases, including inflammatory syndromes and can-
cer, the Institute of Medicine’s Forum on Microbial Threats hosted a two-day
workshop on October 21–22, 2002. The workshop, Linking Infectious Agents
and Chronic Diseases, explored the factors that drive infectious etiologies of
chronic diseases to prominence, and sought to identify more broad-based strate-
gies and research programs that need to be developed. The goals of the workshop
were to:
The issues pertaining to these goals were addressed through invited presenta-
tions and subsequent discussions, which highlighted ongoing programs and ac-
tions taken, and also identified the most vital needs in this important area.
PREFACE xiii
ACKNOWLEDGMENTS
The Forum on Microbial Threats and the IOM wish to express their warmest
appreciation to the individuals and organizations who gave valuable time to
provide information and advice to the Forum through their participation in the
workshop.
The Forum is indebted to the IOM staff who contributed their time and efforts
in planning and executing the workshop and the production of this workshop
xiv PREFACE
Contents
xv
xvi CONTENTS
CONTENTS xvii
APPENDIXES
C Biosketches 194
Members of the Forum on Microbial Threats, 194
Speakers, 205
Forum Staff, 215
The belief that infectious agents may cause certain chronic diseases can be
traced to the mid-19th century, when cancer was studied as a possible infectious
disease. This effort met with little success. In the 1950s and 1960s, much more
biomedical research was directed, again unsuccessfully, at the identification of
microorganisms purported to cause a variety of chronic diseases. In recent years,
however, the picture has begun to change. A number of chronic diseases have
now been linked, in some cases definitively, to an infectious etiology: peptic
ulcer disease with Helicobacter pylori, cervical cancer with several human
papillomaviruses, Whipple’s disease with Tropheryma whipplei, Lyme arthritis
and neuroborreliosis with Borrelia burgdorferi, AIDS with the human immuno-
deficiency virus, liver cancer and cirrhosis with hepatitis B and C viruses, to
name a few. Indeed, evidence continues to mount implicating microorganisms as
etiologic agents of chronic diseases that have substantial morbidity and mortality,
including atherosclerosis and cardiovascular disease, type 1 diabetes, inflamma-
tory bowel disease, and a variety of neurological diseases. The proven and sus-
pected roles of microbes does not stop with physical ailments; infections are in-
creasingly being examined as associated causes of or possible contributors to a
variety of serious, chronic neuropsychiatric disorders and to developmental prob-
lems, especially in children.
It also has become apparent that multiple pathogens sometimes interact in
causing chronic diseases or rendering them more virulent. For example, people
who have concomitant infection with hepatitis C virus and the organism that
causes schistosomiasis—as many individuals do in some developing countries—
often develop schistosomiasis much more rapidly than do people who are not
coinfected. Exploring such pathogen interactions and their effects on the immune
system represent rapidly burgeoning areas of scientific interest.1
This report summarizes a two-day workshop held by the Institute of
Medicine’s Forum on Microbial Threats on October 21–22, 2002, to address this
rapidly evolving field. Invited experts presented research findings on a range of
recognized and potential chronic sequelae of infections, as well as on diverse
pathogenic mechanisms leading from exposure to chronic disease outcomes. Can-
cers, cardiovascular disease, demyelinating syndromes, neuropsychiatric diseases,
hepatitis, and type 1 diabetes were among the conditions addressed. Participants
explored factors driving infectious etiologies of chronic diseases of prominence,
identified difficulties in linking infectious agents with chronic outcomes, and dis-
cussed broad-based strategies and research programs to advance the field. Table
S-1 lists the infectious agents and associated diseases discussed in this report.
Emerging infectious diseases are conceptualized either as newly identified or
appreciated infectious illnesses and conditions, or as previously recognized syn-
dromes that are newly attributed to infection. Some scientists now believe that a
substantial portion of chronic diseases may be causally linked to infectious agents.
Just as the germ theory opened the way for numerous discoveries about the
sources of acute infections, changing ideas about the nature of both infectious
diseases and chronic diseases, coupled with the advent of powerful new labora-
tory techniques, are leading to novel claims concerning the infectious origins of
chronic diseases.
1It should be clearly noted throughout this summary report that the nature of the evidence for
causality of a chronic disease from an infectious agent varies considerably. Each of the cases re-
viewed here represents a wide spectrum of the nature of the relationship between the infectious agent
and the chronic disease. In some cases, the links are definitive (e.g., human papillomavirus and cervi-
cal cancer). In other cases, the relationship has only recently been investigated with little more than
suspected associations from preliminary data (e.g., enteroviruses and Type I diabetes).
occurs with hepatitis C virus infection, although in this case there is virus persis-
tence when the infection occurs in adulthood. Viral infections may also be latent
at the time of diagnosis. For example, there are several viruses for which patients
with multiple sclerosis (MS) exhibit higher antibody levels than control patients.
Johnson reports on one study which revealed that 23 percent of MS patients had
antibodies to two or more viruses present within their central nervous systems,
with one patient presenting with 11 viruses. It is not yet clear whether infection(s)
triggers MS or whether elevated markers of infection are secondary to the under-
lying inflammatory processes of the disease. Such findings emphasize the com-
plexity of directly attributing chronic disease to one or more specific infectious
agents.
• Detecting and/or isolating microbes that are present in a variety of tissues
may pose significant technical difficulties. Current methods to identify novel or
rare microorganisms may be inadequate. During the workshop, David Persing
reported on the deficiencies and weaknesses of conventional methods for identi-
fying and subtyping microorganisms. However, newer molecular technology,
such as broad-range amplification of ribosomal targets directly from tissue or
culture, can complement conventional systems, and these tools have helped in
identifying several new species and pathogenic subtypes. For example, the infec-
tious agent strongly suspected of causing Whipple’s disease remained elusive for
years. Applying broad-range polynuclear chain reaction techniques enabled sci-
entists to amplify and categorize the etiologic Tropheryma whipplei bacterium.
Patrick Moore (Chapter 3) recounts the development of a technique called repre-
sentational difference analysis to identify Kaposi’s sarcoma-associated herpesvi-
rus as a cause of AIDS-associated Kaposi’s sarcoma. These discoveries exem-
plify the diligent effort required to move from identification of a new DNA
sequence to confirming causality in a specific disease. During the workshop,
Persing also described the potential for gene expression arrays (microarrays),
proteomics, and other technologies to identify patterns of host response to an
infection(s) that might explain the pathogenic processes from exposure to chronic
disease and lead to the development of diagnostic tools for these entities. Phylo-
genetic analysis can relate new pathogens for which there are no effective diag-
nostic assays to known agents through conserved epitopes and other properties,
facilitating the evaluation of new infectious causes of disease.
Given the various reasons why it may often prove difficult to satisfy Koch’s
postulates in linking a particular infectious agent to a particular chronic disease,
alternative sets of criteria may need to be developed for determining causation.
Such criteria must take into account the more complex relationships that are be-
ing observed between microbial agents and chronic disease, and they likely will
require collection of more challenging types of experimental data, especially
molecular data, that can help clarify discrete causal links. Toward this goal, sev-
eral promising avenues of research are being pursued, including extending vari-
ous genetic technologies and modifying animal and cell culture models of human
disease to make them more immediately relevant to microbial disease causation.
Ensuing discussions highlighted gaps in scientific knowledge and in the trans-
lation of research data to health care interventions for both well-accepted and
more speculative causal associations. Participants noted the complexity of these
issues, as well as the importance of strengthening the critical linkages among
clinicians, researchers, epidemiologists, and public health officials.
tain the precise timing of infection (which may have happened well in the past) or
the exact nature of the pathogen.
Scientifically sound data on the infectious etiologies of chronic diseases must
derive from new technologies and the optimization of existing assays. The re-
search must be guided by epidemiologic insights gained from well-designed stud-
ies of disease in human populations and from the application of sophisticated
surveillance systems to detect and monitor diseases and pathogens. Standardiza-
tion and reproducibility will be essential. Selection of appropriate cases and con-
trols is imperative, with the use of systematic case studies or experimental de-
signs when this is not possible. Prospective cohort studies should incorporate
appropriate surveillance and be capable of detecting outbreaks of infection as
well as identifying recently infected individuals. Throughout all, researchers will
need to employ comparable definitions of infection and of the chronic disease
being explored. To develop enough human capital for these endeavors, it will be
necessary to attract more scientists to the relevant fields and provide more train-
ing in attendant epidemiological and scientific areas.
Overcoming these obstacles will require the concentrated efforts of research-
ers from a variety of disciplines, including epidemiology, clinical medicine, mo-
lecular biology, and pathology, among others. It also will require harnessing new
analytical tools and approaches that have emerged recently, and continue to
emerge, from molecular biology, genomics, and biotechnology. One of the most
fruitful technologies centers on the ability to detect and manipulate nucleic acid
molecules in microorganisms, thus creating a powerful means for identifying pre-
viously unknown microbial pathogens and for studying the host-pathogen rela-
tionship. Other new tools being employed include broad-range polymerase chain
reaction and representational difference analysis, both of which have played key
roles in linking numerous pathogens with chronic diseases. Equipped with these
and other advanced tools, researchers are becoming better able to move beyond
the limitations of Koch’s postulates and to link infectious agents with chronic
diseases more precisely and with greater confidence than ever before. In addition,
researchers are developing sophisticated approaches for exploring the interplay
of genetic and environmental factors in the causation of a number of important
developmental behavioral disorders.
Participants also identified a number of general characteristics of a compre-
hensive and coordinated effort that would enhance efforts both to identify links
between infectious microorganisms and chronic diseases and to develop and
implement interventions to minimize their health consequences. For example,
they noted need to develop prototypes and standards to guide this work. Stan-
dardized case definitions are needed to facilitate research as well as the clinical
diagnosis of infection (active, persistent, or latent) and the chronic syndromes or
outcomes that result from it. Laboratory assays need to be adopted that are uni-
form in terms of sensitivity, specificity, and reproducibility. High-throughput as-
says meeting similar standards will be key to the study of large cohorts and popu-
The Forum discussions emphasized two major themes: (1) the need to define
the nature and scope of future research that will balance global efforts targeting
various chronic disease syndromes, and (2) the need to develop a coordinated and
systematic strategy to maximize resource use and overcome the inherent techno-
logic and epidemiologic challenges, as well as the organizational barriers, that
now impede progress in this field.
CONCLUSION
The substantial burden posed by chronic diseases of likely infectious etiol-
ogy demands global attention and action. Evidence continues to mount implicat-
ing microorganisms as important etiologic agents of chronic diseases that con-
tribute substantially to morbidity and mortality. However, the identification and
confirmation of infectious causes of chronic diseases is complicated by several
problems, including frequent multifactor causation for many of these diseases
and differences in the environmental background and genetic composition of dif-
ferent populations. Recently developed molecular and immunological techniques
offer new approaches to addressing the technical barriers. However, improved
coordination among basic and clinical scientists, pathologists, and epidemiolo-
gists also will be critical to progress. Standardization of case definitions and ana-
lytical assays combined with sound epidemiologic design will help, as will the
development of broad, new strategies for creating carefully pedigreed specimen
collections and disease registries. Although the task is daunting, taking the prac-
tical and pragmatic pathways described above could clarify many of the uncertain
relationships between infectious agents and chronic diseases.
OVERVIEW
Chronic diseases cause 70 percent of all deaths in the United States. Yet the
factors that cause many of these conditions have been poorly understood until
recently. Advances in numerous detection and diagnostic techniques have re-
vealed that several chronic illnesses result from infectious agents. For example,
the human papillomavirus causes more than 90 percent of cervical cancers. The
hepatitis B virus accounts for more than 60 percent of liver cancer. The Epstein-
Barr virus produces in people simultaneously infected with malaria a cancer
known as Burkitt’s lymphoma, a leading cause of childhood cancer deaths glo-
bally. The bacterium Helicobacter pylori has been linked to a number of disor-
ders, including duodenal ulcers, gastric cancer, and certain types of lymphomas.
Other connections between infections and chronic diseases are suspected,
but not proven. Epstein-Barr virus, for example, has been found in patients with
Hodgkin’s disease and with aggressive breast cancers. Multiple sclerosis acts
suspiciously like an infection, with patients experiencing high antibody levels as
well as exacerbations and remissions. Juvenile-onset diabetes may arise when a
Coxsackie B enterovirus elicits an immune response that damages the pancreas.
Identifying and confirming an infectious cause of a chronic disease is com-
plicated by several factors:
13
The case studies presented in this chapter were chosen to provide insight into
the range of research under way in the field. The chronic diseases covered repre-
sent the full spectrum of those that have been linked in some degree, from “clearly
proven” to “suspected,” with infectious agents; they are caused by a variety of
microorganisms; and their association with disease is supported variously by labo-
ratory and epidemiological studies. Although other diseases and studies might
have been included, some limits were imposed by time constraints and the avail-
ability of speakers.
Eduardo Franco reviewed the evidence that human papillomavirus (HPV)
infection is a cause of cervical cancer. HPV infection precedes lesion develop-
ment and appears to be necessary for cervical cancer to occur. This is one of the
first examples in which an infectious agent has been identified to be necessary for
cancer development. This causal relationship was revealed through the use of
improved diagnostic tools that enabled more accurate identification of HPV. As
the role of infection by certain types of HPV is better elucidated as the cause of
cervical cancer, HPV testing in cervical cancer screening programs becomes an
important part of a primary prevention strategy. Another component of this strat-
egy may be increased use of a recently developed vaccine. Clinical studies indi-
cate that the new HPV 16 VLP vaccine was 100 percent effective in preventing
acquisition of persistent infection with HPV 16, and was 90 percent effective in
preventing any incident HPV 16 infection, transient or persistent. Immunization
against HPV may have greatest value in developing countries, where 80 percent
of the global burden of cervical cancer occurs each year.
William Mason presented the association between hepatitis B virus infection
and liver disease. Infection with the virus remains a worldwide problem, with
more than 350 million people chronically infected. Although a vaccine has been
available for the past 20 years, its high cost prevents universal vaccination. Cur-
rent research, therefore, has focused on the development of effective therapies to
cure those individuals chronically infected with the virus. Mason described the
research presently being conducted in a number of animal model systems, includ-
ing the woodchuck. Along with clinical studies, these models have been able to
characterize infections and evaluate therapies, as well as better elucidate the dif-
ficulties of treating chronic infections with nucleoside analogs.
Michael Dunne described the relationship between infection and cardio-
vascular disease. There is a tight association between hypercholesterolemia and
atherosclerosis; recent research has examined how inflammation within the plaque
*The author’s research on the epidemiology of HPV infection and prevention of cervical cancer is
funded by grants from the Canadian Institutes of Health Research (CIHR) and from the U.S. National
Institutes of Health.
Human Papillomaviruses
HPVs are small, double-stranded DNA viruses of approximately 55 nanom-
eters (nm) with an icosahedral protein capsid containing 72 capsomers. The ge-
nome is circular and contains 7500–8000 base pairs (bp). HPVs have the follow-
ing characteristics:
Normal Invasive
cervical Low grade High grade
cervical
epithelium lesions lesions
cancer
Co-factors : nutrition
FIGURE 1-1 Etiologic model in cervical carcinogenesis showing the primary role of
HPV infection, its relation with sexual activity, and the putative role of cofactors.
• Cannot be cultivated
• Over 150 genotypes identified, of which more than 40 infect the anogenital
tract
• High risk (oncogenic) types: 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59,
68
• Induces both benign (caused by low risk types) and malignant (caused by
high risk types) diseases
• Two major viral oncogenes: E6 (binds to p53) and E7 (binds to retino-
blastoma [Rb] protein)
1.0
.8
Proportion remaining HPV positive
HPV 16/18
.6
Nononcogenic
.4 types
Oncogenic
types other than 16/18
.2
0.0
0 4 8 12 16 20 24
Time since enrollment (months)
FIGURE 1-2 Actuarial curves showing clearance of prevalent HPV infection according
to type present at enrollment in a cohort study of asymptomatic women presenting for
cervical cancer screening.
SOURCE: Adapted from Franco et al. (1999b).
0.5
0.4
Cumulative incidence of any SIL
Transient infection
0.1
HPV visits 1 and 2
0.0
0 12 24 36 48 60 72
Time since enrollment (months)
FIGURE 1-3 Actuarial curves showing the cumulative incidence of cervical squamous
intraepithelial lesions (SIL) according to HPV infection in the first two visits in a cohort
study of asymptomatic women presenting for cervical cancer screening.
SOURCE: Adapted from Schlecht et al. (2001).
yield higher viral loads than transient ones (Caballero et al., 1999) and those with
non-European variants of HPVs 16 and 18 tend to be associated with higher risk
of CIN as compared with those caused by European variants (Villa et al., 2000).
Defining viral persistence is critical because trials of HPV vaccine efficacy
rely on the reduction of the risk of persistent infection as one of the primary
outcomes. Similarly, concerning screening of cervical cancer by HPV testing, a
main drawback is the low positive predictive value of a single test because of the
relatively high prevalence of latent HPV infections in the population, particularly
among young women. The predictive value would increase substantially if test-
ing were to rely on repeated samplings, about 6 months apart, because of the
aforementioned high prognostic value of persistent positivity. However, popula-
tion screening cannot rely on repeated testing to be cost-effective and realistic as
a public health measure. It would be highly desirable if one could, with a single
HPV test, collect enough ancillary information on the virus and on the host that
would allow determining whether or not a single instance of HPV positivity is
likely to represent a persistent infection.
Conclusions
During the last 20 years, the concerted effort among virologists, epidemiolo-
gists, and clinical researchers has helped to elucidate the role of infection by
certain types of HPV as the necessary cause of cervical cancer. This has opened
new frontiers for preventing a disease that is responsible for substantial morbidity
and mortality, particularly among women living in resource-poor countries. Re-
search on two prevention fronts has already begun in several populations in the
form of preliminary trials assessing the efficacy of HPV vaccines and of studies
of the value of HPV testing in cervical cancer screening (see Figure 1-4). Progress
on both counts is very promising. While the benefits of vaccination against HPV
infection as a cervical cancer prevention tool are at least a decade into the future,
the potential benefits of HPV testing in screening for this disease can be realized
now in most populations.
Primary prevention of cervical cancer can also be achieved through preven-
tion and control of genital HPV infection. Health promotion strategies geared at a
change in sexual behavior targeting all sexually-transmitted infections of public
health significance can be effective in preventing genital HPV infection (Franco
et al., 2001). Although there is consensus that symptomatic HPV infection (geni-
tal warts) should be managed via treatment, counseling, and partner notification,
active case-finding of asymptomatic HPV infection is currently not recommended
Primary
screening Triage of
STD education, abnormalities and
HPV vaccination F/up of recurrences
FIGURE 1-4 Opportunities for primary and secondary preventive approaches in the natu-
ral history of cervical cancer.
REFERENCES
Bosch FX, Manos MM, Muñoz N, Sherman M, Jansen AM, Peto J, Schiffman MH, Moreno V,
Kurman R, Shah KV. 1995. Prevalence of human papillomavirus in cervical cancer: a world-
wide perspective. International biological study on cervical cancer (IBSCC) Study Group. Jour-
nal of the National Cancer Institute 87:796–802.
Bosch FX, Lorincz A, Munoz N, Meijer CJ, Shah KV. 2002. The causal relation between human
papillomavirus and cervical cancer. Journal of Clinical Pathology 55:244–265.
Brinton LA, Reeves WC, Brenes MM, et al. 1989a. The male factor in the etiology of cervical cancer
among sexually monogamous women. International Journal of Cancer 44:199–203.
Brinton LA, Reeves WC, Brenes MM, Herrero R, de Britton RC, Gaitan E, Tenorio F, Garcia M,
Rawls WE. 1989b. Parity as a risk factor for cervical cancer. American Journal of Epidemiology
130:486–496.
Caballero OL, Trevisan A, Villa LL, Ferenczy A, Franco EL. 1999. High viral load is associated with
persistent HPV infection and risk of cervical dysplasia. 17th International Papillomavirus Con-
ference, Charleston, SC.
Chellappan S, Kraus VB, Kroger B, Munger K, Howley PM, Phelps WC, Nevins JR. 1992. Adenovi-
rus E1A, simian virus 40 tumor antigen, and human Papillomavirus E7 protein share the capac-
ity to disrupt the interaction between transcription factor E2F and the retinoblastoma gene prod-
uct. Proceedings of the National Academy of Sciences 89:4549–4553.
Cox JT. 1995. Epidemiology of cervical intraepithelial neoplasia: the role of human papillomavirus.
Baillière’s clinical obstetrics and gynaecology 9:1–37.
Cuzick J, Sasieni P, Davies P, Adams J, Normand C, Frater A, van Ballegooijen M, van den Akker-
van Marle E. 2000. A systematic review of the role of human papilloma virus (HPV) testing
within a cervical screening programme: summary and conclusions. British Journal of Cancer
83:561–565.
Dyson N, Howley PM, Munger K, Harlow ED. 1989. The human papilloma virus-16 E7 oncoprotein
is able to bind to the retinoblastoma gene product. Science 243:934–937.
Franco EL. 1991. Viral etiology of cervical cancer: a critique of the evidence. Reviews of Infectious
Diseases 13:1195–1206.
Franco EL, Rohan TE, Villa LL. 1999a. Epidemiologic evidence and human papillomavirus infection
as a necessary cause of cervical cancer. Journal of the National Cancer Institute 91:506–511.
Franco EL, Villa LL, Sobrinho JP, Prado JM, Rousseau MC, Desy M, Rohan TE. 1999b. Epidemiol-
ogy of acquisition and clearance of cervical human papillomavirus infection in women from a
high-risk area for cervical cancer. The Journal of Infectious Diseases 180:1415–1423.
Franco EL, Duarte-Franco E, Ferenczy A. 2001. Cervical cancer: epidemiology, prevention, and role
of human papillomavirus infection. Canadian Medical Association Journal 164:1017–1025.
Herrero R. 1996. Epidemiology of cervical cancer. Journal of the National Cancer Institute Mono-
graphs 21:1–6.
Hildesheim A, Schiffman MH, Gravitt PE, Glass AG, Greer CE, Zhang T, Scott DR, Rush BB,
Lawler P, Sherman ME, et al. 1994. Persistence of type-specific human papillomavirus infec-
tion among cytologically normal women. The Journal of Infectious Diseases 169:235–240.
Ho GY, Kadish AS, Burk RD, Basu J, Palan PR, Mikhail M, Romney SL. 1998a. HPV 16 and
cigarette smoking as risk factors for high-grade cervical intra-epithelial neoplasia. International
Journal of Cancer 78:281–285.
Ho GY, Bierman R, Beardsley L, Chang CJ, Burk RD. 1998b. Natural history of cervicovaginal
papillomavirus infection in young women. New England Journal of Medicine 338:423–428.
IARC Working Group. 1995. Human papillomaviruses. IARC Monographs on the evaluation of car-
cinogenic risks to humans. Vol. 64. Lyon: International Agency for Research on Cancer.
Koutsky LA, Holmes KK, Critchlow CW, Stevens CE, Paavonen J, Beckmann AM, DeRouen TA,
Galloway DA, Vernon D, Kiviat NB. 1992. A cohort study of the risk of cervical intraepithelial
neoplasia grade 2 or 3 in relation to papillomavirus infection. New England Journal of Medicine
327:1272–1278.
Koutsky LA, Ault KA, Wheeler CM, Brown DR, Barr E, Alvarez FB, Chiacchierini LM, Jansen KU.
2002. A controlled trial of a human papillomavirus type 16 vaccine. New England Journal of
Medicine 347:1645–1651.
Liaw KL, Hildesheim A, Burk RD, Gravitt P, Wacholder S, Manos MM, Scott DR, Sherman ME,
Kurman RJ, Glass AG, Anderson SM, Schiffman M. 2001. A prospective study of human
papillomavirus (HPV) type 16 DNA detection by polymerase chain reaction and its association
with acquisition and persistence of other HPV types. The Journal of Infectious Diseases 183:8–
15.
Maciag PC, Schlecht NF, Souza PS, Franco EL, Villa LL, Petzl-Erler ML. 2000. Major histocompat-
ibility complex class II polymorphisms and risk of cervical cancer and human papillomavirus
infection in Brazilian women. Cancer Epidemiology, Biomarkers and Prevention 9:1183–1191.
Makni H, Franco EL, Kaiano J, Villa LL, Labrecque S, Dudley R, Storey A, Matlashewski G. 2000.
p53 polymorphism in codon 72 and risk of human papillomavirus-induced cervical cancer: ef-
fect of inter-laboratory variation. International Journal of Cancer 87:528–533.
Moreno V, Bosch FX, Munoz N, Meijer CJ, Shah KV, Walboomers JM, Herrero R, Franceschi S.
2002. Effect of oral contraceptives on risk of cervical cancer in women with human
papillomavirus infection: the IARC multicentric case-control study. Lancet 359:1085–1092.
Moscicki AB, Shiboski S, Broering J, Powell K, Clayton L, Jay N, Darragh TM, Brescia R, Kanowitz
S, Miller SB, Stone J, Hanson E, Palefsky J. 1998. The natural history of human papillomavirus
infection as measured by repeated DNA testing in adolescent and young women. The Journal of
Pediatrics 132:277–284.
Moscicki AB, Hills N, Shiboski S, Powell K, Jay N, Hanson E, Miller S, Clayton L, Farhat S, Broering
J, Darragh T, Palefsky J. 2001. Risks for incident human papillomavirus infection and low-
grade squamous intraepithelial lesion development in young females. Journal of the American
Medical Association 285:2995–3002.
Muir C, Waterhouse J, Mack T, et al. 1987. Cancer incidence in five continents, Vol. V. IARC
Scientific Publications No. 88. Lyon: International Agency for Research on Cancer.
Nobbenhuis MA, Walboomers JM, Helmerhorst TJ, Rozendaal L, Remmink AJ, Risse EK, van der
Linden HC, Voorhorst FJ, Kenemans P, Meijer CJ. 1999. Relation of human papillomavirus
status to cervical lesions and consequences for cervical-cancer screening: a prospective study.
Lancet 354:20–25.
Parkin DM, Pisani P, Ferlay J. 1999. Estimates of the worldwide incidence of 25 major cancers in
1990. International Journal of Cancer 80:827–841.
Pisani P, Parkin DM, Bray F, Ferlay J. 1999. Estimates of the worldwide mortality from 25 cancers in
1990. International Journal of Cancer 83:18–29.
Potischman N and Brinton LA. 1996. Nutrition and cervical neoplasia. Cancer Causes and Control
7:113–126.
Ries LAG, Eisner MP, Kosary CL, Hankey BF, Miller BA, Clegg L, Edwards BK, eds. 2000. SEER
Cancer Statistics Review: 1973-1997. Bethesda, MD: National Cancer Institute.
Schiller JT. 1999. Papillomavirus-like particle vaccines for cervical cancer. Molecular Medicine To-
day 5:209–215.
Schlecht NF, Kulaga S, Robitaille J, Ferreira S, Santos M, Miyamura RA, Duarte-Franco E, Rohan
TE, Ferenczy A, Villa LL, Franco EL. 2001. Persistent human papillomavirus infection as a
predictor of cervical intraepithelial neoplasia. Journal of the American Medical Association
286:3106–3114.
Thomas M, Matlashewski G, Pim D, Banks L. 1996. Induction of apoptosis by p53 is independent of
its oligomeric state and can be abolished by HPV-18 E6 through ubiquitin mediated degrada-
tion. Oncogene 13:265–273.
Villa LL, Sichero L, Rahal P, Caballero O, Ferenczy A, Rohan T, Franco EL. 2000. Molecular vari-
ants of human papillomavirus types 16 and 18 preferentially associated with cervical neoplasia.
The Journal of General Virology 81:2959–2968.
Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, Snijders PJ, Peto J,
Meijer CJ, Munoz N. 1999. Human papillomavirus is a necessary cause of invasive cervical
cancer worldwide. The Journal of Pathology 189:12-9.
Ylitalo N, Josefsson A, Melbye M, Sorensen P, Frisch M, Andersen PK, Sparen P, Gustafsson M,
Magnusson P, Ponten J, Gyllensten U, Adami HO. 2000. A prospective study showing long-
term infection with human papillomavirus 16 before the development of cervical carcinoma in
situ. Cancer Research 60:6027–6032.
zur Hausen H. Papillomaviruses causing cancer: evasion from host-cell control in early events in
carcinogenesis. 2000. Journal of the National Cancer Institute 92:690–698.
Transmission
Transmission is parenteral, requiring exposure to the blood or blood-con-
taminated materials of infected individuals. The most common mode of exposure
leading to chronic infection occurs at birth when the mother is chronically in-
fected, or during the first year of life. During this period, the risk of an infection
becoming chronic is at least 90 percent. In contrast, the risk of chronic infection
in adults is greater than 10 percent. According to the CDC, the most common
exposure risks in adults in the United States are sexual activity (50 percent of
cases) and intravenous drug abuse (15 percent of cases).
Prevalence
The case fatality rate in adults due to acute hepatitis is about 1 percent. Indi-
viduals with chronic infection, typically acquired in childhood, have a ~25 per-
cent risk of premature death due to either liver cancer or cirrhosis, both resulting
from the persistent liver damage associated with infection. According to WHO,
there are now 350 million chronically infected individuals worldwide. Of these,
60 million are expected to die prematurely of liver cancer or cirrhosis, at a rate of
approximately 1 million per year (5,000 per year in the United States). This does
not account for new cases, which will continue to accumulate in the coming de-
cades.
Vaccines
A vaccine comprised of the viral envelope proteins has been available for
over 20 years. Due in part to high cost, universal vaccination was not initially
feasible in many parts of the world, but lower cost vaccines have subsequently
come into use. Universal vaccination of school children is now in effect in the
United States. In some parts of the world, especially in Africa and regions of
Asia, chronic infection rates exceed 5–10 percent of the population, but vaccina-
tion has not yet been economically feasible in all of these areas, even with low-
cost vaccines. Although attempts are under way to address this problem (Kane,
2003), for various reasons of cost and delivery, HBV is likely to remain a major
public health problem. On top of this problem there is evidence for vaccine es-
cape mutants (He et al., 2001; Torresi et al., 2002; Wilson et al., 2000). Though
these do not yet seem to be a major public health problem, they remain a concern
even for the large pool of individuals that have already received the current vaccine.
In addition, about 5 percent of vaccinated individuals fail to produce a measur-
able antibody response, suggesting that they also remain at risk for HBV infection.
Current Research
A major goal of current research has thus been the development of therapies
to cure chronically infected individuals. A problem in achieving this is that hepa-
tocytes comprise a self-renewing population with a low turnover rate, and this
population often appears to be 100 percent infected. This same barrier is con-
fronted and overcome during immune clearance of transient infections, though it
remains controversial how the virus is actually destroyed (Guidotti et al. 1999;
Guo et al., 2000; Jilbert et al., 1992; Kajino et al., 1994; Thimme et al., 2003).
However, in chronic carriers, the immune system is usually unable to mount such
a response, especially in those infected as children. Some hope for better immuno-
therapies has however been sustained by the fact that interferon alpha administra-
tion induces virus loss in about 20–30 percent of carriers (Hoofnagle and Lau,
1997), typically those with adult-acquired infections. In addition, some carriers
experience spontaneous loss of the virus in association with a flare of liver dis-
ease. In both instances, clearance is probably due to activation of the same set of
immune responses that are active in clearance of transient infections. Key issues
now are how this clearance is carried out, whether it requires destruction of all of
the infected hepatocytes, if the immune system has the capacity to cure an infected
hepatocyte, and if it can be induced in carriers that have failed to respond to
interferon therapy with virus clearance.
Treatment
Another approach to treatment of chronic infections is administration of
nucleoside analog inhibitors of the HBV reverse transcriptase. Lamivudine was
approved by the U.S. Food and Drug Administration (FDA) in 1998 and has been
shown in clinical trials to have a treatment success rate similar to interferon alpha
(Perrillo, 2002). A significant problem with lamivudine is the emergence of drug-
resistant variants of HBV as therapy continues past a year. Another nucleoside,
adefovir dipivoxil, recently received FDA approval and to date drug-resistant
variants have not been reported. Moreover, this drug retains activity against
lamivudine-resistant HBV (Delaney et al., 2001). However, at doses higher than
used for HBV carriers, nephrotoxicity has been observed (Tanji et al., 2001). It
may be that nephrotoxicity will become a problem in HBV therapy due to a cu-
mulative effect if carriers require treatment indefinitely. A number of other
nucleoside analogs are now in Phase II trials. If these compounds are not toxic
during long-term administration, and if viral multi-drug resistance does not de-
velop, it should be possible to eliminate over time the viral cccDNA that main-
tains a cellular infection by a combination of dilution and hepatocyte death.
Achieving this would also allow a critical test of the hypothesis that curing a
chronic infection would significantly reduce the risk of death due to cirrhosis,
which seems likely, and due to liver cancer, which is difficult to predict, because
liver cancer may occur in a liver that appears relatively healthy histologically.
Research Models
HBV research generally reflects public health concerns. How can chronic
infections be cured? Will eliminating the virus reduce the risk of liver cancer and
premature death from liver disease? What is the mechanism of carcinogenesis?
(It is speculated that immune-mediated chronic injury, insertional mutagenesis,
and viral proteins all may play a role.) These questions have been investigated
using clinical samples and a number of model systems.
Woodchucks are naturally infected with woodchuck hepatitis virus (WHV)
(Summers et al., 1978), which is closely related to HBV and, like HBV, induces
liver cancer (but not cirrhosis) during a chronic infection (Popper et al., 1987).
Similarly, domestic ducks are infected with duck hepatitis B virus (DHBV), a
more distant relative of HBV (Mason et al., 1980; Zhou, 1980). Unlike HBV and
WHV, chronic DHBV infection has not been associated with either cirrhosis or
liver cancer, possibly because of a lower antiviral immune response in carriers.
HBV transgenic mice have been powerful tools for studying certain aspects of the
antiviral immune response (Guidotti and Chisari, 2001), even though these mice
do not support a complete HBV infection cycle (Tang and McLachlan, 2002). On
occasion, chimpanzees, which are susceptible to HBV, have been used to address
research issues (Guidotti et al., 1999; Thimme et al., 2003).
Among the model systems, the duck has been heavily used to understand the
virus life cycle at the molecular level, to study the biology of infection, and to
characterize antiviral therapies, primarily with nucleoside analogs. The wood-
chuck model has been less used to study molecular biology issues, but has been
employed extensively in the development of antiviral therapies and in character-
ization of the link between chronic infection and liver cancer. An unresolved
issue arose in the latter studies. It was found that liver cancer in woodchucks is
almost always associated with transcriptional activation of N-myc2 expression in
the liver by insertion of viral enhancer sequences (Fourel et al., 1994; Wei et al.,
1992). Contrary to expectation, insertional activation of N-myc2 does not appear
to be a correlate of liver cancer in HBV carriers. Indeed, with a few rare excep-
tions, it remains unclear if the frequent sporadic integration of viral DNA that
characterizes an infection has a role in most liver cancers that occur in individuals
chronically infected with HBV (Dejean et al., 1986; Gozuacik et al., 2001).
The HBV transgenic mouse, in contrast to the natural infection models, has
been most heavily used to demonstrate the effects of immune cytokines, such as
interferons alpha and gamma, on viral replication intermediates. It was found that
cytokines can induce the rapid clearance of viral proteins, RNAs, and DNAs from
mouse hepatocytes (Guidotti and Chisari, 2001). These observations seem likely
to provide part of the explanation for how virus replication is shut down during
the clearance of transient HBV infections.
Though the relationship to natural infections is still unclear, a number of
studies have shown that mice carrying the HBV transcriptional activator, X, as a
transgene, are at increased risk of developing liver cancer (Kim et al., 1991; Mad-
den et al., 2001; Terradillos et al., 1997). These data suggest that X is in fact a
viral oncogene, but clinical evidence to support this conclusion is still lacking,
and it is difficult to address this issue in the woodchuck model, because X is
needed to establish a productive infection (Chen et al., 1993; Zoulim and Seeger,
1994b).
In addition to characterizing infections and therapies, the animal models have
also provided, along with clinical studies, a better understanding of the difficul-
ties of treating chronic infections with nucleoside analogs. From such studies, it
has been determined that cccDNA can persist in the liver for months, and prob-
ably years, even when virus DNA synthesis is effectively inhibited (Colonno et
al., 2001; Foster et al., 2003; Luscombe et al., 1996; Mason et al., 1994; Zhu et
al., 2001). Persistence of cccDNA may be attributable to two factors: 1) an inher-
ent stability within non-dividing hepatocytes, and 2) the relatively low turnover
(perhaps a few percent per day) of hepatocytes in most carriers. Studies with
animal models have also established that the mutation rate of the viruses is quite
high, with a single-base mutation prevalence of about 10-4 (Pult et al., 2001).
Thus, drug-resistant variants, especially those requiring only one or two base
changes, are likely to be present at the start of therapy. The primary factors needed
for subsequent emergence of drug-resistant variants are the time required for the
hepatocyte population to become susceptible to spread of virus (e.g., for loss of
super-infection resistance), the prevalence of a drug-resistant virus at the start of
therapy, and its growth rate (Zhang and Summers, 2000). In practice, emergence
of mutants can take from months to several years, the variation probably reflect-
ing additional factors, including the effect of nucleoside therapy on the antiviral
immune response of the host (Boni et al., 2001).
Outlook
Discovery of an effective HBV vaccine in the 1960s (Blumberg, 1977) led to
the hope that HBV would be eliminated, or at least substantially reduced in the
human population within the then foreseeable future. This still remains mostly a
hope. Two objectives still need to be fulfilled, universal vaccination (Kane, 2003),
and development of an effective therapy for chronic infection. Even though not
everyone will be protected using the current vaccine, most would be, and the
carrier incidence should decline substantially, first among the young. The goal of
complete elimination seems unlikely without major advances in the treatment
and elimination of chronic infections, particularly treatments that are rapid acting
and cost-effective.
REFERENCES
Blumberg BS. 1977. Australia antigen and the biology of hepatitis B. Science 197:17–25.
Boni C, Penna A, Ogg GS, Bertoletti A, Pilli M, Cavallo C, Cavalli A, Urbani S, Boehme R,
Panebianco R, Fiaccadori F, Ferrari C. 2001. Lamivudine treatment can overcome cytotoxic T-
cell hyporesponsiveness in chronic hepatitis B: new perspectives for immune therapy.
Hepatology 33:963–971.
Chen HS, Kaneko S, Girones R, Anderson RW, Hornbuckle WE, Tennant BC, Cote PJ, Gerin JL,
Purcell RH, Miller RH. 1993. The woodchuck hepatitis virus X gene is important for establish-
ment of virus infection in woodchucks. Journal of Virology 67:1218–1226.
Colonno RJ, Genovesi EV, Medina I, Lamb L, Durham SK, Huang ML, Corey L, Littlejohn M,
Locarnini S, Tennant BC, Rose B, Clark JM. 2001. Long-term entecavir treatment results in
sustained antiviral efficacy and prolonged life span in the woodchuck model of chronic hepatitis
infection. The Journal of Infectious Diseases 184:1236–1245.
Dejean A, Bougueleret L, Grzeschik KH, Tiollais P. 1986. Hepatitis B virus DNA integration in a
sequence homologous to v-erb-A and steroid receptor genes in a hepatocellular carcinoma.
Nature 322:70–72.
Delaney WE, Edwards R, Colledge D, Shaw T, Torresi J, Miller TG, Isom HC, Bock CT, Manns MP,
Trautwein C, Locarnini S. 2001. Cross-resistance testing of antihepadnaviral compounds using
novel recombinant baculoviruses which encode drug-resistant strains of hepatitis B virus. Anti-
microbial Agents and Chemotherapy 45:1705–1713.
Foster WK, Miller DS, Marion PL, Colonno RJ, Kotlarski I, Jilbert AR. 2003. Entecavir therapy
combined with DNA vaccination for persistent duck hepatitis B virus infection. Antimicrobial
Agents and Chemotherapy 47:2624–2635.
Fourel G, Couturier J, Wei Y, Apiou F, Tiollais P, Buendia MA. 1994. Evidence for long-range
oncogene activation by hepadnavirus insertion. The European Molecular Biology Organization
Journal 13:2526–2534.
Gozuacik D, Murakami Y, Saigo K, Chami M, Mugnier C, Lagorce D, Okanoue T, Urashima T,
Brechot C, Paterlini-Brechot P. 2001. Identification of human cancer-related genes by naturally
occurring Hepatitis B Virus DNA tagging. Oncogene 20:6233–6240.
Guidotti LG and Chisari FV. 2001. Noncytolytic control of viral infections by the innate and adaptive
immune response. Annual Review of Immunology 19:65–91.
Guidotti LG, Rochford R, Chung J, Shapiro M, Purcell R, Chisari FV. 1999. Viral clearance without
destruction of infected cells during acute HBV infection. Science 284:825–829.
Guo JT, Zhou H, Liu C, Aldrich C, Saputelli J, Whitaker T, Barrasa MI, Mason WS, Seeger C. 2000.
Apoptosis and regeneration of hepatocytes during recovery from transient hepadnavirus infec-
tions. Journal of Virology 74:1495–1505.
He C, Nomura F, Itoga S, Isobe K, Nakai T. 2001. Prevalence of vaccine-induced escape mutants of
hepatitis B virus in the adult population in China: a prospective study in 176 restaurant employ-
ees. Journal of Gastroenterology and Hepatology 16:1373–1377.
Hoofnagle JH and Lau D. 1997. New therapies for chronic hepatitis B. Journal of Viral Hepatitis
4:41–50.
Jilbert AR, Wu TT, England JM, Hall PM, Carp NZ, O’Connell AP, Mason WS. 1992. Rapid resolu-
tion of duck hepatitis B virus infections occurs after massive hepatocellular involvement. Jour-
nal of Virology 66:1377–1388.
Kajino, K., A. R. Jilbert, J. Saputelli, C. E. Aldrich, J. Cullen, and W. S. Mason. 1994. Woodchuck
hepatitis virus infections: very rapid recovery after a prolonged viremia and infection of virtu-
ally every hepatocyte. Journal of Virology 68:5792–5803.
Kane MA. 2003. Global control of primary hepatocellular carcinoma with hepatitis B vaccine: The
contributions of research in Taiwan. Cancer Epidemiology, Biomarkers & Prevention 12:2–3.
Kim CM, Koike K, Saito I, Miyamura T, Jay G. 1991. HBx gene of hepatitis B virus induces liver
cancer in transgenic mice. Nature 351:317–320.
Luscombe C, Pedersen J, Uren E, Locarnini S. 1996. Long-term ganciclovir chemotherapy for con-
genital duck hepatitis B virus infection in vivo: Effect on intrahepatic-viral DNA, RNA, and
protein expression. Hepatology 24:766–773.
Madden CR, Finegold MJ, Slagle BL. 2001. Hepatitis B virus X protein acts as a tumor promoter in
development of diethylnitrosamine-induced preneoplastic lesions. Journal of Virology 75:3851–
3858.
Mason WS, Seal G, Summers J. 1980. Virus of Pekin ducks with structural and biological relatedness
to human hepatitis B virus. Journal of Virology 36:829–836.
Mason WS, Cullen J, Saputelli J, Wu TT, Liu C, London WT, E Lustbader, Schaffer P, O’Connell
AP, Fourel I, Aldrich CE, Jilbert AR. 1994. Characterization of the antiviral effects of 2′
carbodeoxyguanosine in ducks chronically infected with duck hepatitis B virus. Hepatology
19:398–411.
Moraleda G, Saputelli J, Aldrich CE, Averett D, Condreay L, Mason WS. 1997. Lack of effect of
antiviral therapy in nondividing hepatocyte cultures on the closed circular DNA of woodchuck
hepatitis virus. Journal of Virology 71:9392–9399.
Perrillo RP. 2002. How will we use the new antiviral agents for hepatitis B? Current Gastroenterol-
ogy Reports 4:63–71.
Popper H, Roth L, Purcell RH, Tennant BC, Gerin JL. 1987. Hepatocarcinogenicity of the woodchuck
hepatitis virus. Proceedings of the National Academy of Sciences 84:866–870.
Pult I, Abbott N, Zhang YY, Summers JW. 2001. Frequency of spontaneous mutations in an avian
hepadnavirus infection. Journal of Virology 75:9623–9632.
Seeger C and Mason WS. 2000. Hepatitis B virus biology. Microbiology and Molecular Biology
Reviews 54:51–68.
Summers J and Mason WS. 1982. Replication of the genome of a hepatitis B-like virus by reverse
transcription of an RNA intermediate. Cell 29:403–415.
Summers J, Smolec JM, Snyder R. 1978. A virus similar to human hepatitis B virus associated with
hepatitis and hepatoma in woodchucks. Proceedings of the National Academy of Sciences
75:4533–4537.
Tang H and McLachlan A. 2002. Avian and mammalian hepadnaviruses have distinct transcription
factor requirements for viral replication. Journal of Virology 76:7468–7472.
Tanji N, Tanji K, Kambham N, Markowitz GS, Bell A, D’Agati VD. 2001. Adefovir nephrotoxicity:
possible role of mitochondrial DNA depletion. Human Pathology 32:734–740.
Terradillos O, Billet O, Renard CA, Levy R, Molina T, Briand P, Buendia MA. 1997. The hepatitis B
virus X gene potentiates c-myc-induced liver oncogenesis in transgenic mice. Oncogene 14:395–
404.
Thimme R, Wieland S, Steiger C, Ghrayeb J, Reimann KA, Purcell RH, Chisari FV. 2003. CD8(+) T
cells mediate viral clearance and disease pathogenesis during acute hepatitis B virus infection.
Journal of Virology 77:68–76.
Torresi J, Earnest-Silveira L, Civitico G, Walters TE, Lewin SR, Fyfe J, Locarnini SA, Manns M,
Trautwein C, Bock TC. 2002. Restoration of replication phenotype of lamivudine-resistant hepa-
titis B virus mutants by compensatory changes in the “fingers” subdomain of the viral poly-
merase selected as a consequence of mutations in the overlapping S gene. Virology 299:88–99.
Tuttleman JS, Pourcel C, Summers J. 1986. Formation of the pool of covalently closed circular viral
DNA in hepadnavirus-infected cells. Cell 47:451–460.
Weber M, Bronsema V, Bartos H, Bosserhoff A, Bartenschlager R, Schaller H. 1994. Hepadnavirus
P protein utilizes a tyrosine residue in the TP domain to prime reverse transcription. Journal of
Virology 68:2994–2999.
Wei Y, Fourel G, Ponzetto A, Silvestro M, Tiollais P, Buendia MA. 1992. Hepadnavirus integration:
mechanisms of activation of the N-myc2 retrotransposon in woodchuck liver tumors. Journal of
Virology 66:5265–5276.
Wilson JN, Nokes DJ, Carman WF. 2000. Predictions of the emergence of vaccine-resistant hepatitis
B in The Gambia using a mathematical model. Epidemiology and Infection 124:295–307.
Zhang YY and Summers J. 2000. Low dynamic state of a viral competition in a chronic avian
hepadnavirus infection. Journal of Virology 74:5257–5265.
Zhou YZ. 1980. A virus possibly associated with hepatitis and hepatoma in ducks. Shanghai Medical
Journal 3:641–644.
Zhu Y, Yamamoto T, Cullen J, Saputelli J, Aldrich CE, Miller DS, Litwin S, Furman PA, Jilbert AR,
Mason WS. 2001. Kinetics of hepadnavirus loss from the liver during inhibition of viral DNA
synthesis. Journal of Virology 75:311–322.
Zoulim F and Seeger C. 1994a. Reverse transcription in hepatitis B viruses is primed by a tyrosine
residue of the polymerase. Journal of Virology 68:6–13.
Zoulim F and Seeger C. 1994b. Woodchuck hepatitis virus X protein is required for viral infection in
vivo. Journal of Virology 68:2026–2030.
Local infection
Infection of the
arterial wall
FIGURE 1-5 Pathways through which local infection can lead to atherogenesis.
infect distal tissue. Once at the site, the organisms could drive a local inflamma-
tory process or, in addition, infect other cells within the arterial wall.
A number of potential pathogens have been associated with atherosclerosis
(Danesh, 1999). The strength of the association varies with the organism but is
based on seroepidemiologic studies, histopathologic evidence of disease, animal
model data and various pathophysiologic associations. Among possible viral
pathogens are cytomegalovirus and herpes simplex (Nieto, 1999; Dunne, 2000).
Among bacterial pathogens are various dental organisms, Helicobacter pylori,
and Mycoplasma pneumoniae. The most significant amount of preclinical and
clinical investigation, however, has focused on C. pneumoniae; as an example of
the types of evidence that can implicate a potential infectious pathogen driving
some component of the atherosclerotic process, these data will be reviewed in
more detail.
cutoffs, different case definitions of coronary artery disease, and were performed
in different geographic regions. Overall, it appears that elevated antibody titers to
C. pneumoniae are associated with a three-fold increase in the likelihood of hav-
ing coronary artery disease. The association identified in seroepidemiologic stud-
ies using titers to predict the incidence, distinct from the prevalence, of heart
disease, however, only variably detect an association and, when positive, only in
the range of a 20–40 percent increased risk (Dunne, 2000). While the implica-
tions of these different findings are being evaluated, the main value of these
seroepidemiologic studies may be the attention they have brought to the potential
for any association at all.
Histopathology
The next series of studies involve histopathologic examinations of the athero-
matous plaque. In the first 15 studies reported in the literature which were con-
ducted in the United States and Europe, approximately 45 percent of the total of
574 samples examined were found to contain evidence of C. pneumoniae by ei-
ther immunohistochemistry, electron microscopy, in situ polymerase chain reac-
tion (PCR) or, rarely, culture. The primary criticism of these studies has focused
on the lack of standardization of the assay techniques but, given the bulk of the
observations from these and subsequent studies, it seems likely that this pathogen
can be found in the plaque.
Because antibody titers merely suggest historical exposure to the pathogen,
there has been recent interest in the use of PCR to identify individuals that may
have an active infection with C. pneumoniae. PCR has been used to assess both
histopathologic specimens and circulating white blood cells. In four published
papers, patients with a history of coronary artery disease were more likely than
controls to have C. pneumoniae identified in circulating monocytes by PCR
(Dunne, 2000). In a fifth paper, the incidence was not significantly different but
the C. pneumoniae rRNA copy number was higher in patients with heart disease
(Berger et al., 2000). Of interest, the proportion of individuals with PCR positive
cells in these studies ranged from 9 to 60 percent in the patients with heart disease
and 2 to 46 percent in the controls. While this range of exposure may be ex-
plained by epidemiologic influences, technical concerns about assay methodolo-
gies remain and efforts at standardization have been initiated (Dowell et al., 2001).
When the technical concerns have been addressed, it will also be important to
understand why otherwise normal individuals have evidence of this pathogen
circulating in what should be a sterile space.
Animal Models
In addition to serologic and histologic evidence associating C. pneumoniae
and atherosclerosis, a number of animal models have been established. Evidence
that C. pneumoniae can either initiate or accelerate the atherosclerotic lesion has
come from work with both mice (NIH/s, ApoE-deficient, and LDL-receptor
knock-out strains) and New Zealand White rabbits. These animals generally need
to consume a high cholesterol diet in order to develop observable changes, though
it is possible, in one of the rabbit models, to observe effects without an athero-
genic diet (Fong et al., 1999). In the LDL receptor knockout mouse, intranasal
inoculation with the C. pneumoniae AR39 strain twice monthly for six months
was performed prior to sacrifice of the animals. Uninfected mice fed a high cho-
lesterol diet had a lesion area index (defined as the size of a digitized image of the
lesion divided by the aorta luminal surface and multiplied by one hundred) of 18,
while infected animals given a high cholesterol diet had an index of 42. This 130
percent increase in lesion size suggests that infection with chlamydia can acceler-
ate the growth of an atherosclerotic plaque (Hu et al., 1999).
There are limitations to the interpretation of animal models of atherosclero-
sis. In some of these models the atherosclerotic lesions observed are consistent
with a very early pathologic process that does not mirror the lesions responsible
for causing human disease. The atherosclerotic lesions in these models generally
do not rupture or lead to clinical disease in the animal. While these data do sup-
port the potential for a contribution of chlamydia to lipid accumulation at the site,
they do not provide conclusive evidence that infection will lead to plaque rupture.
cell. A series of experiments (Zhong et al., 1999; 2000), has offered some in-
sights as to why a chronically infected host cell is not destroyed by the immune
system. It appears that chlamydia can selectively inhibit IFN-gamma-inducible
MHC class I and II expression and thereby evade antigen presentation on the cell
surface. Inhibition of this process by bacterial protein synthesis inhibitors such as
chloramphenicol suggests that it is dependent on chlamydial protein synthesis.
Clinically latent infections have been demonstrated with a number of chlamy-
dia species. The blinding eye disease trachoma has occurred decades after expo-
sure to either C. trachomatis or C. pneumoniae. Infertility can result from chronic
infection of the upper genital tract with C. trachomatis, a process that can take
place over years. C. pneumoniae has also been isolated from the respiratory tract
long after resolution of an acute infection.
Atherosclerosis is now considered to be an inflammatory disease (Ross,
1999). The association of C. pneumoniae with atherogenesis is supported by the
possibility that C. pneumoniae contributes to this inflammation. Based on data
from animal models, and supported by the PCR examinations of circulating white
blood cells and histologic examinations of atherosclerotic tissue, a respiratory
tract infection could lead to dissemination of C. pneumoniae in monocytes. These
monocytes release factors that enhance the likelihood of endothelial infection
with chlamydia (Lin et al., 2000). Once infected, the endothelial cells could affect
the local arterial environment in three ways. Transendothelial migration of the
monocytes is enhanced (Molestina et al., 1999). The infected endothelial cells
release tissue factor and platelet aggregation inhibitor, which leads to enhanced
coagulability at the site. And thirdly, mitogenic factors are released through an
NF-Kβ related mechanism, leading to smooth muscle cell proliferation (Miller et
al., 2000). This triad, subendothelial monocyte accumulation, hypercoagulability
at the site of the atheroma and smooth muscle cell proliferation, is the hallmark of
an atherosclerotic plaque and, as such, provides further support for a contribution
of local C. pneumoniae infection to this inflammatory state.
pathogenesis, and better defining the lifecycle of chlamydia, and specifically the
persistent state.
There are a number of challenges to studying the use of antibiotics in clinical
coronary artery disease. While several risk factors for coronary artery disease are
already well established, the relationship between these risk factors and C.
pneumoniae infection has not been fully examined. As such associations become
better known, the use of these risk factors as selection criteria may become use-
ful. Clinical studies will need to address this problem of multiple competing risks
even while the appropriateness of controlling for these factors in any statistical
analyses, or selecting the target group of patients to treat, remains open to debate.
Many questions remain regarding antimicrobial activity within the plaque.
While there is clinical evidence that patients with either genitourinary tract or
respiratory tract infections due to chlamydia can have the clinical course of their
disease positively impacted by antibiotic intervention, it remains unknown
whether antibiotic treatment will affect either the replication or pathogenicity of
chlamydia infections in the atherosclerotic plaque. It may not be possible to either
document infection at the arterial site or substantiate a positive microbiologic
outcome. There remain concerns that to the extent that cells contain chlamydia in
the persistent state, it may not be possible to fully eradicate the organism. Stan-
dard in vitro testing may be inadequate to fully address this issue, given that the
contribution of the immune system to clearance of infected cells is not measured.
Specific concerns about the design of clinical trials also exist. The appropri-
ate patient population to treat is not clear. If C. pneumoniae is the target organ-
ism, patient selection criteria specific to the organism could be useful. Antibody
titers are a crude estimate of previous exposure but may not be adequate to select
those patients actively infected. As identification of infection within the atheroma
is not presently possible, surrogates of active infection are needed. Perhaps, in the
future, there will be a role for the measurement of C. pneumoniae DNA in circu-
lating white blood cells. As is typical with cardiovascular studies of coronary
artery disease, the event rates are typically low. Selection of patients likely to
have a primary event is critical to ensuring that any treatment effect can be ob-
served. Setting the sample size is made difficult by not having any estimate of the
potential treatment effects; in order to avoid missing a potential effect, efficacy
rates may need to be assumed to be low. These two issues require that definitive
studies be large in order to have sufficient statistical power to determine treat-
ment effects. Interpretation of the results from smaller studies is consequently
more problematic.
The results of ongoing clinical trials will be best able to answer questions
that are focused on the merits of the antibiotic intervention in the specific popula-
tion of patients enrolled, and focused on the prespecified endpoints. The results
will be compelling to the extent that the studies are adequately powered and the
chosen endpoints are clinically relevant. The ongoing trials are less likely to be
able to define the mechanism of action underlying any observed treatment effect.
TABLE 1-2 Clinical Trials with Antibiotics for Primary and Secondary
Prevention of Atherosclerosis Diseases
Study Population N Antibiotic Endpoint Outcome
stroke, or death.
REFERENCES
Anderson JL, Muhlestein JB, Carlquist J, Allen A, Trehan S, Nielson C, Hall S, Brady J, Egger M,
Horne B, Lim T. 1999. Randomized secondary prevention trial of azithromycin in patients with
coronary artery disease and serological evidence for Chlamydia pneumoniae infection: the
azithromycin in coronary artery disease: elimination of myocardial infection with chlamydia
(ACADEMIC) study. Circulation 99:1540–1547.
Beatty WL, Morrison RP, Byrne GI. 1994. Persistent chlamydiae: from cell culture to a paradigm for
chlamydial pathogenesis. Microbiological Reviews 58:686–699.
Berger M, Schroder B, Daeschlin G, Schneider W, Busjahn A, Buchalow I, Luft FC, Haller H. 2000.
Chlamydia pneumoniae DNA in non-coronary atherosclerotic plaques and circulating
leokocytes. Journal of Laboratory and Clinical Medicine 136:194–200.
Danesh J. 1999. Coronary heart disease, Helicobacter pylori, dental disease, Chlamydia pneumoniae,
and cytomegalovirus: meta-analyses of prospective studies. American Heart Journal 138:S434–
437.
Dowell SF, Peeling RW, Boman J, Carlone GM, Fields BS, Guarner J, Hammerschlag MR, Jackson
LA, Kuo CC, Maass M, Messmer TO, Talkington DF, Tondella ML, Zaki SR. 2001. Standard-
izing Chlamydia pneumoniae assays: recommendations from the Centers for Disease Control
and Prevention (USA) and the Laboratory Centre for Disease Control (Canada). Clinical Infec-
tious Diseases 33:492–503.
Dunne M. 2000. The evolving relationship between Chlamydia pneumoniae and atherosclerosis. Cur-
rent Opinion in Infectious Diseases 13(6):583–591.
Fong IG, Chiu B, Viira E, Jang D, Fong MW, Peeling R, Mahony JA. 1999. Can an Antibiotic
(macrolide) prevent Chlamydia pneumoniae-induced atherosclerosis in a rabbit model? Clinical
and Diagnostic Laboratory Immunology 6:891–894.
Gilbert A and Lion G. 1889. Artérites infectieuses expérimentales. Comptes Rendus Hebdomadaires
des Séances et Mémoires de la Société de Biologie. 41:583–584.
Gupta S, Leatham EW, Carrington D, Mendall MA, Kaski JC, Camm AJ. 1997. Elevated Chlamydia
pneumoniae antibodies, cardiovascular events, and azithromycin in male survivors of myocar-
dial infarction. Circulation 96:404–407.
Gurfinkel EG, Bozovich G, Daroca A, Beck E, Mautner B. 1997. Randomised trial of roxithromycin
in non-Q-wave coronary syndromes: ROXIS Pilot Study. ROXIS Study Group. [comment].
Lancet 350:404–407.
Hu H, Pierce GN, Zhong G. 1999. The atherogenic effects of chlamydia are dependent on serum
cholesterol and specific to Chlamydia pneumoniae. Journal of Clinical Investigation 103:747–
753.
Lin TM, Campbell LA, Rosenfeld ME, Kuo CC. 2000. Monocyte-endothelial cell coculture enhances
infection of endothelial cells with Chlamydia pneumoniae. Journal of Infectious Diseases
181:1096–1100.
Miller SA, Selzman CH, Shames BD, Barton HA, Johnson SM, Harken AH. 2000. Chlamydia
pneumoniae activates nuclear factor kappaB and activator protein 1 in human vascular smooth
muscle and induces cellular proliferation. Journal of Surgical Research 90:76–81.
Molestina RE, Miller RD, Ramirez, JA Summersgill JT. 1999. Infection of human endothelial cells
with Chlamydia pneumoniae stimulates transendothelial migration of neutrophils and mono-
cytes. Infection & Immunity 67:1323–1330.
Neumann FA, Kastrati A, Miethke T, Pogatsa-Murray G, Mehilli J, Valina C, Jogethaei N, da Costa
CP, Wagner H. Schomig A. 2001. Treatment of Chlamydia pneumoniae infection with
roxithromycin and effect on neointima proliferation after coronary stent placement (ISAR-3): a
randomised, double-blind, placebo-controlled trial. Lancet 357:2085–2089.
Nieto FJ. 1998. Infections and atherosclerosis: new clues from and old hypothesis? American Journal
of Epidemiology 48:937–948.
Nieto FJ. 1999. Viruses and atherosclerosis: a critical review of the epidemiologic evidence. Ameri-
can Heart Journal 138:S453–460.
Reiner Z. 2002. Azithromycin in the secondary prevention of adverse cardiovascular events in C.
pneumoniae-positive post myocardial infarction patients (CROAATS). Paper presented at the
Sixth International Conference on the Macrolides, Azalides, Streptogramins, Ketolides, and
Oxazolidinones, Bologna, Italy, January 23-25, 2002.
Ross R. 1999. Atherosclerosis—an inflammatory disease. [comment]. New England Journal of Medi-
cine 340:115–126.
Saikku P, Leinonen M, Mattila K, Ekman MR, Nieminen MS, Makela PH, Huttunen JK, Valtonen V.
1988. Serological evidence of an association of a novel Chlamydia, TWAR, with chronic coro-
nary heart disease and acute myocardial infarction. Lancet 2:983–986.
Wiesli P, Czerwenka W, Meniconi A, Maly F, Hoffman U, Vetter W, Schulthess G. 2002.
Roxithromycin treatment prevents progression of peripheral arterial occlusive disease in
Chlamydia pneumoniae seropositive men: a randomized, double-blind, placebo-controlled trial.
Circulation 105:2646–2652.
Zahn R, Schneider S, Frilling B, Seidl K, Tebbe U, Weber M, Gottwik M, Altmann E, Seidel F, Rox
J, Hoffler U, Neuhaus KL, Senges J. Working Group of Leading Hospital Cardiologists. 2003.
Antibiotic therapy after acute myocardial infarction: a prospective randomized study. Circula-
tion 107:1253–1259.
Zhong G, Fan T, Lui L. 1999. Chlamydia inhibits interferon gamma-inducible major histocompatibil-
ity complex class II expression by degradation of upstream stimulatory factor 1. Journal of
Experimental Medicine 189:1931–1938.
Zhong G, Liu L, Fan T, Fan P, Ji H. 2000. Degradation of transcription factor RFX5 during the
inhibition of both constitutive and interferon gamma-inducible major histocompatibility com-
plex class I expression in chlamydia-infected cells. Journal of Experimental Medicine 191:1525–
1534.
DEMYELINATING DISEASES
Richard T. Johnson, M.D.
Johns Hopkins University, Baltimore, MD
Box 1-1
Possible Mechanisms of Virus-Induced Demyelination
ing diseases have been pursued in hopes of discovering a role of viruses in mul-
tiple sclerosis, but this goal remains elusive.
Animal Models
Animal viruses can produce acute, chronic, and relapsing/remitting demyeli-
nating central nervous system diseases in their natural or experimental hosts (see
Table 1-3). The best model for human postinfectious encephalomyelitis (acute
disseminated encephalomyelitis), however, is not a viral infection but experimen-
tal autoimmune encephalomyelitis (EAE) induced by injection of myelin proteins
with Freund’s adjuvant. The latency, clinical disease, pathology and immuno-
logical features of these two diseases are similar.
Progressive multifocal leucoencephalopathy (PML) in macaque monkeys
caused by SV40 virus is the animal equivalent of PML in man caused by the
related JC virus. As in the human disease, the disease evolves in latently infected
animals when other infections or illnesses cause immunodeficiencies. Four natu-
rally occurring infections in their native hosts have been the most widely studied
models of virus-induced demyelination. Theiler’s virus, a picornavirus, and JHM
virus, a murine coronavirus, were both originally recovered from paralyzed mice;
canine distemper, a morbillivirus closely related to measles virus, has long been
recognized to cause demyelination in a subacute encephalitis called “old dog dis-
ease”; and visna virus, a natural retrovirus infection of sheep, causes relapsing
and remitting disease with multifocal demyelinating lesions after a long incuba-
tion period. Visna and a related caprine lentivirus (caprine arthritis-ecephalitis
virus) best simulate multiple sclerosis, but they have not been widely exploited
because of the need to use sheep or goats as the experiment animals. In these
lentivirus diseases, infection is limited to macrophages and microglia, and demy-
elination is thought to result from cytokines released by infected cells.
syndrome (different pathology) and acute cerebellar ataxia (unknown pathology) and the rare docu-
mentation of perivenular demyelinating disease.
ADEM constitutes less than one-tenth of the cases of acute encephalitis and now
is most common after nonspecific upper respiratory infections.
The hazard of ADEM after inoculation of vaccinia virus to protect against
smallpox has returned as an issue, since resumption of vaccination is being con-
sidered to counter the possible use of smallpox as a terrorist weapon. A very high
rate of complications in one Dutch vaccination program was presumably due to
use of a more encephalitogenic strain; the low rates during the mass vaccination
in New York in 1947 probably reflects poor surveillance. In Great Britain, during
the more recent outbreak of smallpox in 1962, a rate of postvaccinal encephalo-
myelitis of 1 per 20,000 was estimated, and CDC retrospective surveys estimated
1 per 200,000 in the United States prior to the discontinuation of vaccination.
However, the risk of ADEM when starting vaccination after a hiatus of 30 years
is uncertain, since neurologic complications are more frequent with primary vac-
cination and higher in persons over the age of 20 years.
The clinical presentation of ADEM usually follows the antecedent exanthem
or respiratory or gastrointestinal symptoms by 5 to 21 days. Typically postmeasles
encephalomyelitis occurs 5 to 7 days after the rash when the child is returning to
normal activity. There is the abrupt recurrence of fever, depression of conscious-
ness, and appearance of multifocal neurological findings. The spinal fluid usually
contains myelin basic protein, often shows increased pressure and a mild pleocy-
tosis (but in about one-third of cases, no increased cellularity is found). The MRI
may show very dramatic changes with multiple enhancing lesions in the white
matter.
The histopathology of fatal cases shows perivenular inflammation and de-
myelination throughout the brain and spinal cord. In most instances, virus is not
found within the nervous system. For example, in measles, virus is seldom recov-
erable after the rash which corresponds with the humoral immune response. In
measles, deaths occurring at or before the time of rash, measles virus has been
found in cerebrovascular endothelial cells by in situ PCR; but no virus antigen or
nucleic acid has been found in cells of the CNS in patients dying of encephalomy-
elitis.
The pathogenesis of ADEM is related to infection of immunocompetent cells
and the alteration of immune responses. In both postmeasles and postvaricella
disease activated peripheral blood lymphocytes responsive to myelin basic pro-
tein have been demonstrated. The autoimmune response against CNS myelin ap-
pears to occur without the prerequisite of infection of CNS cells. ADEM appears
to be an autoimmune disease very similar to experimental autoimmune encepha-
lomyelitis.
dinarily rare disease. With the emergence of AIDS over the past two decades,
PML has become a common opportunistic infection causing death in 3–5 percent
of AIDS patients.
The clinical presentation is on a background of severe immunosuppression.
Multifocal neurological symptoms and signs develop insidiously and usually fol-
low an ingravescent course to death. With introduction of HAART therapy and
recovery of T4 counts, stabilization and even improvement has been reported.
There is no fever, no nuchal rigidity, and usually no pleocytosis. A very charac-
teristic MRI pattern is seen, however, with nonenhancing multifocal lesions in
the subcortical white matter.
The neuropathological changes are unique. Plaques of demyelination are seen
preferentially in the grey–white junction. Histologically inflammation is slight or
absent. In areas of demyelination, axons are relatively spared and oligodendro-
cytes are lost. Surrounding these foci, oligodendrocytes are enlarged and contain
intranuclear inclusions. Astocytosis is intense, and many astrocytes contain bi-
zarre mitotic figures and multiple nuclei resembling malignant cells.
Electron microscopic examination of the oligodendrocyte inclusions reveal
profuse pseudocrystalline arrays of papovaviruses. Only occasional viral particles
are seen in astrocytes but they express papovavirus T antigen. JC virus, an ubiq-
uitous human papovavirus, has been associated with almost all cases.
The pathogenesis of demyelination in PML is the opposite of that in ADEM.
JC virus causes an asymptomatic persistent infection in most persons. With in-
tense immunosuppression the virus in some patients is transported to brain, prob-
ably in B cells. With massive replication in oligodendrocytes these cells are de-
stroyed with secondary loss of myelin. There is no evidence of infection of
neurons. Semipermissive infection of astrocytes leads to limited virus production
but many astrocytic changes and proliferation resemble transformation. The tat
protein of HIV may transactivate JC virus accounting for the unique frequency of
PML in HIV-infected patients.
Multiple Sclerosis
A viral cause for multiple sclerosis has been postulated for over 100 years.
Over the past half century this speculation has been highlighted by 3 types of
studies. First, epidemiological evidence implicates childhood exposure factors
(possibly viral infections) in the genesis of multiple sclerosis, and natural history
studies have related “virus-like illnesses” to exacerbations of the disease. Second,
studies of human and animal viral infections have documented that these infec-
tions can have incubation periods of years, cause remitting and relapsing disease
and can cause myelin destruction mediated by a variety of mechanisms. Third,
laboratory studies of patients with multiple sclerosis consistently show that such
patients have greater antibody responses to a variety of viruses than controls and
this includes intrathecal antibody synthesis. This is not to deny the clear-cut ge-
REFERENCES
These references are not specifically cited in text.
Buljevac D, Flach HZ, Hop WC, Hijdra D, Laman JD, Savelkoul HF, van Der Meche FG, van Doorn
PA, Hintzen RQ. 2002. Prospective study on the relationship between infections and multiple
sclerosis exacerbations. Brain 125:952–960.
Gieffers J, Pohl D, Treib J, Dittmann R, Stephan C, Klotz K, Hanefeld F, Solbach W, Haass A, Maass
M. 2001. Presence of Chlamydia pneumoniae DNA in the cerebral spinal fluid is a common
phenomenon in a variety of neurological diseases and not restricted to multiple sclerosis. Annals
of Neurology 49:585–589.
Gonzalez-Scarano F and Rima B. 1999. Infectious etiology in multiple sclerosis: the database contin-
ues. Trends in Microbiology 7:475–477.
Johnson RT. 1994. The virology of demyelinating diseases. Annals of Neurology 36:S54–S60.
Johnson RT. 1998. Viral Infections of the Nervous System 2nd Ed. Philadelphia: Lippincott-Raven.
Johnston JB, Silva C, Holden J, Warren KG, Clark AW, Power C. 2001. Monocyte activation and
differentiation augments human retrovirus expression: implications for inflammatory brain dis-
eases. Annals of Neurology 50:434–442.
Wandinger K, Jabs W, Siekhaus A, Bubel S, Trillenberg P, Wagner H, Wessel K, Kirchner H, Hennig
H. 2000. Association between clinical disease activity and Epstein-Barr virus reactivation in
MS. Neurology 55:178–184.
Yao SY, Stratton CW, Mitchell WM, Sriram S. 2000. CSF oligoclonal bands in MS include antibod-
ies against Chlamydophila antigens. Neurology 56:1168–1176.
Several viruses have been proposed as infectious triggers of diabetes, but the
enteroviruses (family Picornaviridae, genus Enterovirus) are the subject of the
most intense scrutiny at present (Leinikki, 1998; Hyöty et al., 1998). Numerous
studies have provided evidence for an association between enterovirus infection
and prediabetic autoimmunity or clinical diabetes. Diabetes incidence has been
epidemiologically linked to the incidence of enteroviral meningitis or enterovirus
outbreaks (Karvonen et al., 1993). Serologic studies have shown that there is a
correlation between enterovirus seroprevalence in patients with prediabetic au-
toimmunity or diabetes, compared to unaffected control individuals (Hiltunen et
al., 1997; Helfand et al., 1995). Direct enterovirus detection in pancreas, blood,
serum, or stool has suggested a temporal correlation between enterovirus infec-
tion and onset of diabetes (Yoon et al., 1979; Andreoletti et al., 1997; Clements et
al., 1995).
Enteroviruses are among the most common of human viruses, infecting an
estimated 50 million people annually in the United States and possibly a billion
or more annually worldwide (Morens and Pallansch, 1995; Pallansch and Roos,
2001). Most infections are inapparent, but enteroviruses may cause a wide spec-
trum of acute disease, including mild upper respiratory illness (common cold),
febrile rash (hand, foot, and mouth disease and herpangina), aseptic meningitis,
pleurodynia, encephalitis, acute flaccid paralysis (paralytic poliomyelitis), and
neonatal sepsis-like disease. Enterovirus infections result in 30,000 to 50,000
hospitalizations per year in the United States, with aseptic meningitis cases ac-
counting for the vast majority of the hospitalizations (Pallansch and Roos, 2001).
In addition to these acute illnesses, enteroviruses have also been associated with
severe chronic diseases such as myocarditis (Martino et al., 1995; Kim et al.,
2001), Type 1 diabetes mellitus (Leinikki, 1998; Rewers and Atkinson, 1995),
and neuromuscular diseases (Dalakas, 1995). Enteroviruses are transmitted pri-
marily by the fecal-oral route but respiratory transmission to close contacts may
also be important. The incubation period between infection and onset of symp-
toms is usually 4–7 days. The intestinal mucosa or upper respiratory tract is the
site of primary infection, with secondary spread to the central nervous system and
other tissues. Viremia is usually short-lived, often waning before the onset of
symptoms, except in very young children. Virus is excreted in the stool for up to
8 weeks (average 2–4 weeks) but maximal virus shedding occurs before the onset
P1 P2 P3
5'NTR 3'NTR
VP4 VP2 VP3 VP1 2A 2B 2C 3A 3B 3C 3D
5’ NTR
RT-snPCR
012 011
040
187 222
VP3/VP1 188 VP1/2A
189
292
FIGURE 1-6 Schematic representation of the enterovirus genome, indicating regions that
have been targeted for development of PCR diagnostics. The genome is a positive-stranded,
polyadenylated RNA of ~7400 nucleotides, with a viral protein (3B/VPg) covalently linked
to the 5′ end. The genome is divided into five functional regions: the 5′ non-translated
region (NTR) (control of viral translation initiation and initiation of positive-strand RNA
synthesis); P1 (encodes the structural proteins that comprise the virus capsid); P2 and P3
(encode the non-structural proteins involved in RNA replication, proteolytic processing of
polyprotein, and host cell shut-down); and 3′ NTR (involved in initiation of negative-
strand RNA synthesis).
A Dilution: 10 x
M –1 –2 –3 –4 –5 –6 –7 –8 –9 –10 M
x
B Dilution: 10
M –1 –2 –3 –4 –5 –6 –7 –8 –9 –10 M
antisense primer. PCR was performed using a single round of amplification (con-
ventional PCR) or two rounds of amplification (semi-nested PCR). The second
round of the semi-nested amplification used the same primers as the conventional
PCR. Amplification products were visualized by polyacrylamide gel electrophore-
sis and staining with ethidium bromide. The RT-snPCR method (see Figure 1-
7B) was approximately 10,000-fold more sensitive than the conventional RT-
PCR (see Figure 1-7A). The 10–7 dilution corresponds to less than 20 infectious
virus particles.
Enterovirus infection elicits a serotype-specific immune response directed
against epitopes on the surface of the viral capsid. Mucosal immunity is most
important. Antibody alone fully protects from disease, probably by limiting virus
spread from the gut, but antibody does not necessarily protect from infection. The
virus-specific T-cell response, directed against epitopes on both the structural and
non-structural proteins, is probably involved in virus clearance but it is not needed
for protection. Antigenic sites are located in each of the three enterovirus struc-
tural proteins, VP1, VP2, and VP3 (Minor, 1990; Mateu, 1995), but the epitopes
responsible for serotype specificity have not been identified. Since the picornavi-
rus VP1 protein contains a number of immunodominant neutralization domains,
we hypothesized that VP1 sequence should correspond with neutralization prop-
erties (serotype) (Oberste et al., 1999b). Due to the high frequency of recombina-
tion among picornaviruses (Kopecka et al., 1995; King, 1988; Santti et al., 1999),
REFERENCES
Andreoletti L, Hober D, Hober-Vandenberghe C, Belaich S, Vantyghem MC, Lefebvre J, Wattre P.
1997. Detection of coxsackie B virus RNA sequences in whole blood samples from adult pa-
tients at the onset of type I diabetes mellitus. Journal of Medical Virology 52:121–127.
Clements GB, Galbraith DN, Taylor KW. 1995. Coxsackie B virus infection and onset of childhood
diabetes. Lancet 346:221–223.
Committee on Enteroviruses. 1962. Classification of human enteroviruses. Virology 16:501–504.
Dalakas MC. 1995. Enteroviruses and human neuromuscular diseases. Pp. 387–398 in Human En-
terovirus Infections, HA Rotbart, ed. Washington, DC: ASM Press.
Helfand RF, Gary HE Jr, Freeman CY, Anderson LJ, Pallansch MA. 1995. Serologic evidence of an
association between enteroviruses and the onset of type 1 diabetes mellitus. Pittsburgh Diabetes
Research Group. The Journal of Infectious Diseases 172:1206–1211.
Hiltunen M, Hyoty H, Knip M, Ilonen J, Reijonen H, Vahasalo P, Roivainen M, Lonnrot M, Leinikki
P, Hovi T, Akerblom HK. 1997. Islet cell antibody seroconversion in children is temporally
associated with enterovirus infections. Childhood Diabetes in Finland (DiMe) Study Group. The
Journal of Infectious Diseases 175:554–560.
Hyöty H, Hiltunen M, Lonnrot M. 1998. Enterovirus infections and insulin dependent diabetes mel-
litus—evidence for causality. Clinical and Diagnostic Virology 9:77–84.
Hyypiä T, Horsnell C, Maaronen M, Khan M, Kalkkinen N, Auvinen P, Kinnunen L, Stanway G.
1992. A distinct picornavirus group identified by sequence analysis. Proceedings of the Na-
tional Academy of Sciences 89:8847–8851.
Karvonen M, Tuomilehto J, Libman I, LaPorte R. 1993. A review of the recent epidemiological data
on the worldwide incidence of type 1 (insulin-dependent) diabetes mellitus. World Health
Organisation DIAMOND Project Group. Diabetologia 36:883–892.
Kim KS, Hufnagel G, Chapman NM, Tracy S. 2001. The group B coxsackieviruses and myocarditis.
Reviews in Medical Virology 11:355–368.
King AMQ. 1988. Genetic recombination in positive strand RNA viruses. Pp. 149–165 in RNA Ge-
netics, E Domingo, JJ Holland, and P Ahlquist, eds. Boca Raton, FL: CRC Press, Inc.
King AMQ et al. 2000. Picornaviridae. Pp. 657–678 in Virus Taxonomy: Seventh Report of the
International Committee on Taxonomy of Viruses, MH Van Regenmortel et al., eds. San Diego:
Academic Press.
Kopecka H, Brown B, Pallansch M. 1995. Genotypic variation in coxsackievirus B5 isolates from
three different outbreaks in the United States. Virus Research 38:125–136.
Leinikki P. 1998. Viruses and type 1 diabetes: elusive problems and elusive answers. Clinical and
Diagnostic Virology 9:65–66.
Lim KA and Benyesh-Melnick M. 1960. Typing of viruses by combinations of antiserum pools.
Application to typing of enteroviruses (coxsackie and ECHO). Journal of Immunology 84:309–
317.
Martino TA et al. 1995. Enteroviral myocarditis and cardiomyopathy: a review of clinical and experi-
mental studies. Pp. 291–351 in Human Enterovirus Infections, HA Rotbart, ed. Washington,
DC: ASM Press.
Marvil P, Knowles NJ, Mockett AP, Britton P, Brown TD, Cavanagh D. 1999. Avian encephalomy-
elitis virus is a picornavirus and is most closely related to hepatitis A virus. Journal of General
Virology 80:653–662.
Mateu MG. 1995. Antibody recognition of picornaviruses and escape from neutralization. Virus Re-
search 38:1–24.
Minor PD. 1990. Antigenic structure of picornaviruses. Current Topics in Microbiology and Immu-
nology 161:121–154.
Morens DM and Pallansch MA. 1995. Epidemiology. Pp. 3–23 in Human Enterovirus Infections, HA
Rotbart, ed. Washington, DC: ASM Press.
Niklasson B, Kinnunen L, Hornfeldt B, Horling J, Benemar C, Hedlund KO, Matskova L, Hyypia T,
Winberg G. 1999. A new picornavirus isolated from bank voles (Clethrionomys glareolus).
Virology 255:86–93.
Oberste MS, Maher K, Kilpatrick DR, Flemister MR, Brown BA, Pallansch MA. 1999a. Typing of
human enteroviruses by partial sequencing of VP1. Journal of Clinical Microbiology 37:1288–
1293.
Oberste MS, Maher K, Kilpatrick DR, Pallansch MA. 1999b. Molecular evolution of the human
enteroviruses: correlation of serotype with VP1 sequence and application to picornavirus classi-
fication. Journal of Virology 73:1941–1948.
Oberste MS, Maher K, Flemister MR, Marchetti G, Kilpatrick DR, Pallansch MA. 2000. Comparison
of classic and molecular approaches for the identification of “untypable” enteroviruses. Journal
of Clinical Microbiology 38:1170–1174.
Oberste MS, Schnurr D, Maher K, al-Busaidy S, Pallansch M. 2001. Molecular identification of new
picornaviruses and characterization of a proposed enterovirus 73 serotype. Journal of General
Virology 82:409–416.
Pallansch MA and Roos RP. 2001. Enteroviruses: polioviruses, coxsackieviruses, echoviruses, and
newer enteroviruses. Pp. 723–775 in Fields Virology, DM Knipe and PM Howley, eds. Phila-
delphia: Lippincott Williams and Wilkins.
Panel for Picornaviruses. 1963. Picornaviruses: classification of nine new types. Science 141:153–
154.
Rewers M and Atkinson M. 1995. The possible role of enteroviruses in diabetes mellitus. Pp. 353–
385 in Human Enterovirus Infections, HA Rotbart, ed. Washington, DC:ASM Press.
Rotbart HA and Romero JR. 1995. Laboratory diagnosis of enteroviral infections. Pp. 401–418 in
Human Enterovirus Infections, HA Rotbart, ed. Washington, DC: ASM Press.
Rotbart HA, Ahmed A, Hickey S, Dagan R, McCracken GH Jr, Whitley RJ, Modlin JF, Cascino M,
O’Connell JF, Menegus MA, Blum D. 1997. Diagnosis of enterovirus infection by polymerase
chain reaction of multiple specimen types. The Pediatric Infectious Disease Journal 16:409–
411.
Santti J, Hyypia T, Kinnunen L, Salminen M. 1999. Evidence of recombination among enteroviruses.
Journal of Virology 73:8741–8749.
See DM and Tilles JG. 1998. The pathogenesis of viral-induced diabetes. Clinical and Diagnostic
Virology 9:85–88.
Vosloo W, Knowles NJ, Thomson GR. 1992. Genetic relationships between southern African SAT-2
isolates of foot-and-mouth-disease virus. Epidemiology and Infection 109:547–558.
Yamashita T, Sakae K, Tsuzuki H, Suzuki Y, Ishikawa N, Takeda N, Miyamura T, Yamazaki S. 1998.
Complete nucleotide sequence and genetic organization of Aichi virus, a distinct member of the
Picornaviridae associated with acute gastroenteritis in humans. Journal of Virology 72:8408–
8412.
Yang CF, De L, Yang SJ, Ruiz Gomez J, Cruz JR, Holloway BP, Pallansch MA, Kew OM. 1992.
Genotype-specific in vitro amplification of sequences of the wild type 3 polioviruses from
Mexico and Guatemala. Virus Research 24:277–296.
Yoon JW. 1990. The role of viruses and environmental factors in the induction of diabetes. Current
Topics in Microbiology and Immunology 164:95–123.
Yoon JW, Austin M, Onodera T, Notkins AL. 1979. Isolation of a virus from the pancreas of a child
with diabetic ketoacidosis. New England Journal of Medicine 300:1173–1179.
*The research described in this presentation was supported by the Stanley Medical Research Insti-
tute.
Box 1-2
Clinical and Epidemiological Features of Schizophrenia
Positive symptoms:
• Hallucinations
• Delusions
• Disordered thinking
Negative symptoms:
• Withdrawal
• Amotivation
• Retricted expressiveness
• Symptomatic improvement
• High rate of side effects
• Do not affect overall disease process
in schizophrenia has focused on possible genetic etiologies. The rationale for this
approach is based on numerous studies indicating a strong risk associated with
having a biological parent with this disease. Extensive genetic analyses of fami-
lies with schizophrenia have led to the identification of a number of broad ge-
nomic regions which appear to be inherited in a non-random fashion by individu-
als with schizophrenia. However, despite intensive searches, no genes of major,
or even minor effect, have been consistently linked to the schizophrenia pheno-
type (see Box 1-3).
Due to the limited success of genetic investigations, there has been renewed
interest in the role of environmental factors in the etiopathogenesis of schizophre-
nia. This approach derives its rationale from a number of epidemiological studies
which indicate that environmental factors may contribute to the risk of schizo-
phrenia in some individuals. Many of these studies identify environmental events
occurring during fetal development and early infancy as risk factors for the devel-
opment of schizophrenia in adult life. Risk factors which have been identified
Box 1-3
Genetics of Schizophrenia
DNA
Scz
Ctr
Herv-W
HERVw GTTCAGGGATAGCCCCCATCTATTTGGCCAGGCATTAGCCCAAGACTTGAGTCAATTCTCATACCTGGACACTCTTGTCCTTCAG
C1 C
A1 A TG
A2 A TG
A3 C C G- G
A4 A T C G TG
A5 A
A6 T CA TA C G TG
FIGURE 1-9 Endogenous retrovirus was found in the CSFs of approximately 30 percent
of individuals with recent-onset schizophrenia and 5 percent of individuals with chronic
forms of the disease.
SOURCE: Karlsson et al. (2001).
transcription has also been found to be increased in the CSFs of individuals with
multiple sclerosis (Perron et al., 1997). It has also been demonstrated to be active
during human fetal development and to encode a protein with syncytium forming
activity in the human placenta (Mi et al., 2000). Furthermore , the envelope pro-
tein of HERV-W is capable of causing polyclonal T-lymphocyte activation (Per-
ron et al., 2001). HERV-W may thus also provide a link between environmental
events active both during fetal development and adult life.
The transcription of endogenous retroviruses can be activated by a number of
infectious agents and other environmental factors. We have examined the preva-
lence of potential activating infections in different stages of schizophrenia. We
have found an increased level of antibodies to Toxoplasma gondii in individuals
with the recent onset of schizophrenia (see Figure 1-10) (Yolken et al., 2001).
This finding is consistent with epidemiological studies documenting an increased
rate in schizophrenia in individuals who were exposed to cats in early life (Torrey
et al., 2000). We have also found that serological evidence of infection with Her-
pes Simplex Virus Type 1 and Toxoplasma gondii are associated with increased
levels of cognitive and memory impairments in individuals with established forms
of schizophrenia (Dickerson et al., 2003b). We also examined the possible asso-
ciation between infections in pregnancy in the occurrence of schizophrenia in
2 1.00
0.70
0.60
1 0.50
0.40
0.30
0.20
0.10
0 0.00
Case Control Case Control Case Control
later life. These analyses were accomplished by the testing of sera which had
been obtained from healthy pregnant women as part of the National Collaborative
Perinatal Study performed in the United States during the 1950s and 1960s. Ini-
tial analyses of this cohort indicates that the offspring of mothers who had evi-
dence of infection, as indicated by increased levels of IgG, IgM, and of IgG
antibodies to Herpes Simplex Virus type 2, have higher rates of schizophrenia in
adult life (see Figure 1-11) (Buka et al., 2001). There was also a risk of schizo-
phrenia associated with IgM antibodies to Toxoplasma gondii, although the anti-
genic source of these antibodies is still under investigation.
These studies indicate that infectious agents play a role in the generation of
schizophrenia in some individuals. The activation of endogenous retroviruses
within the central nervous system is likely to be one of several mechanisms by
means of which infections can lead to disease. If this is the case, it is possible that
the treatment of infectious agents which activate retroviral transcription may be
capable of modulating the course of disease at different times in the lifelong
course of disease. For example, the treatment of active infection with herpes sim-
plex virus might prevent endogenous retrovirus activation due to this organism. It
is of note in this regard that several of the medications which are commonly used
for the treatment of schizophrenia also have the ability to inhibit the replication of
infectious agents (Jones-Brando et al., 1997). Preliminary analysis of a clinical
FIGURE 1-11 Association between HSV infections in pregnant women and the occur-
rence of schizophrenia in their adult offspring. The adult offspring of mothers whose sera
showed evidence of HSV infection during pregnancy have higher rates of schizophrenia
than the adult offspring of mothers whose sera did not show such evidence during pregnancy.
SOURCE: Reprinted from Buka et al. (2001).
REFERENCES
Buka SL, Tsuang MT, Torrey EF, Klebanoff MA, Bernstein D, Yolken RH. 2001. Maternal infections
and subsequent psychosis among offspring. Archives of General Psychiatry 58:1032–1037.
Dickerson F, Boronow JJ, Stallings C, Origoni A, Yolken R. 2003a. Valacyclovir reduces symptoms
in individuals with schizophrenia who are seropositive for cytomegalovirus. Paper presented at
the International Congress on Schizophrenia Research, Colorado Springs, March 2003.
Dickerson FB, Boronow JJ, Stallings C, Origoni AE, Ruslanova I, Yolken RH. 2003b. Association of
serum antibodies to herpes simplex virus 1 with cognitive deficits in individuals with schizo-
phrenia. Archives of General Psychiatry 60:466–472.
Johnston-Wilson NL, Bouton CM, Pevsner J, Breen JJ, Torrey EF, Yolken RH. 2001. Emerging
technologies for large-scale screening of human tissues and fluids in the study of severe psychi-
atric disease. The International Journal of Neuropsychopharmacology 4:83–92.
Jones-Brando LV, Buthod JL, Holland LE, Yolken RH, Torrey EF. 1997. Metabolites of the
antipsycotic agent clozapine inhibit the replication of human immunodeficiency virus type 1.
Schizophrenia Research 25:63–70.
Karlsson H, Bachmann S, Schroder J, McArthur J, Torrey EF, Yolken RH. 2001. Retroviral RNA
identified in the cerebrospinal fluids and brains of individuals with schizophrenia. Proceedings
of the National Academy of Sciences 98:4634–4639.
Mi S, Lee X, Li X, Veldman GM, Finnerty H, Racie L, LaVallie E, Tang XY, Edouard P, Howes S,
Keith JC Jr, McCoy JM. 2000. Syncytin is a captive retroviral envelope protein involved in
human placental morphogenesis. Nature 403:785–789.
Mortensen PB, Pederson CB, Westergaard T, Wohlfahrt J, Ewald H, Mors O, Andersen PK, Melbye
M. 1999. Effects of family history and place and season of birth on the risk of schizophrenia.
New England Journal of Medicine 340:603–608.
Perron H, Garson JA, Beden F, Beseme F, Paranhos-Baccala G, Komurian-Pradel F, Mallet F, Tuke
PW, Voisset C, Blond JL, Lalande B, Seigneurin JM, Mandrand B. 1997. Molecular identifica-
tion of a novel retrovirus repeatedly isolated from patients with multiple sclerosis. The Collabo-
rative Research Group on Multiple Sclerosis. Proceedings of the National Academy of Sciences
94:7583–7588.
Perron H, Jouvin-Marche E, Michael M, Quanonian-Paroz A, Camelo S, Dumon A, Jolivet-Reynaud
C, Marcel F, Souillet Y, Barel E, Gebeihrer L, Santoro L, Marcel S, Seigreurin JM, Marche PN,
Lafon M. 2001. Multiple sclerosis retrovirus particles and recombinant envelope trigger on
abnormal immune response in vitro, by inducing polyclonal Vbetal 6 T-lymphocyte activation.
Virology 287:321–332.
Torrey EF and Yolken RH. 1998. At issue: is household crowding a factor for schizophrenia and
bipolar disorder. Schizophrenia Bulletin 24:321–324.
Torrey EF and Yolken RH. 2000. Familial and genetic mechanisms in schizophrenia. Brain Research
Reviews 31:113–117.
Torrey EF, Miller J, Rawlings R, Yolken RH. 1997. Seasonality of births in schizophrenia and bipolar
disorder: a review of the literature. Schizophrenia Research 28:1–38.
Torrey EF, Rawlings R, Yolken RH. 2000. The antecedents of psychoses: a case-control study of
selected risk factors. Schizophrenia Research 46:17–23.
Yee F and Yolken RH. 1997. Identification of differentially expressed RNA transcripts in neuropsy-
chiatric disorders. Biological Psychiatry 41:759–761.
Yolken RH and Torrey EF. 1995. Viruses, schizophrenia and bipolar disorder. Clinical Microbiology
Reviews 8:131–145.
Yolken RH, Karlsson H, Yee F, Johnston-Wilson NL, Torrey EF. 2000. Endogenous retroviruses and
schizophrenia. Brain Research Reviews 31:193–199.
Yolken RH, Bachmann S, Rouslanova I, Lillehoj E, Ford G, Torrey EF, Schroeder J. 2001. Antibod-
ies to Toxoplasma gondii in individuals with first-episode schizophrenia. Clinical Infectious
Diseases 32:842–844.
and numerous factors can contribute. Over the years, it has been shown that envi-
ronmental factors (e.g., radiation and heavy metals) and lifestyle habits (e.g.,
tobacco smoking) can increase the rates of particular cancers. In some cases,
infectious agents have been identified as causative to certain cancers. In animal
model systems, several classes of viruses have been shown to cause cancers,
including retroviruses (e.g., avian sarcoma/leukemia viruses and murine leuke-
mia viruses) and small DNA viruses (e.g., polyoma and SV40). Viruses associ-
ated with human cancers include retroviruses (human T-cell leukemia virus—
adult T-cell leukemia), human papillomavirus (cervical cancer), hepatitis virus
types B and C (hepatocellular carcinoma) and gamma herpes viruses (EBV—
lymphomas and nasopharyngeal carcinoma; HHV8—Kaposi’s sarcoma). In ad-
dition, the bacterium Helicobacter pylori has been associated with stomach can-
cer. Proving the involvement of viruses and bacteria in human cancers has
typically taken many years. Steps involved include appreciating an epidemiologi-
cal pattern of the particular cancer suggesting an infectious agent, identification
of an infectious agent whose distribution fits the epidemiological pattern, and
ultimately demonstrating in an animal model or in vitro culture system that the
putative infectious agent is carcinogenic.
Lung cancer is one of the most common human neoplasms. While a substan-
tial portion of lung cancer can be attributed to tobacco smoking, other factors
may also contribute to development of disease. Moreover, in some cases tobacco
smoking is not involved in causation of the tumor. Human adenocarcinoma of the
lung represents neoplasms of secretory epithelia cells. In the distal airways (al-
veoli and bronchioles), the targets of transformation are Type II pneumocytes and
Clara cells. Bronchiolo alveolar carcinoma (BAC) is a sub-classification of lung
adenocarcinoma in which the tumor cells line the alveoli or bronchioles and spread
in a sideways (lepidic) fashion (Mornex et al., 2003). BAC does not appear to be
tightly associated with tobacco smoking, and the incidence of this cancer may be
rising. Thus the possibility that an infectious agent may be involved in BAC or
human lung adenocarcinoma in general has been suggested by a number of inves-
tigators (Jackson et al., 2000; Koyi et al., 2001; Laurila et al., 1997).
A very interesting animal model for human lung adenocarcinoma exists:
ovine pulmonary adenocarcinoma (OPA), a contagious lung cancer of sheep (Fan,
2003). The disease was first described in the late 1800s in South Africa, and was
named jaagsiekte—“driving sickness” in Afrikaans (York and Querat, 2003). The
disease is prevalent worldwide, and is particularly well-documented in Europe
and Africa. It is estimated that the lifetime risk of developing OPA in high inci-
dence flocks is approximately 25 percent (Sharp and DeMartini, 2003). The
spread of OPA may result from inhalation of aerosols. A noteworthy feature of
the disease is production of excess surfactant by the tumor cells—the normal
function of Type II pneumocytes and Clara cells is to produce lung surfactant and
other molecules important for lung physiology. As a result, animals with end-
stage OPA exhibit respiratory distress; “tipping” of OPA animals results in lung
fluid (excess surfactant) draining from the nose. In the mid-1970s, researchers in
the United Kingdom showed that filtered OPA lung fluid could transfer the dis-
ease to unaffected animals, indicating a viral etiology of OPA (York and Querat,
2003). In the 1960s and 1970s, further evidence was obtained that supported this
notion. In particular, OPA lung fluid was shown to contain reverse transcriptase
activity (characteristic of retroviruses), and OPA lung fluid was shown to contain
antigens that cross-react with two retroviruses—Mason Pfizer monkey virus
(MPMV) and murine mammary tumor virus (MMTV) (Sharp and Herring, 1983).
MPMV and MMTV are relatively closely related viruses, belonging to the
betaretrovirus class. However, the experiments were complicated by the presence
of another retrovirus, an ovine lentivirus, that was also present in many of the
animals with OPA. This confounded experiments to isolate and purify the caus-
ative agent of OPA. Moreover, attempts to propagate the OPA-inducing virus
from lung fluid in tissue culture were unsuccessful. A major advance was made in
1990, when York et al. deduced the presence and sequence of a novel retrovirus
in OPA lung fluid (York et al., 1991, 1992). This was accomplished by first
developing a technique that partially removed the contaminating ovine lentivirus
from the OPA lung fluid (treatment with a fluorocarbon). The treated lung fluid
was then banded to equilibrium in a sucrose density gradient, and RNA was
extracted from the peak of reverse transcriptase activity. This RNA was then
reverse transcribed in vitro using purified reverse transcriptase and an oligodT
primer, and a series of overlapping partial cDNA clones was obtained. Sequenc-
ing of the cDNA clones and overlapping the resulting sequences revealed a novel
complete retroviral sequence; this retrovirus was designated jaagsiekte sheep
retrovirus or JSRV. Consistent with the previous serology, sequence homology
analyses indicated that JSRV is also a beta retrovirus, with sequence similarities
to both MPMV and MMTV. A diagram of the JSRV sequence is shown in Figure
1-12. Disappointingly, attempts to isolate a replication-competent retrovirus from
assembled cDNA clones were unsuccessful.
The availability of the JSRV sequence allowed generation of important mo-
lecular reagents for detection of the putative virus. Initial Southern blot experi-
ments indicated that, as for many other retroviruses, endogenous JSRV-related
proviruses are present in the germ line of all sheep and goats (DeMartini et al.,
2003; Hecht et al., 1996; York et al., 1991). There are 15–20 endogenous JSRV-
related proviruses in most sheep. On an evolutionary scale, these endogenous
viruses entered the sheep germ line relatively recently—1–5 million years ago
(Palmarini et al., 2000a). The existence of endogenous proviruses complicated
experiments, in that it was necessary to distinguish the endogenous proviruses
from the exogenous JSRV present in lung fluid. PCR-based assays were devel-
oped that allowed distinguishing exogenous from endogenous JSRV’s (Palmarini
et al., 1996). This allowed demonstration that tumor samples from OPA animals
consistently contain exogenous JSRV DNA above and beyond the endogenous
JSRV-related sequences. The JSRV cDNA clones were also used for production
JSRV21
pro pol
env
orf-x
gag
DNA
FIGURE 1-13 Foci of transformation induced by the transfection of clone JSRV DNA
into mouse NIH 3T3 cells. Such transfection is a standard assay for detecting viral and
cellular oncogenes. Panels (a) and (c) show untransfected NIH3T3 cells. Panel (b) shows
a focus of transformed cells resulting from transfection with CMV-driven plasmid DNA
(pCMV2JS21). Panel (d) shows a pCMV2JS21 transfected culture that had been passaged
several times prior to plating under focus-forming conditions.
The finding that the JSRV envelope gene contains oncogenic potential is
unusual for replication-competent retroviruses. Most other replication-competent
retroviruses do not normally cause tumors by a direct mechanism (i.e.,
oncogenes). More typically, oncogenesis is a byproduct of the replication cycle
(e.g., insertional activation of cellular proto-oncogenes). However, the fact that
all exogenous JSRVs have a transforming envelope suggests that this property is
important for replication of the virus. We have proposed a hypothesis to explain
this. In studies of the endogenous JSRV-related proviruses, we found that the
endogenous JSRV LTRs do not show transcriptional specificity for lung epithe-
lial cells (Palmarini et al., 2000a). The endogenous viruses provide a view into
the primordial progenitor of JSRV, since they reflect the JSRV progenitor from 1
million to 5 million years ago. (Mutation rates of retroviral DNAs decrease mark-
edly when they are transmitted in the proviral [DNA] form.) Thus the progenitor
to exogenous JSRV likely replicated through different cells in the animals than
lung epithelial cells. Indeed, the endogenous JSRV proviruses in current day sheep
are not expressed in lung epithelial cells, but they are expressed in cells of the
female reproductive tract (Palmarini et al., 2001b). During evolution of exog-
enous JSRV, presumably alterations in the enhancer sequences in the LTR arose
that conferred transcriptional specificity for lung epithelial cells. However, in the
normal adult lung there is relatively little division and growth of Type II
pneumocytes and close cells. Most retroviruses require cell division for efficient
infection and production. Thus during evolution of exogenous JSRV, the muta-
tion in the cytoplasmic tail of the envelope TM protein would allow for more
efficient infection and expression in lung epithelial cells, to which JSRV is tran-
scriptionally restricted.
As mentioned above, JSRV-induced OPA is histologically very similar to
human adenocarcinoma and BAC. In light of the lack of association of human
BAC with tobacco smoking and its increasing incidence, the possibility of a viral
involvement in human lung adenocarcinoma has also been raised. Several inves-
tigators have specifically explored whether a human virus related to JSRV might
be associated with human lung cancer. In particular, De las Heras and colleagues
recently reported a study in which they screened a series of human lung cancers
and other tumors for immunological staining with a polyclonal antibody to JSRV
CA protein (De las Heras et al., 2000). They found that approximately 30 percent
of human BACs and nearly 25 percent of human lung adenocarcinomas showed
immunohistochemical staining with the JSRV CA antibody. In contrast, little or
no reactivity was detected in squamous cell carcinomas of the lung and other
tumors. Thus the reactivity appears to be rather specific for human lung adeno-
carcinomas and BACs. Another laboratory has been able to replicate these immu-
nohistochemistry findings (J. DeMartini, personal communication). On the other
hand, several investigators have attempted to clone a JSRV-related retrovirus
from these human tumors by using PCR amplification with degenerative oligo-
nucleotide primers. So far no one has succeeded.
REFERENCES
De las Heras M, Barsky SH, Hasleton P, Wagner M, Larson E, Egan J, Ortin A, Gimenez-Mas JA,
Palmarini M, Sharp JM. 2000. Evidence for a protein related immunologically to the jaagsiekte
sheep retrovirus in some human lung tumors. The European Respiratory Journal 15:330–332.
DeMartini J, Carlson J, Leroux C, Spencer T, Palmarini M. 2003. Endogenous retroviruses related to
jaagsiekte sheep retrovirus. Pp. 117–137 in Jaagsiekte Sheep Retrovirus and Lung Cancer, H
Fan, ed. Berlin: Springer-Verlag.
Fan H, ed. 2003. Jaagsiekte Sheep Retrovirus and Lung Cancer, Vol. 275. Berlin: Springer-Verlag.
Hecht SJ, Stedman KE, Carlson JO, DeMartini JC. 1996. Distribution of endogenous type B and type
D sheep retrovirus sequences in ungulates and other mammals. Proceedings of the National
Academy of Sciences 93:3297–3302.
Holland MJ, Palmarini M, Garcia-Goti M, Gonzalez L, de las Heras M, McKendrick I, Sharp JM.
1999. Jaagsiekte retrovirus is widely distributed both in T and B lymphocytes and in mono-
nuclear phagocytes of sheep with naturally and experimentally acquired pulmonary adenomato-
sis. Journal of Virology 73:4004–4008.
Jackson LA, Wang SP, Nazar-Stewart V, Grayston JT, Vaughan TL. 2000. Association of Chlamydia
pneumoniae immunoglobulin A seropositivity and risk of lung cancer. Cancer Epidemiology
Biomarkers and Prevention 9:1263–1266.
Koyi H, Branden E, Gnarpe J, Gnarpe H, Steen B. 2001. An association between chronic infection
with Chlamydia pneumoniae and lung cancer. A prospective 2-year study. APMIS 109:572–
580.
Laurila AL, Anttila T, Laara E, Bloigu A, Virtamo J, Albanes D, Leinonen M, Saikku P. 1997.
Serological evidence of an association between Chlamydia pneumoniae infection and lung can-
cer. International Journal of Cancer 74:31–34.
Maeda N, Palmarini M, Murgia C, Fan H. 2001. Direct transformation of rodent fibroblasts by
jaasiekte sheep retrovirus DNA. Proceedings of the National Academy of Sciences 98:4449–
4454.
Mornex JF, Thivolet F, de las Heras M, Leroux C. 2003. Pathology of human bronchioloalveolar
carcinoma and its relationship to the ovine disease. Pp. 225–248. In Fan H, editor. Jaagsiekte
Sheep Retrovirus and Lung Cancer. Berlin: Springer-Verlag.
Palmarini M, Cousens C, Dalziel RG, Bai J, Stedman K, DeMartini JC, Sharp JM. 1996. The exog-
enous form of Jaagsiekte retrovirus is specifically associated with a contagious lung cancer of
sheep. Journal of Virology 70:1618–1623.
Palmarini M, Sharp JM, de las Heras M, Fan H. 1999. Jaagsiekte sheep retrovirus is necessary and
sufficient to induce a contagious lung cancer in sheep. Journal of Virology 73:6964–6972.
Palmarini M, Hallwirth C, York D, Murgia C, de Oliveira T, Spencer T, Fan H. 2000a. Molecular
cloning and functional analysis of three type D endogenous retroviruses of sheep reveal a differ-
ent cell tropism from that of the highly related exogenous jaagsiekte sheep retrovirus. Journal of
Virology 74:8065–8076.
Palmarini M, Datta S, Omid R, Murgia C, Fan H. 2000b. The long terminal repeat of Jaagsiekte sheep
retrovirus is preferentially active in differentiated epithelial cells of the lungs. Journal of Virol-
ogy 74:5776–5787.
Palmarini M, Maeda N, Murgia C, De-Fraja C, Hofacre A, Fan H. 2001a. A phosphatidylinositol 3-
kinase docking site in the cytoplasmic tail of the jaagsiekte sheep retrovirus transmembrane
protein is essential for envelope-induced transformation of NIH 3T3 cells. Journal of Virology
75:11002–11009.
Palmarini M, Gray CA, Carpenter K, Fan H, Bazer FW, Spencer TE. 2001b. Expression of endog-
enous betaretroviruses in the ovine uterus: effects of neonatal age, estrous cycle, pregnancy and
progersterone. Journal of Virology 75:11319–11327.
Sharp J and DeMartini J. 2003. Natural history of JSRV in sheep. Pp. 55–79. In Fan H, editor.
Jaagsiekte Sheep Retrovirus and Lung Cancer. Berlin: Springer-Verlag.
Sharp JM and Herring AJ. 1983. Sheep pulmonary adenomatosis: demonstration of a protein which
cross-reacts with the major core proteins of Mason-Pfizer monkey virus and mouse mammary
tumour virus. The Journal of General Virology 64:2323–2327.
Willems L, Kettmann R, Dequiedt F, Portetelle D, Voneche V, Cornil I, Kerkhofs P, Burny A,
Mammerickx M. 1993. In vivo infection of sheep by bovine leukemia virus mutants. Journal of
Virology 67:4078–4085.
York D and Querat G. 2003. A history of ovine pulmonary adenocarcinoma (Jaagsiekte) and experi-
ments leading to the deduction of the JSRV nucleotide sequence. Pp. 1–23. In Fan H, editor.
Jaagsiekte Sheep Retrovirus and Lung Cancer. Berlin: Springer-Verlag.
York DF, Vigne R, Verwoerd DW, Querat G. 1991. Isolation, identification, and partial cDNA clon-
ing of genomic RNA of jaagsiekte retrovirus, the etiological agent of sheep pulmonary ad-
enomatosis. Journal of Virology 65:5061–5067.
York DF, Vigne R, Verwoerd DW, Querat G. 1992. Nucleotide sequence of the jaagsiekte retrovirus,
an exogenous and endogenous type D and B retrovirus of sheep and goats. Journal of Virology
66:4930–4939.
mated that nearly 20 percent of all visits to dermatologists are related to the treat-
ment of acne. Acne often debuts during changes in hormonal levels in pre-teens;
however, it is also very common as an adult-onset condition, often associated
with hormonal fluctuation during the menstrual cycle and pregnancy. While not
life-threatening, acne can persist for years and is known to have serious psycho-
social effects such as decreased self-esteem, depression, frustration, and social
withdrawal. In addition to dermatological pathology, P. acnes is also suspected to
be discreetly involved in post-operative infections, prostheses failure, and more
recently, in inflammation of lumbar nerve roots leading to sciatica.
P. acnes, previously known by the name Corynebacterium parvum, has been
studied extensively by immunologists for its ability to stimulate the reticuloen-
dothelial system (Adlam and Scott, 1973). Not too long ago, an important
cytokine, interleukin (IL)-18 was cloned from the liver of mice primed with P.
acnes followed by challenge with LPS (Okamura et al., 1995). In the early eight-
ies, certain bacteria, including BCG and P. acnes, were commonly used to stimu-
late the innate immune response against cancer in mice and human cells (Cantrell
and Wheat, 1979; Davies, 1982). One of the great ironies of this organism is that
it is a powerful nonspecific immune stimulant that resides naturally in the skin;
its role as an immunostimulant in humans is appreciated when cases of severe
acne also develop adjuvant-type arthritis.
Some investigators have gone so far as to suggest that severe acne, by virtue
of the nonspecific immunostimulatory effects of P. acnes, might have played a
role in natural protection against life-threatening diseases such as malaria and
plague. In contrast, the acquired immune response to P. acnes has received little
attention in humans.
Pathogenesis of Acne
Chronic inflammatory acne cannot be defined as an infectious disease, since
the bacteria are normally present on the skin of a vast majority of individuals,
irrespective of the presence of acne lesions. P. acnes apparently only triggers the
disease when it meets favorable dermatophysiological terrain; P. acnes coloniza-
tion of the skin is therefore necessary but not sufficient for the establishment of
the pathology. The 4 major recognized pathophysiological features of acne in-
clude androgen stimulated seborrhea, hyperkeratinization and obstruction of the
follicular epithelium, proliferation of P. acnes, and then inflammation.
Comedogenesis, the transformation of the pilosebaceous follicle into the pri-
mary acne lesion, the comedone, is the product of abnormal follicular keratiniza-
tion related to excessive sebum secretion. During this process, P. acnes often gets
trapped in layers of corneocytes and sebum and rapidly colonizes the comedonal
kernel, resulting in a microcomedone, a structure invisible to the naked eye
(Plewig and Kligman, 2000). A microcomedone can develop into larger struc-
tures, called comedones. Comedones can be a closed structure (whitehead) that
appears like a colored bump on the skin or an open structure (blackhead). Unlike
open comedones, closed comedones cannot evacuate the thread-looking conglom-
erate of cell debris, sebum, P. acnes and its products to the skin surface, and this
makes them more prone to inflammation and rupture. In inflammatory acne,
comedones rupture and the follicular material becomes dispersed in the dermis
rather than on the skin surface. Depending on the extent of the damage to the
comedone wall, various types of inflammatory lesions are produced and these are
classified as papules, pustules, or nodules. Nodules are the most severe types of
acne lesions and scarring may be associated with any form of severe inflamma-
tory acne.
A break in the lining of the comedone was initially attributed to free fatty
acids generated by P. acnes-mediated triglyceride hydrolysis, but for several rea-
sons, it is now thought that substances produced by P. acnes are directly involved
in the rupture the comedone epithelial lining (Holland et al., 1981). The bacteria
secrete many polypeptides, among which are numerous extracellular enzymes
such as proteases, hyaluronidases, neuraminidases, and others that could be in-
volved in epithelium permeabilization and inflammatory infiltration (Noble,
1984). P. acnes is also known to produce chemotactic factors (Puhvel and
Sakamoto, 1977), proinflammatory cytokine inducing-factors (Vowels et al.,
1995), and to activate both the direct and indirect complement pathways (Webster
et al., 1978). The infiltrate of an early inflamed lesion consists of polymorpho-
nuclear cells that certainly contribute to the lining breakage, but eventually, as
time goes by and infection becomes chronic, these cells attract and are replaced
by mononuclear cells, predominantly T-cells of the CD4 phenotype (Norris and
Cunliffe, 1988; Layton et al., 1994). As the inflammation propagates to the lining
of adjacent sebaceous follicules, it can start a chain reaction that results in mul-
tiple lesions connected together and called a sinus. Studies by Hoffler et al. (1985)
have revealed differences in the production of various enzymes by Propionibac-
terium isolates of acne lesions versus bacteria isolated from healthy controls.
These studies are important for differentiating bacterial antigens that lead healthy
controls to generate a protective immune response and those that might be in-
volved in pathogenesis.
Antibody against P. acnes antigenic determinants are found in the blood of
most adults, whether they have had acne or not (Ingham et al., 1987); amounts
may vary between the two populations, and possibly the nature of the determi-
nants the antibodies recognize (Holland et al., 1993). Recent investigations by
our group suggest that differential recognition might involve surface molecules
with physiological functions. P. acnes specific IgG and IgA are also found at the
level of the follicular infudibulum (Knop et al., 1983); these antibodies might be
of great importance in limiting or preventing P. acnes proliferation, and maybe
more importantly, in preventing comedonal lining destruction by P. acnes-de-
rived soluble factors. Our preliminary data suggests that a robust P. acnes spe-
cific T-cell response is also common in adult donors, but its specificity at the
antigen level is currently under investigation. We like to think that there possibly
exists a P. acnes-specific protective immunity against acne. This hypothesis is
supported by the fact that some people never get acne, as well as by the observa-
tion that acne is mostly a disease of young people, (although there are numerous
exceptions), and that even in countries where people are unable to afford sophis-
ticated medications, chronic disease of adolescents eventually resolves with age.
Finally, there have been successful human trials of therapeutic vaccination against
P. acnes, and although the rate of success has not been high, some individuals
refractory to conventional approaches experienced remission (Goldman et al.,
1979; Vymola et al., 1970).
REFERENCES
Adamou JE, Heinrichs JH, Erwin AL, Walsh W, Gayle T, Dormitzer M, Dagan R, Brewah YA,
Barren P, Lathigra R, Langermann S, Koenig S, Johnson S. 2001. Identification and character-
ization of a novel family of pneumococcal proteins that are protective against sepsis. Infection
and Immunity 69:949–958.
Adlam C and Scott MT. 1973. Lympho-reticular stimulatory properties of Corynebacterium parvum
and related bacteria. Journal of Medical Microbiology 6:261–274.
Cantrell JL and Wheat RW. 1979. Antitumor activity and lymphoreticular stimulation properties of
fractions isolated from Corynebacterium parvum. Cancer Research 39:3554–3563.
Chakravarti DN, Fiske MJ, Fletcher LD, Zagursky RJ. 2000. Application of genomics and proteomics
for identification of bacterial gene products as potential vaccine candidates. Vaccine 19:601–
612.
Chu RM, Tummala RP, Hall WA. 2001. Focal intracranial infections due to Propionibacterium acnes:
report of three cases. Neurosurgery 49:717–720.
Clark WL, Kaiser PK, Flynn HW Jr, Belfort A, Miller D, Meisler DM. 1999. Treatment strategies and
visual acuity outcomes in chronic postoperative Propionibacterium acnes endophthalmitis. Oph-
thalmology 106:1665–1670.
Davies M. 1982. Bacterial cells as anti-tumour agents in man. Reviews on Environmental Health
4:31–56.
Goldman L, Michael JG, Riebel S. 1979. The immunobiology of acne. A polyvalent proprionibacteria
vaccine. Cutis 23:181–184.
Herrmann M, Vaudaux PE, Pittet D, Auckenthaler R, Lew PD, Schumacher-Perdreau F, Peters G,
Waldvogel FA. 1988. Fibronectin, fibrinogen, and laminin act as mediators of adherence of
clinical staphylococcal isolates to foreign material. The Journal of Infectious Diseases 158:693–
701.
Hoffler U, Gehse M, Gloor M, Pulverer G. 1985. Enzyme production of propionibacteria from pa-
tients with acne vulgaris and healthy persons. Acta Dermato-Venereologica 65:428–432.
Holland KT, Ingham E, Cunliffe WJ. 1981. A review, the microbiology of acne. The Journal of
Applied Bacteriology 51:195–215.
Holland KT, Holland DB, Cunliffe WJ, Cutcliffe AG. 1993. Detection of Propionibacterium acnes
polypeptides which have stimulated an immune response in acne patients but not in normal
individuals. Experimental Dermatology 2:12–16.
Ingham E, Gowland G, Ward RM, Holland KT, Cunliffe WJ. 1987. Antibodies to P. acnes and P.
acnes exocellular enzymes in the normal population at various ages and in patients with acne
vulgaris. The British Journal of Dermatology 116:805–812.
Ishige I, Usui Y, Takemura T, Eishi Y. 1999. Quantitative PCR of mycobacterial and propionibacterial
DNA in lymph nodes of Japanese patients with sarcoidosis. Lancet 354:120–123.
Kirschbaum JO and Kligman AM. 1963. The pathogenic role of Corynebacterium acnes in acne
vulgaris. Archives of Dermatology 88:832–833.
Knop J, Ollefs K, Frosch PJ. 1983. Anti-P. acnes antibody in comedonal extracts. The Journal of
Investigative Dermatology 80:9–12.
Layton AM, Henderson CA, Cunliffe WJ. 1994. A clinical evaluation of acne scarring and its inci-
dence. Clinical and Experimental Dermatology 19:303–308.
Lazar JM and Schulman DS. 1992. Propionibacterium acnes prosthetic valve endocarditis: a case of
severe aortic insufficiency. Clinical Cardiology 15:299–300.
Mohsen AH, Price A, Ridgway E, West JN, Green S, McKendrick MW. 2001. Propionibacterium
acnes endocarditis in a native valve complicated by intraventricular abscess: a case report and
review. Scandinavian Journal of Infectious Diseases 33:379–380.
Noble WC. 1984. Skin microbiology: coming of age. Journal of Medical Microbiology 17:1–12
Norris JF and Cunliffe WJ. 1988. A histological and immunocytochemical study of early acne le-
sions. The British Journal of Dermatology 118:651–659.
Okamura H, Nagata K, Komatsu T, Tanimoto T, Nukata Y, Tanabe F, Akita K, Torigoe K, Okura T,
Fukuda S. 1995. A novel costimulatory factor for gamma interferon induction found in the livers
of mice causes endotoxic shock. Infection and Immunity 63:3966–3972.
Plewig G and Kligman AM, eds. 2000. Acne and Rosacea, 3rd ed., 744 pages. New York: Springer-
Verlag.
Puhvel SM and Sakamoto M. 1977. Chemoattractant properties of Corynebacterium parvum and
pyran copolymer for human monocytes and neutrophils. Journal of the National Cancer Insti-
tute 58:781–783.
Stirling A, Worthington T, Rafiq M, Lambert PA, Elliott TS. 2001. Association between sciatica and
Propionibacterium acnes. Lancet 357:2024–2025.
Thompson TP and Albright AL. 1998. Propionibacterium [correction of Proprionibacterium] acnes
infections of cerebrospinal fluid shunts. Childs Nervous System 14:378–380.
Tunney MM, Patrick S, Curran MD, Ramage G, Hanna D, Nixon JR, Gorman SP, Davis RI, Ander-
son N. 1999. Detection of prosthetic hip infection at revision arthroplasty by immunofluores-
cence microscopy and PCR amplification of the bacterial 16S rRNA gene. Journal of Clinical
Microbiology 37:3281–3290.
Underdahl JP, Florakis GJ, Braunstein RE, Johnson DA, Cheung P, Briggs J, Meisler DM. 2000.
Propionibacterium acnes as a cause of visually significant corneal ulcers. Cornea 19:451–454.
Vymola F, Buda J, Lochmann O, Pillich J. 1970. Successful treatment of acne by immunotherapy.
Journal of Hygiene, Epidemiology, Microbiology, and Immunology 14:135–138.
Vowels BR, Yang S, Leyden JJ. 1995. Induction of proinflammatory cytokines by a soluble factor of
Propionibacterium acnes: implications for chronic inflammatory acne. Infection and Immunity
63:3158–3165.
Webster GF, Leyden JJ, Norman ME, Nilsson UR. 1978. Complement activation in acne vulgaris: in
vitro studies with Propionibacterium acnes and Propionibacterium granulosum. Infection and
Immunity 22:523–529.
Wizemann TM, Heinrichs JH, Adamou JE, Erwin AL, Kunsch C, Choi GH, Barash SC, Rosen CA,
Masure HR, Tuomanen E, Gayle A, Brewah YA, Walsh W, Barren P, Lathigra R, Hanson M,
Langermann S, Johnson S, Koenig S. 2001. Use of a whole genome approach to identify vaccine
molecules affording protection against Streptococcus pneumoniae infection. Infection and Im-
munity 69:1593–1598.
Yu JL, Mansson R, Flock JI, Ljungh A. 1997. Fibronectin binding by Propionibacterium acnes. FEMS
Immunology and Medical Microbiology 19:247–253.
OVERVIEW
Successful disease control efforts in some economically developing coun-
tries have increased life expectancy and resulted in changes in demographics from
predominantly youthful populations to older and aging ones. Consequently, dur-
ing the next 20 years, chronic diseases are expected to become increasingly im-
portant in economically developing regions and to encompass chronic conditions
currently attributed to industrialized nations. Not only will changing economics,
demographic shifts with lower childhood mortality, and changing lifestyles affect
this trend, but migration from rural to urban areas and into previously uninhabited
ecosystems may expose populations to new infectious agents that underlie chronic
disease. Both newly identified and well-recognized infectious etiologies of
chronic disease, including infections known to enter a chronic state, such as tu-
berculosis and malaria, will acquire increasing importance to domestic and global
health. As such, countries with limited research capacities and health care ser-
vices will face increasing burdens from both infectious and chronic disease.
Richard Guerrant illustrated the wide-ranging nature of the threats from
chronic diseases caused by infections, using as an example the long-term conse-
quences of early childhood enteric and parasitic infections. The chronic impact of
repeated malnourishing diarrheal illnesses is greater than that of acute deaths
from enteric illness, which claims more than 6,000 children each day. Early diar-
rheal illnesses have significant long-term effects not only on physical fitness, but
on growth, cognition, and school performance. Diarrhea appears to be a cofactor
with malnutrition in that it reduces nutritional absorption.
81
Josemir Sander detailed the relationship between epilepsy, the most common
serious neurological condition worldwide, and a number of parasites. Epilepsy is
a symptom complex, so diagnosis relies on clinical history rather than a specific
test. Incidence is higher in developing countries than in the industrialized world,
and appears to be higher in rural areas than in urban areas. Furthermore, endemic
infections may be responsible for the increased incidence in low-income coun-
tries.
Maureen Durkin discussed ostensibly preventable or controllable infections
that are important causes of childhood cognitive disability, paralysis, epilepsy,
blindness, and deafness in developing countries. These infections include con-
genital disorders, such as syphilis, rubella, and cytomegalovirus, as well as infec-
tions occurring during infancy and childhood, such as malaria, meningitis, Japa-
nese viral encephalitis, measles, poliomyelitis, and trachoma.
Eduardo Gotuzzo described clinical experience with HTLV-1, a retrovirus
that causes adult T-cell leukemia and is endemic in much of Latin America.The
virus produces 3 different clinical patterns: cancer, autoimmune disease, and im-
munosuppression disease. In developing countries, 80 percent of lymphomas are
non-Hodgkins lymphoma, and 10 pecent of the non-Hodgkins lymphomas seen
by the Peruvian national cancer center are associated with HTLV-1. A second
clinical presentation is tropical dysplastic paraparesia (TSP). The third clinical
pattern associated with the infection is immunosuppression.
Sanaa Kamal described chronic hepatitis C infection with and without schis-
tosomiasis. Patients typically present in their thirties or forties with gastro-
intestinal bleeding, usually massive, and compromised liver function and status.
These patients progress rapidly to end stage disease, usually dying in their forties.
Coinfected individuals have significantly higher fibrosis levels and are unable to
achieve spontaneous viral clearance.
Altaf Lal described interactions between the human immunodeficiency virus
(HIV) and malaria to illustrate how different pathogens interact with each other
and how they modulate the disease process. Infant mortality is higher in babies
born to mothers who are infected with placental malaria and HIV-1, and these
infants have lower levels of acquired passive immunity. Concurrent infections
also promote pathogen diversity. The interactions, however, are extremely com-
plex. For example, acute measles suppresses HIV replication significantly.
*Parts of this paper have been published in a perspective article (Guerrant et al, 2002a) and in a
review (Guerrant et al., 2002b).
advantages that they can also help assess effectiveness of interventions as well as
the burden of disease and are standardized to permit age weighting and compara-
bility across studies.
All conditions affecting health as well as interventions that prevent or re-
verse the adverse effects of these conditions are measured in economic as well as
human terms. These include, in addition to the causes of death and the YPLL due
to premature mortality, the morbidity costs or YLD from conditions that impair
the ability of individuals to reach their full human and economic potential or
productivity. As causes of premature mortality are brought under control world-
wide, the morbidity costs are becoming increasingly recognized and their
quantitation is increasingly important. Thus, in addition to diseases or conditions
like meningitis, AIDS, or automobile accidents that are often fatal at young ages
and are thus responsible for disproportionately greater years of life lost, we must
also weigh the burden of chronic diseases, like arthritis or depression, that often
disable much more than they kill. Both YPLL and YLD are included in the
DALYs that are being used to assess the burdens of all diseases or conditions that
threaten healthy life worldwide, as well as the “cost-effectiveness” of interven-
tions designed for their amelioration. Both mortality (YPLL) and morbidity
(YLD) pose profound economic costs, whether a young, productive working par-
ent dies with AIDS or violence, or whether a child with repeated bouts of diar-
rhea, parasitic infection, or malnutrition fails to develop normally to meet his or
her full human and economic potential.
It is just such an analysis that has brought appropriate attention to conditions
like neuropsychiatric diseases or depression that kill few but disable many. Like-
wise, from placebo-controlled prospective studies of albendazole treatment of
helminthic infections in Kenyan and Jamaican schoolchildren, intestinal hel-
minths have been found to impair growth, fitness, and even cognitive function
(Adams et al., 1994; Nokes et al., 1992a,b; Nokes and Bundy, 1992; Stephenson
et al., 1993). Such studies have enabled Chan and Bundy to suggest potential
recalculation of the long-term impact of childhood helminthic infections on
DALYs to essentially double their previous values (Chan et al., 1994; Guerrant
and Blackwood, 1999).
Indeed, the disability component of the DALY calculations for malnutrition
and the “tropical cluster” (trypanosomiasis, Chagas’ disease, schistosomiasis, and
leishmaniasis), like neuropsychiatric conditions, chronic obstructive lung disease,
and rheumatoid arthritis, outweigh their mortality components (Guerrant and
Blackwood, 1999; Murray and Lopez, 1997). However, the initially calculated
DALY for diarrheal diseases, from a 1997 assessment (Murray and Lopez, 1997),
initially comprised 95 percent mortality (YPLL) and only 5 percent disability
(YLD, from the transient 10 percent incapacitation during just the overt diarrheal
illness [i.e., liquid stools] itself). No long-term disability from repeated dehydrat-
ing and malnourishing diarrheal illnesses in the critical formative developmental
first 2 years of life is considered, largely because there had been no data to sug-
gest such long-term effects (Guerrant and Blackwood, 1999).
TABLE 2-1 Evidence for Lasting Disability Effects from Early Childhood
Diarrhea
Disease Outcome References
Growth shortfalls
Cryptosporidial infections Increased diarrhea morbidity and Agnew et al., 1998
and persistent diarrhea nutritional shortfalls for up to Lima et al., 2000
18 months Newman et al., 1999
Cognitive impairment Impaired cognitive function at 6–9 y.o. Guerrant et al., 1999
Early childhood diarrhea by McCarthy Draw-A-Design
(0–2 y.o.) (p = 0.017 when controlling for early
childhood helminthic infections), and
WISC coding and reverse digit
span testing (p = 0.045)
Early childhood diarrhea Impaired Test of Nonverbal Intelligence Niehaus et al., 2002
(0–2 y.o.) (TONI-III) scores at 6–10 y.o., when
controlling for maternal education,
breast feeding duration, and early
helminthic infections; and WISC
coding and digit span scores were lower
in children who had one or more
persistent diarrheal illnesses in their
first 2 years of life.
been under such intensive surveillance (Lima, Guerrant et al., unpublished obser-
vations).
We are now finding that these correlations of early childhood diarrhea are
also extending to school performance, with significant associations of diarrhea in
the first 2 years of life with delayed age at starting school and age for grade that
remain even after controlling for maternal education and (also affected) stature.
Late starters are also two-fold more likely to have experienced cryptosporidial
infections (Lorntz et al., 2000).
A recent report describes the significant associations of stunting in the first 2
years of life and multiple episodes of Giardia infection with impaired intelli-
gence quotients on the WISC-R test among children in Peru (Berkman et al.,
2002). This is the setting in which diarrhea is also associated with reduced WISC-
R scores albeit not independently of its association with stunting. This is also the
1The statistical symbol p stands for the probability that the observed difference could have been
obtained by chance alone, given random variation and a single test of the null hypothesis.
hood diarrheal illnesses of poverty is potentially far greater and more critical a
global investment than is generally appreciated, i.e., a global “tax” that is paid for
the impaired work productivity in the global economy because these largely pre-
ventable illnesses continue unabated. Thus, beyond their obvious human toll, the
diseases of poverty may well require an economic investment (as they are readily
prevented) that we cannot afford not to make.
• Scenario 5 assumes that only 5 percent of 0–4 year olds (or half experi-
ence a 1 percent life-long disability).
Conclusions
Critical to understanding and making this case for investing adequate re-
sources in the presentation or amelioration of the diseases of poverty like diarrhea
is obtaining solid information about the potential long-term correlates with ill-
ness rates and even subclinical infections, controlling to the extent possible the
numerous confounding variables, and careful studies of potential interventions
that could alter these adverse outcomes. Only improved data and careful, accurate
analyses will direct adequate attention to alleviation of these diseases of poverty
that are so potentially costly to human and societal development for us all.
REFERENCES
Adams EJ, Stephenson LS, Latham MC, Kinoti SN. 1994. Physical activity and growth of Kenyan
school children with hookworm, Trichuris trichiura and Ascaris lumbricoides infections are
improved after treatment with albendazole. Journal of Nutrition 124:1199–1206.
Agnew DG, Lima AA, Newman RD, Wuhib T, Moore RD, Guerrant RL, Sears CL. 1998.
Cryptosporidiosis in northeastern Brazilian children: association with increased diarrhea mor-
bidity. Journal of Infectious Diseases 177:754–760.
Basta SS, Soerkirman, D Karyadi, NS Scrimshaw. 1979. Iron deficiency anaemia and the productivity
of males in Indonesia. The American Journal of Clinical Nutrition 32: 916–925.
Berkman DS, Lescano AG, Gilman RH, Lopez SL, Black MM. 2002. Effects of stunting, diarrhoeal
disease, and parasitic infection during infancy on cognition in late childhood: a follow-up study.
Lancet 359:564–571.
Bern C, Martines J, de Zoysa I, Glass RI. 1992. The magnitude of the global problem of diarrhoeal
disease: a ten-year update. Bulletin of the World Health Organization 70:705–714.
Brantley RK, Williams KR, Silva TM, Sistrom M, Thielman NM, Ward H, Lima AA, Guerrant RL.
2003. AIDS-associated diarrhea and wasting in Northeast Brazil is associated with
subtherapeutic plasma levels of antiretroviral medications and with both bovine and human
subtypes of Cryptosporidium parvum. Brazilian Journal of Infectious Diseases 7:16–22.
Chan MS, Medley GF, Jamison D, Bundy DA. 1994. The evaluation of potential global morbidity
attributable to intestinal nematode infections. Parasitology 109:373–387.
Checkley W, Gilman RH, Epstein LD, Suarez M, Diaz JF, Cabrera L, Black RE, Sterling CR. 1997.
Asymptomatic and symptomatic cryptosporidiosis: their acute effect on weight gain in Peruvian
children. American Journal of Epidemiology 145:156–163.
Checkley W, Epstein LD, Gilman RH, Black RE, Cabrera L, Sterling CR. 1998. Effects of
Cryptosporidium parvum infection in Peruvian children: growth faltering and subsequent catch-
up growth. American Journal of Epidemiology 148:497–506.
Dobbing J. 1985. Infant nutrition and later achievement. The American Journal of Clinical Nutrition
41:477–484.
Dobbing J. 1990. Boyd Orr memorial lecture. Early nutrition and later achievement. Proceedings of
the Nutrition Society 49:103–118.
Dobbing J and Sands J. 1985. Cell size and cell number in tissue growth and development. An old
hypothesis reconsidered. Archives Francaises de Pediatrie 42:199–203.
Golden MH. 1994. Is complete catch-up possible for stunted malnourished children? European Jour-
nal of Clinical Nutrition 48:S58–S70.
Guerrant DI, Moore SR, Lima AA, Patrick PD, Schorling JB, Guerrant RL. 1999. Association of early
childhood diarrhea and cryptosporidiosis with impaired physical fitness and cognitive function
four-seven years later in a poor urban community in northeast Brazil. The American Journal of
Tropical Medicine and Hygiene 61:707–713.
Guerrant RL. 2001. The unacceptable costs of the diseases of poverty. Current Infectious Disease
Reports 3:1–3.
Guerrant RL and Blackwood BL. 1999. Threats to global health and survival: the growing crises of
tropical infectious diseases—our “unfinished agenda.” Clinical Infectious Diseases 28:966–986.
Guerrant RL, Kosek M, Lima AA, Lorntz B, Guyatt HL. 2002a. Updating the DALYs for diarrhoeal
disease. Trends in Parasitology 18:191–193.
Guerrant RL, Kosek M, Moore S, Lorntz B, Brantley R, Lima AA. 2002b. Magnitude and impact of
diarrheal diseases. Archives of Medical Research 33:351–355.
Kosek M, Bern C, Guerrant RL. 2003. The global burden of diarrhoeal disease, as estimated from
studies published between 1992 and 2000. Bulletin of the World Health Organization 81:197–
204.
Lima AA, Silva TM, Gifoni AM, Barrett LJ, McAuliffe IT, Bao Y, Fox JW, Fedorko DP, Guerrant
RL. 1997. Mucosal injury and disruption of intestinal barrier function in HIV-infected individu-
als with and without diarrhea and cryptosporidiosis in northeast Brazil. American Journal of
Gastroenterology 92:1861–1866.
Lima AA, Moore SR, Barboza MS, Soares AM, Schleupner MA, Newman RD, Sears CL, Nataro JP,
Fedorko DP, Wuhib T, Schorling JB, Guerrant RL. 2000. Persistent diarrhea signals a critical
period of increased diarrhea burdens and nutritional shortfalls: A prospective cohort study among
children in northeastern Brazil. Journal of Infectious Diseases 181:1643–1651.
Lorntz et al. 2000. Presentation at the ASTMH.
Moore SR, Lima AA, Schorling JB, Barboza MS Jr., Soares AM, Guerrant RL. 2000. Changes over
time in the epidemiology of diarrhea and malnutrition among children in an urban Brazilian
shantytown, 1989 to 1996. International Journal of Infectious Diseases 4:179–186.
Moore SR, Lima AA, Conaway MR, Schorling JB, Soares AM, Guerrant RL. 2001. Early childhood
diarrhoea and helminthiases associate with long-term linear growth faltering. International Jour-
nal of Epidemiology 30:1457–1464.
Murray CJ and Lopez AD, eds. 1997. The Global Burden of Disease: A Comprehensive Assessment
of Mortality and Disability from Diseases, Injuries, and Risk Factors in 1900 and Projected to
2020. Cambridge, MA: Harvard University Press.
Murray CJ, Lopez AD, Jamison DT. 1994. The global burden of disease in 1990: summary results,
sensitivity analysis and future directions. Bulletin of the World Health Organization 72:495–
509.
Ndamba J, Makaza N, Munjoma M, Gomo E, Kaondera KC. 1993. The physical fitness and work
performance of agricultural workers infected with Schistosoma mansoni in Zimbabwe. Annals
of Tropical Medicine & Parasitology 87:553–561.
Newman RD, Sears CL, Moore SR, Nataro JP, Wuhib T, Agnew DA, Guerrant RL, Lima AA. 1999.
Longitudinal study of Cryptosporidium infection in children in northeastern Brazil. Journal of
Infectious Diseases 180:167–175.
Niehaus MD, Moore SR, Patrick PD, Derr LL, Lorntz B, Lima AA, Guerrant RL. 2002. Early child-
hood diarrhea is associated with diminished cognitive function 4 to 7 years later in children in a
northeast Brazilian shantytown. The American Journal of Tropical Medicine and Hygiene
66:590–593.
Nokes C and Bundy DA. 1992. Trichuris trichiura infection and mental development in children.
Lancet 339:500.
Nokes C, Grantham-McGregor SM, Sawyer AW, Cooper ES, Bundy DA. 1992a. Parasitic helminth
infection and cognitive function in school children. Proceedings of the Royal Society of London.
Series B Biological Sciences 247:77–81.
Nokes C, Grantham-McGregor SM, Sawyer AW, Cooper ES, Robinson BA, Bundy DA. 1992b.
Moderate to heavy infections of Trichuris trichiura affect cognitive function in Jamaican school
children. Parasitology 104:539–547.
Snyder JD and Merson MH. 1982. The magnitude of the global problem of acute diarrhoeal disease:
a review of active surveillance data. Bulletin of the World Health Organization 60:605–613.
Soewondo S, Husaini M, Pollitt E. 1989. Effects of iron deficiency on attention and learning pro-
cesses in preschool children: Bandung, Indonesia. The American Journal of Clinical Nutrition
50:667–673.
Steiner TS, Lima AA, Nataro JP, Guerrant RL. 1998. Enteroaggregative Escherichia coli produce
intestinal inflammation and growth impairment and cause interleukin-8 release from intestinal
epithelial cells. Journal of Infectious Diseases 177:88–96.
Stephenson LS, Latham MC, Adams EJ, Kinoti SN, Pertet A. 1993. Physical fitness, growth and
appetite of Kenyan school boys with hookworm, Trichuris trichiura and Ascaris lumbricoides
infections are improved four months after a single dose of albendazole. Journal of Nutrition
123:1036–1046.
ing a role (Johnson and Sander, 2001). It might be difficult, however, to say
whether individuals share predisposition or are exposed to the same environmen-
tal sources. In epilepsy due to infections, it could also be argued that the inter-
action between infective agents and social, genetic, and environmental factors
determines the extent of the risk (Bittencourt et al., 1999).
Endemic infections such as malaria, neurocysticercosis and paragonomiasis
are associated with epilepsy in certain environments particularly in low-income
countries (Sander, 2003). Neurocysticercosis, for instance, is the commonest
cause of newly diagnosed epilepsy in large areas of the tropical belt, and malaria
is the commonest cause of fever in febrile convulsions in endemic areas (Medina
et al., 1990; Waruiru et al., 1996; Carpio, 2002). These infections are probably
responsible for the higher incidence of epilepsy in low-income economies and
this makes epilepsy one of the world’s most preventable non-communicable con-
ditions (Commission on Tropical Diseases of the International League Against
Epilepsy, 1994; Bittencourt et al., 1999; Bergen and Silberberg, 2002). This pa-
per briefly reviews central nervous infections and infestations that may lead to
chronic epilepsy. The contribution of social and geographic factors and the
putative pathophysiology are discussed in general terms and the natural history of
the commonest infections is reviewed. Seizures that occur during the acute phase
of an infection are termed acute symptomatic seizures and do not constitute epi-
lepsy even if repeated, and are not covered here.
Pathophysiology
Seizures in the aftermath of CNS infectious diseases are usually partial or
focal in nature, i.e., they start in the epileptic focus, a localised area of (usually
damaged) cortex (Bittencourt et al., 1999). The route of entry of infective agents
to the CNS may be arterial—(through the blood-brain barrier or the choroid
plexus), by passive venous transport through the spinal plexuses, by direct inva-
sion through trauma or from cranial sinuses. Viruses may enter the CNS by the
haematogenous route or via neuronal routes (Eeg-Olofsson, 2003). The infec-
tious agent needs to reach and damage the cerebral cortex for seizures to develop,
and this may be achieved through various mechanisms (Bittencourt et al., 1999).
Fungal infections are often dependent on the immunological status of the person,
and are therefore more prevalent in immunocompromised subjects. Cortical dam-
age will not invariably lead to epilepsy but is a major risk factor affected by the
location, severity and individual predisposition, which is likely to be genetically
determined (Sander and Shorvon, 1996). There may be months, or even years,
between the insult and the onset of epilepsy and the reasons for this are not well
understood. The existence of critical modulators, which can turn damaged corti-
cal tissue into an epileptic focus, has been postulated (Walker et al., 2002).
Arteritis, ischaemia and infarction are the main pathological outcome of se-
vere viral or bacterial CNS disease and if this affects the cortical ribbon it may be
the substrate for an epileptic focus (Bittencourt et al., 1999). Cerebral malaria
may lead to capillary thrombosis, which is probably caused by intravascular ag-
gregates of parasitised erythrocytes in cerebral tissues, particularly in white mat-
ter (Molyneux, 2000). Astroglial reaction results in the formation of granulomata
and infarcts affecting the cortical ribbon and leading to seizures. Most other pro-
tozoan and helminthic infestations of the CNS lead to formation of granulomata,
which, if located in cortical tissues, may lead to partial seizures (Bittencourt et
al., 1999).
Viral Infections
Among the many viruses that have been associated with the development of
encephalitis are arboviruses, coxsackie, rubella, measles, herpes simplex, flavivirus
(Japanese encephalitis), and cytomegalovirus. Patients may present with seizures
during the acute encephalitic process but more often develop neurological dis-
ability, including epilepsy, as a long-term complication (Eeg-Olofsson, 2003).
Herpes simplex virus is the commonest and most severe viral encephalitis in
immunocompetent subjects and epilepsy as its aftermath is particularly devastat-
ing (Marks et al., 1992).
HIV infections may be complicated by a subacute cortical and subcortical
encephalopathy with progressive dementia, myoclunus and tonic-clonic seizures
(Modi et al., 2000). Partial seizures in patients with HIV are usually the result of
secondary infections with cytomegalovirus, cryptococcus or toxoplasmosis.
Bacterial Infections
Bacterial infections of the CNS usually involve the meninges or cerebral
parenchyma and present as either meningitis or cerebral abscess. Acute bacterial
meningitis is usually caused by H. influenzae, N. meningitidis, S. pneumoniae or
streptococcus infections. Although it may occur in any age group, children are
the group more likely to contract bacterial meningitis. Five to ten percent of sur-
vivors of acute bacterial meningitis will develop chronic epilepsy and this is usu-
ally associated with learning deficits and other neurological disabilities (Marks et
al., 1992; Bittencourt et al., 1999; Oostenbrink et al., 2002).
Cerebral abscesses and intracranial empyemas are usually associated with a
clear port of entry like sinusitis, otitis media, dental abscess or cardiac
valvopathies (Bittencourt et al., 1999). In the majority of cases anaerobic organ-
isms are involved. Epilepsy in the aftermath of a cortical abscess is the rule, and
it is usually highly refractory to treatment and often associated with other neuro-
logical disabilities. Tuberculosis of the central nervous system may involve the
meninges and cerebral parenchyma and is associated with neurological disabili-
ties in a large number of survivors (Bittencourt et al., 1999). Many of these will
have partial epilepsy that is often refractory to treatment.
Fungal Infections
Fungal infections of the CNS are rare in immunocompetent subjects, particu-
larly in high-income economies. The fungi are acquired by inhalation of spores
that lodge initially in the lungs or paranasal sinuses and may seed to any organ,
although with certain topographic preferences depending on the organism
(Bittencourt et al., 1999). C. neoformans, C. immitis, H. capsulatum, C. albicans,
A. fumigatus and A. flavus, and Mucoraceae sp. are the fungal species most likely
to be involved and all of them may eventually provoke seizures.
Protozoal Infections
Plasmodium falciparum and Toxoplasma gondii are associated with epilepsy,
although the former is by far the bigger culprit. Cerebral malaria may develop
abruptly or subacutely, during systemic uncomplicated, as well as during severe,
falciparum malaria and may have severe consequences. Survivors are at high risk
of neurological disabilities including epilepsy (Waruiru et al., 1996; Molyneux,
2000; Versteeg et al., 2003). It is likely that this is responsible for the higher
prevalence of epilepsy in endemic area. Intrauterine T. gondii infections are asso-
ciated with a severe congenital encephalopathy with epilepsy as one of the symp-
toms. It may also cause seizures in immunocompromised patients. Recently, a
suggestion has been made that it may be responsible for many cases of cryptoge-
nic partial epilepsy but this has not been fully elucidated (Stommel et al., 2001).
Helminthic Infestations
A number of helminthic infestations can occasionally reach the CNS and
lead to seizures. Taenia solium is probably the commonest of these helminthic
infestations but Paragonomiasis westermani, Echinoccocus granulosis,
Spargonomiasis mansonoides and Schistosoma japonicum and S. mansoni have
also been implicated (Pal et al., 2000; Bittencourt et al., 1999). Recently, sugges-
tions have been made that Toxocara canis could be a major culprit for the higher
prevalence of epilepsy in low-income economies (Nicoletti et al., 2002).
Taeniasis and cysticercosis are caused by Taenia solium (Carpio, 2002). They
are closely related to poor sanitation, and the coexistence of humans and pigs is a
major factor. Humans are the final host for Taenia solium while hogs are the
intermediate host. Eating uncooked pork contaminated with taenia cysts will lead
to intestinal taeniasis. When humans, instead of pigs, ingest taenia eggs they may
become the intermediate host and this may lead to neurocysticercosis. In pigs the
cysts tend to lodge in subcutaneous and muscle tissues but in humans there is an
attraction for the brain, particularly well irrigated areas like the cortex and the
choroidal plexus, Here infestation may lead to epilepsy and other neurological
symptoms. Indeed, neurological problems resulting from neurocysticercosis are
very common in vast areas of South America, West Africa and Asia (Medina et
al., 1990; Bergen, 1998; Sander, 2003). Neurocysticercosis is probably the most
preventable form of epilepsy worldwide.
Cerebral hydatidosis is caused by Echinococcus granulosus and occurs in
sheep-raising areas. It is acquired mainly by eating food contaminated with dog
feces. Epilepsy is a rare complication of this condition (Bittencourt et al., 1999).
Paragonimiasis is a parasitic disease caused by Paragonimiasis westermanii
and is common in some endemic areas in the Far East. Like neurocysticercosis, it
may be associated with epilepsy when humans become the intermediate host (usu-
ally a fish). It is acquired by eating undercooked or raw crab or crayfish
(Bittencourt et al., 1999).
A recent report has suggested the possibility of Toxocara canis being the
culprit for partial epilepsy in low-income countries (Nicoletti et al., 2002). An
odds ratio of 18.2 for the development of late onset epilepsy has been reported in
association with positive serology for Toxocara canis. This same study in Bolivia
found an odds ratio for positive serology for Taenia solium of 3.6. This is inter-
esting as, over 30 years ago, Woodruff claimed that dog ownership was a major
risk factor for epilepsy, but this was never taken forward (Woodruff et al., 1966).
Further studies are urgently needed to clarify this issue.
Conclusion
Much of the existing evidence indicates that epilepsy resulting from infec-
tions is a major cause of neurological disability in low-income countries. Indeed,
it is probably responsible for the higher incidence of epilepsy in these areas and is
the commonest preventable cause of epilepsy worldwide. Improvement in basic
sanitation is likely to be crucial to decrease the global burden of epilepsy. Much
remains to be done in this area. Studies are urgently needed to elucidate the whole
spectrum of attributable risk factors for epilepsy. More research is also needed to
understand the molecular basis of all epilepsies particularly the ones caused by
infectious agents.
REFERENCES
Annegers JF, Rocca WA, Hauser WA. 1996. Causes of epilepsy: contributions of the Rochester epi-
demiology project. Mayo Clinic Proceedings 71:570–575.
Bergen DC. 1998. Preventable neurological diseases worldwide. Neuroepidemiology 17:67–73.
Bergen DC and Silberberg D. 2002. Nervous system disorders: a global epidemic. Archives of Neurol-
ogy 59:1194–1196.
Bittencourt PR, Sander JW, Mazer S. 1999. Viral, bacterial, fungal and parasitic infections associated
with seizure disorders. Pp. 145–174 in Handbook of Clinical Neurology, Vol. 72: The Epilep-
sies, H.Meinardi, ed. Amsterdam: Elsevier Sciences.
Carpio A. 2002. Neurocysticercosis: an update. Lancet Infectious Diseases 2:751–762.
Cockerell OC, Johnson AL, Sander JW, Hart YM, Goodridge DM, Shorvon SD. 1994. Mortality from
epilepsy: results from a prospective population-based study. Lancet 344:918–921.
Commission on Tropical Diseases of the International League Against Epilepsy. 1994. Relationship
between epilepsy and tropical diseases. Epilepsia 35:89–93.
Eeg-Olofsson O. 2003. Virological and immunological aspects of seizure disorders. Brain and Devel-
opment 25:9–13.
Hauser WA and Annegers JF. 1991. Risk factors for epilepsy. Epilepsy Research Supplement 4:45–
52.
Heaney DC, MacDonald BK, Everitt A, Stevenson S, Leonardi GS, Wilkinson P, Sander JW. 2002.
Socioeconomic variation in incidence of epilepsy: prospective community based study in south
east England. British Medical Journal 325:1013–1016.
Johnson MR and Sander JW. 2001. The clinical impact of epilepsy genetics. Journal of Neurology,
Neurosurgery, and Psychiatry 70:428–430.
Marks DA, Kim J, Spencer DD, Spencer SS. 1992. Characteristics of intractable seizures following
meningitis and encephalitis. Neurology 42:1513–1518.
Medina MT, Rosas E, Rubio-Donnadieu F, Sotelo J. 1990. Neurocysticercosis as the main cause of
late-onset epilepsy in Mexico. Archives of Internal Medicine 150:325–327.
Modi G, Modi M, Martinus I, Saffer D. 2000. New-onset seizures associated with HIV infection.
Neurology 55:1558–1561.
Molyneux ME. 2000. Impact of malaria on the brain and its prevention. Lancet 355:671–672.
Nicoletti A, Bartoloni A, Reggio A, Bartalesi F, Roselli M, Sofia V, Rosado Chavez J, Gamboa
Barahona H, Paradisi F, Cancrini G, Tsang VC, Hall AJ. 2002. Epilepsy, cysticercosis, and
toxocariasis: a population-based case-control study in rural Bolivia. Neurology 58:1256–1261.
Oostenbrink R, Moons KG, Derksen-Lubsen G, Grobbee DE, Moll HA. 2002. Early prediction of
neurological sequelae or death after bacterial meningitis. Acta Paediatrica 91:391–398.
Pal DK, Carpio A, Sander JW. 2000. Neurocysticercosis and epilepsy in developing countries. Jour-
nal of Neurology, Neurosurgery, and Psychiatry 68:137–143.
Sander JW. 2003. The epidemiology of epilepsy revisited. Current Opinion in Neurology 16:165–
170.
Sander JW and Shorvon SD. 1996. Epidemiology of the epilepsies. Journal of Neurology, Neurosur-
gery, and Psychiatry 61:433–443.
Scott RA, Lhatoo SD, Sander JW. 2001. The treatment of epilepsy in developing countries: where do
we go from here? Bulletin of the World Health Organization 79:344–351.
Stommel EW, Seguin R, Thadani VM, Schwartzman JD, Gilbert K, Ryan KA, Tosteson TD, Kasper
LH. 2001. Cryptogenic epilepsy: an infectious etiology? Epilepsia 42:436–438.
Versteeg AC, Carter JA, Dzombo J, Neville BG, Newton CR. 2003. Seizure disorders among rela-
tives of Kenyan children with severe falciparum malaria. Tropical Medicine and International
Health 8:12–16.
Walker MC, White HS, Sander JW. 2002. Disease modification in partial epilepsy. Brain 125:1937–
1950.
Waruiru CM, Newton CR, Forster D, New L, Winstanley P, Mwangi I, Marsh V, Winstanley M,
Snow RW, Marsh K. 1996. Epileptic seizures and malaria in Kenyan children. Transactions of
the Royal Society of Tropical Medicine and Hygiene 90:152–155.
Woodruff AW, Bisseru B, Bowe JC. 1966. Infection with animal helminths as a factor in causing
poliomyelitis and epilepsy. British Medical Journal 5503:1576–1579.
Too often, infectious diseases have been distinguished from chronic dis-
eases, as though these are mutually exclusive categories competing for recogni-
tion as a leading public health priority. Nowhere is this view less sustainable
than in the field of childhood disability, particularly in developing countries.
Worldwide, infections are among the leading causes of chronic, developmental
disabilities in children, along with and sometimes interacting with genetic and
nutritional causes (Institute of Medicine, 2001). In developing countries today,
infections that are ostensibly preventable or controllable continue to be impor-
tant causes of damage to the developing nervous system resulting in early and
long-term cognitive, motor, seizure, hearing, vision, and behavioral disabilities.
Infectious causes of developmental disabilities thus take a major and potentially
unavoidable toll on the population health and economies of low-income coun-
tries today. This paper reviews some of the major infectious causes of develop-
Congenital Infections
Numerous prenatal infections can damage the developing nervous system or
senses, causing long-term disabilities in children (Levine et al., 2001). The occur-
rence, nature, and severity of effects vary not only with the type of organism but
also often with the timing of the exposure. For example, first or second trimester
exposure to toxoplasmosis, cytomegalovirus, and varicella infections may result
in a range of impairments recognizable at birth, including microcephaly, hydro-
cephaly, growth retardation, blindness, seizures, and skin disorders (Remington
et al., 1995; Dunn et al., 1999), whereas exposure late in pregnancy or during
delivery may result in unapparent infection at birth and onset of developmental
delay during infancy or childhood (Koppe et al., 1986).
Congenital Syphilis
The first congenital disability to be linked to an infectious cause (the spiro-
chete Treponema pallidum), congenital syphilis is preventable through routine
antenatal screening and treatment with penicillin. As a result, it is now a rela-
tively rare occurrence in developed countries, but in some low-resource settings
where routine antenatal care is lacking or where cost barriers prevent access to
treatment, recent studies have reported that 4 to 11 percent of births occur to
women with positive syphilis tests at delivery (Southwick et al., 2001; Frank and
Duke, 2000; Walker and Walker, 2002). The outcomes of congenital syphilis
range from fetal and infant death to premature birth, and survival with or without
neurological manifestations, which can include deafness, interstitial keratitis, and
mental retardation. The most severe outcomes generally result when conception
occurs during the early stages of maternal syphilis infection. Outcomes are less
severe when conception occurs during the latent state of maternal infection, and
clinical manifestations of congenital syphilis are thought to be least severe when
onset of maternal infection occurs during the third trimester of pregnancy (Wicher
and Wicher, 2001). Animal studies suggest that outcomes may also be modulated
by the genetic background of the conceptus (Wicher et al., 1994). Prevention of
congenital syphilis requires interventions to reduce the risk of sexual transmis-
sion to women of childbearing age, and expansion of antenatal screening and
access to treatment. Although the effectiveness and cost-effectiveness of these
interventions have been established in developed countries, a recent study in South
Africa identified logistical difficulties that prevent timely diagnostic results and
access to treatment even when antenatal screening can be accomplished
(Beksinska et al., 2002). These difficulties include late presentation for antenatal
care, transportation delays that delay access to accurate laboratory results, and
Congenital Toxoplasmosis
Congenital toxoplasmosis results from transplacental transmission of infec-
tion with the protozoan parasite Toxoplasma gondii following an acute episode of
maternal infection during pregnancy. The clinical manifestations can include
chorioretinitis, intracranial calcification, necrotizing encephalopathy, microceph-
aly, cranial nerve palsies, spastic hemi- or quadriparesis, seizures, cognitive dis-
ability, and death. Clinical signs may be absent at birth, but infants with congeni-
tal toxoplasmosis may develop cognitive and vision disabilities by late childhood.
While the risk of transplacental transmission has been found to increase with
increasing gestational age, approaching 90 percent during the third trimester, the
severity of clinical manifestations appears to decrease with increasing gestational
age (Jones et al., 2001). Those previously uninfected are susceptible to acute
toxoplasmosis infection through ingestion of raw or inadequately cooked infected
meat, contaminated unwashed fruits and vegetables, or oocytes from the feces of
infected cats. Although little is known about the frequency of congenital toxo-
plasmosis in low- and middle-income countries generally, a recent study from
Brazil reported a prevalence of 1 per 3,000 live births (Neto et al., 2000), more
than three times the rate reported in developed countries (Jara et al., 2001). Evi-
dence of the cost-effectiveness and safety of early detection (via prenatal or new-
born screening) and treatment of acute infection with antiparasitics is not consis-
tent or conclusive at this time (Jones et al., 2001; Roizen et al., 1995). Thus,
prevention of congenital toxoplasmosis in low-income countries requires further
research and perhaps more emphasis on educational programs regarding the risks
and specific hygienic precautions that can prevent acute infections during preg-
nancy.
Congenital Rubella
Congenial rubella leads to a range of adverse pregnancy outcomes or birth
defects but only when maternal rubella virus infection occurs within the first 18
weeks of pregnancy. Outcomes include fetal death, spontaneous abortion, still-
birth, premature birth and, among surviving infants, sensorineural deafness, cata-
racts and other visual impairments, mental retardation, autistic features, cardiac
defects, and increased susceptibility to juvenile diabetes and other chronic condi-
tions (Peckham and Newell, 2001). The earlier in gestation that the fetus becomes
infected, the greater the likelihood of multiple defects. Although congenital ru-
bella has been nearly eliminated in successfully vaccinated populations and with
a very high benefit-to-cost ratio (Plotkin et al., 1999), epidemics continue to oc-
cur in many developing countries (Lawn et al., 2000). Cutts and Vynnycky have
concluded from an extensive review of evidence that “Congenital rubella syn-
drome is an under-recognized public health problem in many developing coun-
tries. There is an urgent need for collection of appropriate data to estimate the
cost-effectiveness of a potential global rubella control program” (Cutts and
Vynnycky, 1999). A difficulty facing developing countries is that vaccination can
prevent congenital rubella only if high coverage is maintained. Incomplete vac-
cine coverage may actually increase the risk of congenital rubella infection by
reducing opportunities for natural immunity and increasing the mean age of in-
fection, thus increasing the susceptibility to infection of women of childbearing
age (Panagiotopoulos et al., 1999). The availability of a combined measles and
rubella vaccine may increase the feasibility of achieving adequate rubella vacci-
nation and improve opportunities to prevent congenital rubella throughout the
world (Banatvala, 1998).
Meningitis
Meningitis from major bacterial agents probably occurs more commonly in
the developing than in developed countries, though specific data are lacking.
Children under age 5 and the elderly are at highest risk. In developing countries,
pneumonia is the most common presentation of Haemophilus influenzae Type b
meningitis; it has been estimated that this cause of meningitis in developing coun-
tries has a case fatality rate of 30 percent and results in permanent nervous system
impairment in 20 percent of survivors (WHO, 2001a). Meningococcal meningitis
occurs sporadically in developed countries, but major epidemics of the disease
occur every several years in sub-Saharan Africa and South America. Case fatality
exceeds 50 percent in the absence of early and adequate treatment, and it is esti-
mated that 15 to 20 percent of survivors are left with deafness, seizures, and
mental retardation (Levine et al., 1998). Primary prevention of Haemophilus
influenzae Type b meningitis can be achieved by means of vaccination of all
infants or by chemoprophylaxis following close contact with an affected child.
Vaccination is the only practical method of preventing infection on a population
level. In developed countries where immunization against this disease during in-
fancy is routine, the incidence of Haemophilus influenzae Type b meningitis has
dropped dramatically (Levine et al., 1998). It has been argued that vaccination
against Haemophilus influenzae Type b infection is cost-effective in developing
countries as well (Levine et al., 1998), but information on the frequency of the
disease and its sequelae in developing countries is needed to guide the implemen-
tation of control strategies. Epidemics of meningococcal meningitis can be con-
trolled effectively by means of mass immunization campaigns resulting in over
80 percent coverage, while infection in endemic situations can be prevented by
chemoprophylaxis administered to close contacts of patients (Levine et al., 1998).
Information on the cost-effectiveness of these interventions in developing coun-
tries is needed.
Measles
Measles is an acute viral disease that is still a leading cause of death world-
wide, largely because of its occurrence among children under age 5 in developing
countries. Rarely (about 1/1,000 cases), measles infection causes encephalitis,
which can result in long-term nervous system sequelae among survivors. While
Vitamin A deficiency has been shown to increase the severity of measles infec-
tion, it is thought the infection can, in turn, exacerbate Vitamin A deficiency and
lead to blindness (Strebel, 1998). Vaccination using live, attenuated measles vi-
rus produces long-lasting immunity. Eradication of measles is theoretically fea-
sible, given the effectiveness of available vaccines and the likelihood that hu-
mans are the only reservoir capable of sustaining transmission of the measles
virus. Widespread vaccination has successfully prevented the spread of measles
in a number of developing countries, and is considered one of the most cost-
effective public health interventions ever undertaken (Strebel, 1998). However,
measles continues to be a major contributor to childhood death and disease world-
wide. Global eradication of this cause of developmental disability will require
sustained efforts.
Poliomyelitis
Polio was eradicated from the Western Hemisphere, the Western Pacific re-
gion, and Eastern Europe following a concerted international initiative (WHO,
2000b). This enteroviral disease, however, continues to threaten children in tropi-
cal Africa and to a lesser extent in South and Southeast Asia. Once established in
the intestines, poliovirus can enter the blood stream and invade the central ner-
vous system. As it multiplies, the virus destroys motor neurons and leads to irre-
versible paralysis. Immunization programs have effectively eradicated poliomy-
elitis from much of the world, but the disease remains endemic in much of
sub-Saharan Africa and parts of South and Southeast Asia. Reported immuniza-
tion coverage with the oral polio vaccine is still low in most African countries
(WHO, 2001b). Although worldwide eradication of polio as a cause of childhood
paralysis can be achieved by vaccination during infancy, meeting this goal will
require major commitments that may be difficult to sustain in the face of the
decline of the disease in much of the world (WHO, 2001b).
Conclusions
Children in developed countries have benefited for decades from interven-
tions such as maternal vaccination to prevent congenital rubella, pediatric vacci-
nations to prevent potentially brain-damaging childhood infections such as
Haemophilus influenza Type b, and early detection and effective management of
bacterial infections that can lead to meningitis or hearing loss. In addition,
antiretroviral therapies have become available in developed countries to prevent
pediatric HIV transmission. Unfortunately, cost and attitudinal and logistical bar-
riers prevent these interventions from reaching children at greatest risk in the
developing world. Extension of such interventions to low-income countries is a
necessary step toward the reduction of international inequalities in child health.
To effectively respond to the impacts of infectious causes of developmental
disabilities worldwide, proven methods of prevention must be implemented and
expanded within primary health care systems in low-income countries. Specific
interventions should be tailored to local epidemiology and resources and needs,
and should include vaccination programs with high coverage to prevent condi-
tions such as congenital rubella, bacterial meningitis and poliomyelitis, develop-
ment of laboratory facilities and networks to facilitate accurate diagnoses, and
commitment of resources to prevent other infectious diseases, such as pediatric
AIDS, malaria, neurocysticercosis, leprosy, viral encephalitis, and trachoma.
Additional recommendations articulated in the Institute of Medicine report on
1. Increase training and expertise at all levels of health care, as well as in the
educational and research sectors, in the intersection between infectious disease
control and child development.
2. Develop and maintain Internet cababilities to facilitate international com-
munication among those involved in the implementation of primary prevention
and rehabilitation programs for children with developmental disabilities in low-
income countries.
3. In the context of the successes of current primary health care child sur-
vival initiatives in low-income countries, it is essential that increased emphasis
be placed in low-income countries on prevention and early identification of de-
velopmental disabilities within the primary and maternal and child health care
systems. Those systems must in turn be linked to and supported by secondary and
tertiary medical services, as well as rehabilitation programs.
4. Develop increased capacity for evidence-based research by establishing
regional coordinating centers in low-income countries to enable the conduct of
clinical and community trials of the effectiveness of interventions to prevent in-
fectious causes of developmental disabilities.
5. Support research on factors that are crucial to understanding how to pre-
vent developmental disabilities in low-income countries, such as the etiology and
prevention of adverse perinatal outcomes and the impact of maternal education
and alleviation of poverty on the prevention of infections resulting in develop-
mental disabilities.
6. Develop practical methods for surveillance of infections leading to child-
hood disabilities.
7. Document nervous system sequelae of cerebral malaria and their prevention.
8. Determine the cost-effectiveness of methods for the prevention of preva-
lent infections that result in developmental disabilities.
REFERENCES
Banatvala JE. 1998. Rubella—could do better. Lancet 351:849–850.
Beksinska ME, Mullick S, Kunene B, Rees H, Deperthes B. 2002. A case study of antenatal syphilis
screening in South Africa: successes and challenges. Sexually Transmitted Diseases 29:32–37.
Belman AL. 1990. AIDS and pediatric neurology. Neurologic Clinics 8:571–603.
Breslau N, DelDotto JE, Brown GG, Kumar S, Ezhuthachan S, Hufnagle KG, Peterson EL. 1994. A
gradient relationship between low birth weight and IQ at age 6 years. Archives of Pediatric and
Adolescent Medicine 148:377–383.
Cook JA. 1998. Trachoma. Bulletin of the World Health Organization 76(Suppl 2):139–140.
Cutts FT and Vynnycky E. 1999. Modelling the incidence of congenital rubella syndrome in develop-
ing countries. International Journal of Epidemiology 28:1176–1184.
Dickson R, Awasthi S, Williamson P, Demellweek C, Garner P. 2000. Effects of treatment for intes-
tinal helminth infection on growth and cognitive performance in children: systematic review of
randomized trials. British Medical Journal 320:1697–1701.
Donders GG, Desmyter J, De Wet DH, Van Assche FA. 1993. The association of gonorrhoea and
syphilis with premature birth and low birthweight. Genitourinary Medicine 69:98–101.
Dunn D, Wallon M, Peyron F, Petersen E, Peckham C, Gilbert R. 1999. Mother-to-child transmission
of toxoplasmosis: risk estimates for clinical counselling. Lancet 353:1829–1833.
Durkin MS, Khan NZ, Davidson LL, Huq S, Munir S, Rasul E, Zaman SS. 2000. Prenatal and post-
natal risk factors for mental retardation among children in Bangladesh. American Journal of
Epidemiology 152:1024–1033.
European Mode of Delivery Collaboration. 1999. Elective caesarean-section versus vaginal delivery
in prevention of vertical HIV-1 transmission: a randomized clinical trial. Lancet 353:1035–
1039.
Fishkin PE, Armstrong FD, Routh DK, Harris L, Thompson W, Miloslavich K, Levy JD, Johnson A,
Morrow C, Bandstra ES, Mason CA, Scott G. 2000. Brief report: relationship between HIV
infection and WPPSI-R performance in preschool-age children. Journal of Pediatric Psychol-
ogy 25:347–351.
Frank D and Duke T. 2000. Congenital syphilis at Goroka Base Hospital: incidence, clinical features
and risk factors for mortality. Papua New Guinea Medical Journal 43:121–126.
Helen Keller International. 2001. Trachoma. Available at: http://www.hki.org/programs/
trachoma.html.
Institute of Medicine. 2001. Neurological, Psychiatric, and Developmental Disorders: Meeting the
Challenge in the Developing World. Washington, DC: National Academy Press.
International Perinatal HIV Group. 1999. The mode of delivery and the risk of vertical transmission
of human immunodeficiency virus type 1: a meta-analysis of 15 prospective cohort studies. New
England Journal of Medicine 340:977–987.
Jain S, Reddy RG, Osmani SN, Lockwood DN, Suneetha S. 2002. Childhood leprosy in an urban
clinic, Hyderabad, India: clinical presentation and the role of household contacts. Leprosy Re-
view 73:248–253.
Jara M, Hsu HW, Eaton RB, Demaria A Jr. 2001. Epidemiology of congenital toxoplasmosis identi-
fied by population-based newborn screening in Massachusetts. Pediatric Infectious Disease
Journal 20:1132–1135.
Jones JL, Lopez A, Wilson M, Schulkin J, Gibbs R. 2001. Congenital toxoplasmosis: a review. Ob-
stetrical and Gynecological Survey 56:296–305.
Koppe JG, Loewer-Sieger DH, de Roever-Bonnet H. 1986. Results of 20-year follow-up of congeni-
tal toxoplasmosis. Lancet 1:254–256.
Lawn JE, Reef S, Baffoe-Bonnie B, Adadevoh S, Caul EO, Griffin GE. 2000. Unseen blindness,
unheard deafness, and unrecorded death and disability: congenital rubella in Kumasi, Ghana.
American Journal of Public Health 90:1555–1561.
Levine MI, Chervenak FA, Whittle M, eds. 2001. Fetal and Neonatal Neurology and Neurosurgery.
London: Churchill Livingston.
Levine OS, Schwartz B, Pierce N, Kane M. 1998. Development, evaluation and implementation of
Haemophilus influenzae type b vaccines for young children in developing countries: current
status and priority actions. Pediatric Infectious Disease Journal 17 (9 Suppl):S95–113.
Macmillan C, Magder LS, Brouwers P, Chase C, Hittelman J, Lasky T, Malee K, Mellins CA, Velez-
Borras J. 2001. Head growth and neurodevelopment of infants born to HIV-1 infected drug-
using women. Neurology 57:1402–1411.
Neto EC, Anele E, Rubim R, Brites A, Schulte J, Becker D, Tuuminen T. 2000. High prevalence of
congenital toxoplasmosis in Brazil estimated in a 3-year prospective neonatal screening study.
International Journal of Epidemiology 29:941–947.
O’Shea TM and Dammann O. 2000. Antecedents of cerebral palsy in very low birthweight infants.
Clinics in Perinatology 27:285–302.
Panagiotopoulos T, Antoniadou I, Valassi-Adam E. 1999. Increase in congenital rubella occurrence
after immunization in Greece: retrospective survey and systematic review. British Medical Jour-
nal 319:1462–1467.
Peckham CS and Newell ML. 2001. Viral infections. Pp. 553–572 in Fetal and Neonatal Neurology
and Neurosurgery, MI Levine, FA Chervenak, M Whittle, eds. London: Churchill Livingston.
Peckam CS, Chin KS, Coleman JC, Henderson K, Hurley R, Preece PM. 1983. Cytomegalovirus
infection in pregnancy: preliminary findings from a prospective study. Lancet 1:1352–1355.
Plotkin SA, Katz M, Cordero JF. 1999. The eradication of rubella. Journal of the American Medical
Association 281:561–562.
Remington JS, McLeod R, Desmonts G. 1995. Toxoplasmosis. Pp. 140–267 in Infectious Diseases of
the Fetus and Newborn Infant, JS Remington and JO Klein, eds. Philadelphia: WB Saunders.
Roizen N, Swisher CN, Stein MA, Hopkins J, Boyer KM, Holfels E, Mets MB, Stein L, Patel D,
Meier P, et al. 1995. Neurologic and developmental outcome in treated congenital toxoplasmo-
sis. Pediatrics 95:11–20.
Siraprapasiri T, Sawaddiwudhipong W, Rojanasuphot S. 1997. Cost benefit analysis of Japanese
encephalitis vaccination program in Thailand. Southeast Asian Journal of Tropical Medicine
and Public Health 28:143–148.
Smith R, Malee K, Charurat M, Magder L, Mellins C, Macmillan C, Hittleman J, Lasky T, Llorente
A, Moye J. 2000. Timing of perinatal human immunodeficiency virus type 1 infection and rate
of neurodevelopment. The Women and Infant Transmission Study Group. Pediatric Infectious
Disease Journal 19:862–871.
Southwick KL, Blanco S, Santander A, Estenssoro M, Torrico F, Seoane G, Brady W, Fears M, Lewis
J, Pope V, Guarner J, Levine WC. 2001. Maternal and congenital syphilis in Bolivia, 1996:
prevalence and risk factors. Bulletin of the World Health Organization 79:33–42.
Strebel PM. 1998. Measles. Bulletin of the World Health Organization 76 (Suppl. 2):154–155.
Walker DG and Walker GJ. 2002. Forgotten but not gone: the continuing scourge of congenital syphi-
lis. Lancet Infectious Diseases 2:432–436.
Wicher V and Wicher K. 2001. Pathogenesis of maternal-fetal syphilis revisited. Clinical Infectious
Diseases 33:354–363.
Wicher V, Baughn RE, Wicher K. 1994. Congenital and neonatal syphilis in guinea pigs show a
different pattern of immune response. Immunology 82:404–409.
Wolf MJ, Wolf B, Beunen G, Casaer P. 1999. Neurodevelopmental outcome at 1 year in Zimbabwean
neonates with extreme hyperbilirubinaemia. European Journal of Pediatrics 158:111–114.
World Health Organization. 1998. Malaria: Fact Sheet Number 94. Available at: http://www.who.int/
inf-fs/en/fact094.html.
World Health Organization. 2000a. Control of Major Blinding Diseases and Disorders: Fact Sheet
Number 214. Available at: http://www.who.int/inf-fs/en/fact214.html.
World Health Organization. 2000b. Global Polio Eradication Initiative. Available at: http://
www.polioeradication.org/vaccines/polioeradication/all/partners/default.asp.
World Health Organization. 2001a. Haemophilus Influenzae Type b Disease. [Online] Available at:
http://www.who.int/vaccines-diseases/diseases/hib.shtml.
World Health Organization. 2001b. Polio Eradication: Final 1% Poses Greatest Challenge. Available
at: http://www.who.int/inf-pr-2001/en/pr2001-17.html.
Human T-cell lymphotropic virus type 1 (HTLV-1) was the first human
retrovirus to be described. It was discovered simultaneously in the United States
and in Japan in 1980 (Poiesz et al., 1980; Hinuma et al., 1981). As documented
for all retroviruses, HTLV-1 produces a permanent cell infection. Therefore, all
carriers are potential sources of transmission of the infection.
Epidemiology
Geographical Distribution
HTLV-1 has an ubiquitous distribution, with well-described endemic areas.
An area is called endemic for HTLV-1 if 2–10 percent of the healthy adult popu-
lation is infected. The islands of Kyushu and Okinawa, in southwestern Japan, are
hyperendemic areas for HTLV-1, 15 percent of the healthy adult population carry
the virus (Blattner, 1990). Moderate rates of infection have been reported in West
Africa, Australia, and the Caribbean (Caribbean Epidemiology Center, 1990;
Delaporte et al., 1989; Nerurkar et al., 1993). In South America, Brazil, Colom-
bia, and Peru are HTLV-1 endemic areas (Zurita et al., 1997; Gabbai et al., 1993;
Zaninovic et al., 1994); the virus is also present in Ecuador (Guderian et al.,
1994), Paraguay (de Cabral et al., 1995), Chile (Cartier and Cartier, 1996) and
Argentina (Bouzas et al., 1994). In Peru, the virus is highly prevalent in some
population and ethnic groups. Sixteen percent of immigrants from Japan—par-
ticularly from Okinawa—are seropositive. However, in the first generation of
these immigrants born in Peru, the virus is prevalent in 4 percent of the popula-
tion, and is not present in the second generation (Gotuzzo et al., 1996). Similar
trends were reported in Hawaii (Blattner et al., 1986) and Bolivia (Tsugane et al.,
1988). A study of HTLV-1 infection in asymptomatic women in Peru found preva-
lence rates of 3.8 percent among Afro-American women in Chincha, a coastal
town south of Lima, 1.3 percent among the Quechua population of the central
highlands (Ayacucho) and 3.8 percent in the population of northern Lima
(Sanchez-Palacios, 2003). In other regions in South America, in which there is a
strong presence of African Americans, such as Tumaco (Colombia) and Bahia
(Brazil), the prevalence of HTLV-1 ranges from 2–5 percent in the healthy adult
population.
Transmission
HTLV-1 is transmitted through modes similar to those described for HIV,
but there are also important differences that are explained by the requirement of
infected lymphocytes for the transmission of HTLV-1.
Vertical Transmission
Intrauterine transmission of HTLV-1 is very rare, and prolonged
breastfeeding seems to be the main risk factor associated with this type of trans-
mission. In Peru, breastfeeding is the most common route of transmission of
HTLV-1. In a study of 120 HTLV-1-infected Peruvian women and their off-
spring, infection was not detected in children who were not breastfed, but was
documented in 14 percent of those who received maternal milk for less than 6
months and in 31 percent of those breastfed for more than 6 months (E. Gotuzzo,
unpublished data). Moreover, in a hyperendemic area in southwestern Japan,
screening pregnant women and abstaining from breastfeeding has been docu-
mented to dramatically decrease the prevalence of HTLV-1 (Katamine, 1999).
HTLV-1-related disease in mothers may also be associated with the increased
risk of transmission of the virus to their children, as suggested by a recent study
which found that HTLV-1 is present in 43 percent of children born from mothers
with strongyloidiasis, and 20 percent of children born from mothers with tropical
spastic paraparesis (p < 0.01). Gender also seemed to be a factor, as HTLV-1 is
transmitted to 17 percent of males and to 32 percent of females (p < 0.01) (E.
Gotuzzo, submitted for publication).
Parenteral Transmission
HTLV-1 is transmitted less efficiently than HIV in whole blood transfusions.
Fresh frozen plasma, which can transmit HIV, has not been associated with the
transmission of HTLV-1. In addition, the efficacy of HTLV-1 transmission de-
creases when blood is stored for more than one week (Okochi et al., 1984). These
observations point to the need for viable lymphocytes to establish infection with
HTLV-1. Transmission through transfusion of whole blood has been estimated to
infect between 50 and 60 percent of recipients (Larson and Taswell, 1988). A
national survey in Peru indicated that 1.2 percent of 142,500 blood donors were
HTLV-1 seropositive. Epidemiologic studies of the general population in Carib-
bean countries have consistently shown that the prevalence of HTLV-1 signifi-
cantly increases with age, is higher in women, specifically in low socioeconomic
strata, and correlates with a history of blood transfusion (Murphy et al., 1996).
The efficacy of HTLV-1 transmission through needle sharing by intravenous drug
users is very low (Gradilone et al., 1986).
Cancer in Lima, Peru, each year, and 10 percent (30) of these cases are associated
with HTLV-1.
tremor versus 9 percent among slow progressors, p < 0.001) (E. Gotuzzo, unpub-
lished data).
Urinary symptoms are a frequent complaint of TSP patients. Initially, pa-
tients report difficulties to initiate voiding. Not uncommonly, patients mention
the need to put external pressure on their lower abdomen in order to urinate. In
severe cases, patients cannot maintain voiding without compressing the abdo-
men, sometimes leading to urinary retention. Recurrent urinary infections are
common, probably reflecting disorders in bladder emptying. Dysfunction of the
detrusor muscle has been implicated in the urinary tract involvement in TSP.
Manifestations of immune hyperactivity other than TSP—such as Sjögren’s
syndrome, uveitis, arthritis, Behçet’s disease and thyroiditis—have been repeat-
edly observed among patients with TSP.
Currently, there is neither specific nor standardized treatment for HTLV-1
and TSP. Prolonged periods of systemic steroids appear to improve clinical symp-
toms of TSP and recently, antiretroviral drugs effective in treating HIV, such as
lamivudine and zidovudine, have been used with relative success in treating pa-
tients with TSP (Sheremata et al., 1993). A combination of corticosteroids,
antiretroviral drugs, and rehabilitation might considerably improve the quality of
life of TSP patients—particularly if treatment is started early in the course of the
disease in those cases with rapid progression (Araujo et al., 1995).
group (5 percent, 1 out of 21) and in a group with intestinal strongyloidiasis (10
percent, 6 of 62) was statistically significant (p < 0.001) (Gotuzzo et al., 1999). A
report of decreased therapeutic efficacy of thiabendazole exists among patients
with concomitant S. stercoralis-HTLV-1 infection in Okinawa (Sato et al., 1994).
Terashima showed that the failure of the standard treatment against intestinal
strongyloidiasis with thiabendazole or ivermectin was an important marker for
suspecting HTLV-1 infection. Some reports suggest that there is a relation be-
tween strongyloidiasis and ATLL in HTLV-1-positive patients. It is not clear
whether Strongyloides acts as a trigger, shortening the incubation time of leuke-
mia, or a marker of high proviral load.
Conclusion
HTLV-1 produces three different clinical patterns: lymphoproliferative dis-
ease (ATLL), autoimmune syndromes (TSP), and infections associated with im-
munosuppression (strongyloidiasis, crusted scabies and others). The infection is
endemic in several countries in Latin America. HTLV-1 is transmitted mainly
through breastfeeding, transfusion of whole blood, and as a STD. These modes of
transmission are vulnerable to simple and effective methods of control, such as
REFERENCES
Araujo AQ, Leite AC, Dultra SV, Andrada-Serpa MJ. 1995. Progression of neurological disability in
HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Journal of the Neuro-
logical Sciences 129:147–151.
Bartholomew C, Blattner W, Cleghorn F. 1987a. Progression to AIDS in homosexual men co-infected
with HIV and HTLV-I in Trinidad. Lancet 2:1469.
Bartholomew C, Saxinger C, Clark JW, Gail M, Dudgeon A, Mahabir B, Hull-Drysdale B, Cleghorn
F, Gallo RC, Blattner WA. 1987b. Transmission of HTLV-1 and HIV among homosexual men
in Trinidad. Journal of the American Medical Association 257:2604–2608.
Blank A, Herrera M, Lourido MA, Rueda R, Blank M. 1995. Infective dermatitis in Colombia. Lancet
346:710.
Blattner WA. 1990. Epidemiology of HTLV-1 and associated diseases. In Human Retrovirology:
HTLV-1, WA Blattner, ed. New York: Raven Press.
Blattner WA, Nomura A, Clark JW, Ho GY, Nakao Y, Gallo R, Robert-Guroff M. 1986. Modes of
transmission and evidence for viral latency from studies of human T-cell lymphotropic virus
type I in Japanese migrant populations in Hawaii. Proceedings of the National Academies of
Sciences USA 83:4895–4898.
Bouzas MB, Zapiola I, Quiruelas S, Gorvein D, Panzita A, Rey J, Carnese FP, Corral R, Perez C, Zala
C, et al. 1994. HTLV Type I and HTLV Type II infection among Indians and Natives from
Argentina. AIDS Research and Human Retroviruses 10:1567–1571.
Brites C, Weyll M, Pedroso C, Badaro R. 2002. Severe and Norwegian scabies are strongly associated
with retroviral (HIV/HTLV-1) infection in Bahia, Brazil [letter]. AIDS 16:1292–1293.
Caribbean Epidemiology Center. 1990. Public Health Implications of HTLV-1 in the Caribbean.
Weekly Epidemiological Record 65:63–65.
Cartier L and Cartier E. 1996. HTLV-I/II in Chile. Pp. 150–158 in HTLV, Truths and Questions, V
Zaninovic, ed. Cali, Colombia: Fundación MAR.
Cortes E, Detels R, Aboulafia D, Li XL, Moudgil T, Alam M, Bonecker C, Gonzaga A, Oyafuso L,
Tondo M. 1989. HIV-1, HIV-2 and HTLV-I infection in high risk groups in Brazil. New En-
gland Journal of Medicine 320:953–958.
de Cabral MB, Vera ME, Samudio M, Arias AR, Cabello A, Moreno R, Zapiola I, Bouzas MB,
Muchinik G. 1995. HTLV-I/II antibodies among three different Indian groups from Paraguay.
Journal of Acquired Immune Deficiency Syndromes and Human Retrovirology 19:548–549.
Delaporte E, Peeters M, Durand JP, Dupont A, Schrijvers D, Bedjabaga L, Honore C, Ossari S,
Trebucq A, Josse R, et al. 1989. Seroepidemiological survey of HTLV-1 infection among ran-
domized population of western central African countries. Journal of AIDS 2:410–413.
Gabbai AA, Bordin JO, Vieira-Filho JP, Kuroda A, Oliveira AS, Cruz MV, Ribeiro AA, Delaney SR,
Henrard DR, Rosario J, et al. 1993. Selectivity of human T lymphotropic virus type I (HTLV-I)
and HTLV-II infection among different populations in Brazil. American Journal of Tropical
Medicine and Hygiene 49:664–671.
Gessain A, Barin F, Vernant JC, Gout O, Maurs L, Calender A, de The G. 1985. Antibodies to human
T-lymphotropic virus type-I in patients with tropical spastic paraparesis. Lancet 2:407–410.
Gotuzzo E, Escamilla J, Phillips IA, Sanchez J, Wignall FS, Antigoni J. 1992. The impact of human
T lymphotropic virus type I/II infection on the prognosis of sexually acquired cases of acquired
immunodeficiency syndrome. Archives of Internal Medicine 152:1429–1432.
Gotuzzo E, Yamamoto V, Kanna M, et al. 1996. Human T lymphotropic virus type I infection among
Japanese immigrants in Peru. International Journal of Infectious Diseases 1:75–77.
Gotuzzo E, Terashima A, Alvarez H, Tello R, Infante R, Watts DM, Freedman DO. 1999. Strongy-
loides stercoralis hyperinfection associated with human T cell lymphotropic virus type-I infec-
tion in Peru. American Journal of Tropical Medicine and Hygiene 60:146–149.
Gradilone A, Zani M, Barillari G, Modesti M, Agliano AM, Maiorano G, Ortona L, Frati L, Manzari
V. 1986. HTLV-1 and HIV infection in drug addicts in Italy. Lancet 2:753–754.
Guderian R, Guevara A, Cooper P, Rugeles MT, Arango C. 1994. HTLV-1 infection and tropical
spastic paraparesis in Esmeraldas Province of Ecuador. Transactions of the Royal Society of
Tropical Medicine and Hygiene 88:399–400.
Henriquez C, Gotuzzo E, Cairampoma, et al. Impact of infection with HIV and/or HTLV-1 on the
outcome of hospitalization for tuberculosis in a public hospital in Peru. Abstract at the 4th World
Congress on Tuberculosis, Washington, DC, June 3–5, 2002.
Hinuma Y, Nagata K, Hanaoka M, Nakai M, Matsumoto T, Kinoshita KI, Shirakawa S, Miyoshi I.
1981. Adult T cell leukemia antigen in an ATL cell line and detection of antibodies to the
antigen in human sera. Proceedings of the National Academy of Sciences USA 78:6476–6480.
Jeffrey KJ, Usuku K, Hall SE, Matsumoto W, Taylor GP, Procter J, Bunce M, Ogg GS, Welsh KI,
Weber JN, Lloyd AL, Nowak MA, Nagai M, Kodama D, Izumo S, Osame M, Bangham CR.
1999. HLA alleles determine human T-lymphotropic virus-I (HTLV-1) proviral load and the
risk of HTLV-1-associated myelopathy. Proceedings of the National Academy of Sciences USA
96:3848–3853.
Kaplan JE, Osame M, Kubota H, Igata A, Nishitani H, Maeda Y, Khabbaz RF, Janssen RS. 1990. The
risk of development of HTLV-1 associated myelopathy/tropical spastic paraparesis among per-
sons infected with HTLV-1. AIDS 3:1096–1101.
Katamine S. 1999. Milk-borne transmission of human T-cell lymphotropic virus Type 1 (HTLV-1)
and its intervention in Nagasaki. Acta Medica Nagasakiensia 44:1–6.
Khabbaz R, Darrow WW, Hartley TM, Witte J, Cohen JB, French J, Gill PS, Potterat J, Sikes RK,
Reich R, et al.. 1990. Seroprevalence and risk factors for HTLV-1 infection among female
prostitutes in the United States. Journal of the American Medical Association 263:60–64.
LaGrenade L, Hanchard B, Fletcher V, Cranston B, Blattner W. 1990. Infective dermatitis of Jamai-
can children: a marker for HTLV-1 infection. Lancet 336:1345–1347.
Larson C and Taswell H. 1988. Human T-cell leukemia virus (HTLV-1) and blood transfusion. Mayo
Clinic Proceedings 63: 869–875.
Mani KS, Mani AJ, Montgomery, RD. 1969. A spastic paraplegic syndrome in South India. Journal
of the Neurological Sciences 9:179–199.
Murphy EL, Hanchard B, Figueroa JP, Gibbs WN, Lofters WS, Campbell M, Goedert JJ, Blattner
WA. 1989. Modelling the risk of adult T cell leukemia/lymphoma in persons infected with
human T lymphotropic virus type I. International Journal of Cancer 43:250–253.
Murphy EL, Wilks K, Hanchard B, Cranston B, Figueroa JP, Gibbs WN, Murphy J, Blattner WA.
1996. A case-control study of risk factors for seropositivity to HTLV-1 in Jamaica. Interna-
tional Journal of Epidemiology 25:1083–1089.
Nakada K, Kohakura M, Komoda H, Hinuma Y. 1984. High incidence of HTLV antibody in carriers
of Strongyloides stercoralis [letter]. Lancet 1:633.
Nakagawa M, Izumo S, Ijichi S, Kubota H, Arimura K, Kawabata M, Osame M. 1995. HTLV-1-
associated myelopathy: analysis of 213 patients based on clinical features and laboratory find-
ings. Journal of Neurovirology 1:50–61.
Nerurkar VR, Song KJ, Saitou N, Melland RR, Yanagihara R. 1993. Interfamilial genomic diversity
and molecular phylogeny of human T-cell lymphotrophic virus type I from Papua New Guinea
and the Solomon Islands. Virology 196:506–513.
Neva FA. 1986. Biology and immunology of human strongyloidiasis. Journal of Infectious Diseases
153:397–406.
Okochi K, Sata H, Hinuma Y. 1984. A retrospective study on transmission of adult T-cell leukemia
virus via blood transfusion: seroconversion in recipients. Vox Sanguinis 46:245–253.
Page JB, Lai SH, Chitwood DD, Klimas NG, Smith PC, Fletcher MA. 1990. HTLV-I/II seropositivity
and death from AIDS among HIV-1 seropositive intravenous drug users. Lancet 335:1439–
1441.
Paterson WD, Allen BR, Beveridge GW. 1973. Norwegian scabies during immunosuppressive
therapy. British Medical Journal 4:211–212.
Phillips I, Hyams KC, Wignall FS, Moran AY, Gotuzzo E, Sanchez J, Roberts CR. 1991. HTLV-1 co-
infection in a HIV-1 infected Peruvian population. Journal of Acquired Immune Deficiency
Syndromes 4:301–302.
Pierik LT and Murphy EL. 1991. The clinical significance of HTLV-I and HTLV-II infection in the
AIDS epidemic. Pp. 41–57 in AIDS Clinical Review, P Volberding and MA Jacobson, eds. New
York: Marcel Dekker.
Poiesz BJ, Ruscetti FW, Gadzar AF, Bunn PA, Minna JD, Gallo RC. 1980. Detection and isolation of
type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T
cell lymphoma. Proceedings of the National Academy of Sciences USA 77:7415–7419.
Sanchez-Palacios C, Gotuzzo E, Vandamme AM, Maldonado Y. 2003. Seroprevalence and risk fac-
tors for human T-cell lymphotropic virus (HTLV-1) infection among ethnically and geographi-
cally diverse Peruvian women. International Journal of Infectious Diseases 7:132–137.
Sato Y, Shiroma Y, Kiyuna S, Toma H, Kobayashi J. 1994. Reduced efficacy of chemotherapy might
accumulate concurrent HTLV-1 infection among strongyloidiasis patients in Okinawa, Japan.
Transactions of the Royal Society of Tropical Medicine and Hygiene 88:59.
Seiki M, Hattori S, Hirayama Y, Yoshida M. 1983. Human adult T-cell leukemia virus: Completed
nucleotide sequence of the provirus genoma integrated in leukemia cell DNA. Proceedings of
the National Academy of Sciences USA 80:3618–3622.
Sheremata WA, Benedict D, Squilacote DC, Sazant A, DeFreitas E. 1993. High-dose zidovudine
induction in HTLV-1-associated myelopathy: safety and possible efficacy. Neurology 43:2125–
2129.
Sweet RD. 1966. A pattern of eczema in Jamaica. British Journal of Dermatology 78:93–100.
Tajima K and Hinuma Y. 1992. Epidemiology of HTLV-I/II in Japan and the world. Gann Mono-
graph on Cancer Research 39:129–149.
Tajima K, Tominaga S, Suchi, Kawagoe T, Komoda H, Hinuma Y, Oda T, Fujita K. 1982. Epidemio-
logical analysis of the distribution of antibody to adult T-cell leukemia-virus-associated antigen:
possible horizontal transmission of adult T-cell leukemia virus. Gann 73:893–901.
Takatsuki K, Uchiyama J, Sagawa K, Yodoi J. 1977. Adult T-cell leukemia in Japan. Pp. 73–77 in
Topics in Hematology, S Sano, F Takaku, S Irino, eds. Amsterdam: Excerpta Medica.
Tsugane S, Watanabe S, Sugimura H, Otsu T, Tobinai K, Shimoyama M, Nanri S, Ishii H. 1988.
Infectious status of human T lymphotropic virus type I and hepatitis B virus among Japanese
immigrants in the Republic of Bolivia. American Journal of Epidemiology 128:1153–1161.
Wignall FS, Hyams KC, Phillips IA, Escamilla J, Tejada A, Li O, Lopez F, Chauca G, Sanchez S,
Roberts CR. 1992. Sexual transmission of human T-cell lymphotropic virus type I in Peruvian
prostitutes. Journal of Medical Virology 38:44–48.
Zaninovic V, Sanzón F, López F, Velandia G, Blank A, Blank M, Fujiyama C, Yashiki S, Matsumoto
D, Katahira Y, et al. 1994. Geographic independence of HTLV-I and HTLV-II foci in the Andes
Highland, the Atlantic Coast, and the Orinoco of Colombia. AIDS Research and Human
Retroviruses 10:97–101.
Zurita S, Costa C, Watts D, Indacochea S, Campos P, Sanchez J, Gotuzzo E. 1997. Prevalence of
human retroviral infection in Quillabamba and Cuzco, Peru: a new endemic area for human T-
cell lymphotropic virus type 1. American Journal of Tropical Medicine and Hygiene 56:561–
565.
file (Koziel, 1999). However, most studies are based on few individuals who
recovered infection either spontaneously or after interferon therapy (Gerlach et
al., 1999; Lohr et al., 1998). Thus longitudinal prospective studies analyzing the
early antiviral immune responses in acute hepatitis C infection are crucial for
understanding the pathogenesis of the disease and potentially in vaccine design.
Once chronic infection is established HCV-specific CD4+ T-cells compartmen-
talize in the liver and differ functionally and clontypically from those in the pe-
ripheral blood (Schirren et al., 2000; Bertoletti et al., 1997). The significance of
the intrahepatic CD4+ responses and their relation to liver injury have not been
comprehensively investigated since most studies focus on the peripheral com-
partment due to the difficulty in obtaining liver biopsies.
Khoby et al., 2000; Hammam et al., 2000). Morbidity in humans infected with S.
mansoni results primarily from deposition of parasite ova in the portal areas in-
ducing a T-cell-dependent granulomatous response which progresses to irrevers-
ible fibrosis and severe portal hypertension in more than 60 percent of cases. S.
mansoni infection in mice is characterized by a strong Th2-associated immune
response coupled with a defect in Th1-cell effector function (Sabin and Pearce,
1995). Although a predominant Th2 profile was shown to be beneficial in
polyparasitism, where mice infected with S. mansoni are capable of eliminating
Trichuris muris infection more efficiently than non-infected mice (Curry et al.,
1995), it is assumed to be harmful in most viral infections.
Concomitant schistosomiasis and HCV infection is common in Egypt and
other developing countries (Kamal et al., 2000a; Angelico et al., 1997; Pereira et
al., 1995). Patients with concomitant HCV and schistosomiasis exhibit a unique
clinical, virological, and histological pattern manifested by virus persistence with
high HCV RNA titers, higher necroinflammatory and fibrosis scores in their liver
biopsies and poor response to interferon therapy (Kamal et al., 2000a; Angelico
et al., 1997; Pereira et al., 1995; Kamal et al., 2000b). This results in a markedly
accelerated disease course once chronic HCV infection has been established. Our
understanding of the pathomechanisms leading to this accelerated disease pro-
gression in HCV/S. mansoni coinfection is still extremely limited. This
coinfection should be a valuable model to study the effect of one pathogen on the
pathogenesis of the other agent, especially the influence of an altered T helper
cell response on the other arms of the immune response as well as the clinical
outcome. The model of HCV/S. mansoni coinfection also offers a unique oppor-
tunity to define the role of HCV-specific T-cells in viral control as well as the
pathogenesis of HCV-related liver disease.
Comprehensive study of the different aspects of HCV/S. mansoni coinfection
has been conducted and the data were presented in several publications. These
studies, described below, provide insight into the mechanisms through which
infection with one pathogen can influence the immunopathogenesis and the clini-
cal course of another.
Conclusion
In summary, HCV infection is a worldwide problem for which there has been
insufficient success with treatment options presently available. The lack of a clear
understanding of the immunological events during acute and chronic infection
has hampered vaccine development and immunotherapeutic approaches to treat-
ment. From an immunological point of view the interplay between T helper cell
responses and CTL has been difficult to assess in humans, and infection with S.
mansoni offers the unique situation of studying the impact of an altered response
on the outcome and progression of HCV-related liver disease.
REFERENCES
Abdel Aziz F, Habib M, Mohamed M, Abdel Hamid M, Gamil F, Madkour S, Mikhail N, Thomas D,
Fix A, Strickland T, Anwar W, Ismail S. 2000. Hepatitis C virus infection in a community in the
Nile Delta: population description and HCV prevalence. Hepatology 32:111–115.
Alter MJ. 1997. Epidemiology of hepatitis C. Hepatology 26:62S–65S.
Angelico M, Renganathan E, Gandin C, Fathy M, Profili MC, Refai W, De Santis A, Nagi A, Amin
G, Capocaccia L, Callea F, Rapicetta M, Badr G, Rocchi G. 1997. Chronic liver disease in
Alexandria governorate, Egypt: contribution of schistosomiasis and hepatitis virus infections.
Journal of Hepatology 26:236–243.
Bertoletti A, D´Elios MM, Boni C, De Carli M, Zignego AL, Durazzo M, Missale G, Penna A,
Fiaccadori F, Del Prete G, Ferrari C. 1997. Different cytokine profiles of intrahepatic T cells in
chronic hepatitis B and hepatitis C virus infections. Gastroenterology 112:193–199.
Cerny A and Chisari FV. 1999. Pathogenesis of chronic hepatitis C: immunological features of he-
patic injury and viral persistence. Hepatology 30:595–601.
Chitsulo L, Engels D, Montresor A, Savioli L. 2000. The global status of schistosomiasis and its
control. Acta Tropica 77:41–51.
Curry AJ, Else KJ, Jones F, Bancroft A, Grencis RK, Dunn DW. 1995. Evidence that cytokine-
mediated immune interactions induced by Schistosoma mansoni alter disease outcome in mice
concurrently infected with Trichuris muris. The Journal of Experimental Medicine 181:769–
774.
Diepolder HM, Zachoval R, Hoffmann RM, Jung MC, Gerlach T, Pape GR. 1996. The role of hepa-
titis C virus specific CD4+ T lymphocytes in acute and chronic hepatitis C. Journal of Molecu-
lar Medicine 74:583–588.
El-Khoby T, Galal N, Fenwick A, Barakat R, El-Hawey A, Nooman Z, Habib M, Abdel Wahab F,
Gabr NS, Hammam HM, Hussein MH, Mikhail NN, Cline BL, Strickland GT. 2000. The epide-
miology of schistosomiasis in Egypt: summary of findings in nine governorates. The American
Journal of Tropical Medicine and Hygiene 62:88–99.
Gerlach T, Diepolder H, Jung M, Gruner N, Schraut W, Zachoval R, Hoffman R, Schirren A,
Santantonio T, Pape G. 1999. Recurrence of hepatitis C virus after loss of virus specific CD4+
T-cell response in acute hepatitis C. Gastroenterology 117:993–941.
Malaria, TB, and HIV/AIDS are important public health problems in sub-
Saharan Africa and some parts of Asia. Both HIV and malaria exert their heaviest
toll in sub-Saharan Africa, where the progression of HIV-related disease is con-
sidered to be most rapid. The interaction between HIV/AIDS and malaria can be
viewed in the mechanistic context, where immunomodulation by one organism
can impact the natural course of infection of the co-existing pathogen, and in
programmatic context, where the treatment for one disease may have beneficial
impact on the other disease and/or the treatment for one disease may not be effec-
tive in the presence of the co-infecting pathogen.
Initial studies of the interactions between HIV and malaria focused on the
ability of malaria parasites to act as opportunistic organisms in immunosuppressed
HIV-positive persons. As recent reviews demonstrate, most of the earlier studies,
conducted primarily in adults, did not show an effect of HIV infection on the
prevalence or severity of malaria (Chandramohan and Greenwood, 1998; Corbett
et al., 2002).
Earlier studies conducted in Zaire, Uganda, Rwanda, and Zambia, showed
no or marginal effect of HIV infection on malaria parasitemia (Simooya et al.,
1988; Chattopadhya et al., 1991; Greenberg, 1992). However, recent studies con-
ducted in Malawi reported increased prevalence rates of malaria parasitemia and
parasite density in HIV-infected pregnant women (Chandramohan and Green-
wood, 1998). The higher prevalence of malaria parasitemia was seen in HIV-
infected women of all gravidities, indicating that the parity-specific immunity to
malaria, which is normally associated with multigravidae, was impaired in HIV-
infected women (Chandramohan and Greenwood, 1998). More importantly, these
studies revealed that infants born to HIV- and malaria-positive mothers were at a
significantly higher risk for low birth weight. Increased prevalence of peripheral
parasitemia and placental malaria has also been seen in HIV-positive pregnant
women in western Kenya, which has higher rates of malaria transmission than
Malawi (Chandramohan and Greenwood, 1998). The increased prevalence of
parasitemia in HIV-positive women seemed to be pregnancy associated, because
parasitemia in HIV-positive women reduced to the level seen in HIV-negative
women 2–6 months postpartum.
HIV infection has been shown to induce poor responses to antimalarial treat-
ment with sulfadoxine-pyrimethamine (S/P) in pregnant women. A recent study
conducted in western Kenya indicated that although a standard two-dose S/P regi-
men worked well in controlling peripheral parasitemia and placental malaria dur-
ing pregnancy in HIV-negative women, it failed to prevent peripheral and placen-
tal parasitemia in HIV-infected women. Poor response to S/P antimalarial
treatment was also reported in pregnant Malawian women with HIV-1 infection.
Because parasitemia was reduced drastically after each treatment and monthly S/
P dosing worked well in both HIV-positive and HIV-negative women, it is pos-
sible that the poor treatment response was due to rapid re-infection rather than
delayed parasite clearance. No difference in quinine treatment failure was seen
It has been suggested that the progression from HIV-1 infection to AIDS is
more rapid in sub-Saharan African patients than in persons living in developed
countries (Gilks, 1993; Mulder et al., 1994; Grant et al., 1997). In addition to the
lack of access to health care and treatment, chronic immune stimulation from
increased exposure to other infectious agents are probable co-factors of immune
activation. Earlier investigations of the relationship between HIV-1 and malaria
focused mainly on the effect of HIV-1 infection on malaria. Only one study ex-
amined the effect of malaria on HIV-1 infection, and failed to detect any effect of
malaria infection on HIV-1 progression. No measurements of changes in CD4+
T-cell counts and viral load, which are two current predictors of HIV disease
progression, were done in this study.
Recent in vitro and in vivo studies, nevertheless, indicate that malaria can
potentially affect the course of HIV infection in several aspects. The initial evi-
dence of a possible effect of malaria on HIV-1 infection came from a retrospec-
tive analysis of data from a cohort study of mothers and infants in rural Malawi. It
was demonstrated that infants born to mothers with both placental malaria and
HIV-1 infection had post-neonatal mortality 4.5 times higher than infants born to
mothers with only placental malaria, and 2.7–7.7 times higher than infants born
to mothers with only HIV-1 infection (Chandramohan and Greenwood, 1998;
Corbett et al., 2002). This increased mortality in infants born to mothers with dual
HIV and malaria was attributed to the increased transmission of HIV from mo-
thers to infants, although no HIV testing was conducted in these infants.
Because immune activation is an important prerequisite for efficient HIV
infection and viral replication, we evaluated the effect of malarial antigen stimu-
lation on HIV-1 infection. Stimulation with soluble malarial antigens or malarial
pigment from P. falciparum enhanced HIV-1 replication in PBMC from naive
donors by 10- to 100-fold. The malarial antigen-upregulated HIV-1 replication
was mediated through induction of TNF-α via the activation of long terminal
repeat (LTR)-directed viral transcription (Xiao et al., 1998). Preliminary studies
conducted with PBMC from HIV-positive individuals residing in western Kenya
indicated that recall immune responses induced by soluble malarial antigens can
increase HIV-1 replication (Xiao et al., unpublished observation). PBMC from 3
of 10 HIV-1 infected individuals showed active in vitro viral production after the
antigen stimulation.
These in vitro observations have been confirmed by the result of a recent
prospective, cohort study of 47 HIV-positive adults with active falciparum ma-
laria and 42 HIV-positive adults without malaria in Malawi. It was shown that
HIV-positive individuals with active malaria had a mean plasma HIV-1 viral load
7-fold higher than HIV-positive individuals without malaria (Hoffman et al.,
1999). Plasma HIV-1 RNA concentrations did not correlate significantly with P.
falciparum parasite density or the duration of fever. However, antimalarial che-
motherapy with S/P resulted in a small (37 percent) but significant reduction in
HIV-1 RNA load by week 4 post-treatment in individuals with HIV and malaria
coinfection.
Another potential interaction between HIV and malaria is at the invasion
stage of both pathogens. HIV-1 has been recently shown to bind erythrocytes
from Caucasian persons through the Duffy antigen receptor for chemokines
(DARC), a receptor that is also used by the invasion of P. vivax merozoites into
reticulocytes (Lachgar et al., 1998). It has been proposed that erythrocytes may
function as a reservoir for HIV-1, and this binding to CD4 (–) cells via DARC by
HIV may be used as a mechanism for the entry of HIV-1 into endothelial cells
and neurons (Lachgar et al., 1998). Because P. falciparum-infected erythrocytes
adhere to brain endothelial cells and cause brain hemorrhage, it is conceivable
that the sequestration of parasitized erythrocytes in the brain with HIV viral par-
ticles attached may facilitate the entry of HIV into neurons in individuals that are
DARC-positive. This may promote the occurrence of neurologic disorders, which
are frequently seen in AIDS patients.
Programmatic concerns for interactions between malaria and HIV/AIDS are
mainly at the level of diagnosis and treatment. Earlier diagnostic studies showed
false positivity of blood samples from malaria-affected individuals during HIV
testing (Biggar et al., 1996). This was probably due to nonspecificity of the early
HIV diagnostic kits, because antigen cross-reactivity between retroviruses and
malaria parasites has been reported (Lal et al., 1994). As far as treatment of un-
complicated and complicated malaria is concerned, blood transfusion for the treat-
ment of severe malarial anemia and presumptive treatment of febrile illness have
emerged as two important problems. Recent studies have shown that many of the
presumed malarial febrile illnesses were actually the result of primary HIV-1
infection (Nwanyanwu et al., 1997). This problem may be more severe in areas
with high prevalence of HIV and malaria, leading to unnecessary use of
antimalarials for the treatment of fever. This overuse of antimalarials may con-
tribute to the rapid emergence of drug resistance. As far as the transfusion-related
transmission of HIV is concerned, earlier studies clearly revealed that use of
unscreened blood for the treatment of severe malarial anemia was a factor in the
transmission of HIV.
While this paper provides an account of published work on the interaction
between malaria and HIV/AIDS, there is compelling evidence of interaction be-
tween other microorganisms and HIV/AIDS. It is likely that the interactions be-
tween several microorganisms present together in an individual may modulate
the pathogenesis and transmission of major infectious agents.
From a mechanistic point of view, however, a common thread that seems to
tie this interaction together is immune activation induced by infectious agents
prevalent in malaria-endemic areas. Therefore, removing risk factors of immune
activation (i.e., co-infectious agents) by effective use of drugs, physical interven-
tions to interrupt transmission, such as bednets for malaria, and other prevention
methods should have the dual effect of reduced risk of rapid progression of HIV-
related disease (by elimination or suppression of viral activating factors) and re-
duced morbidity and mortality by a co-infecting pathogenic organism.
The schematics of our current knowledge and the likely outcomes of the
interaction between HIV and malaria are shown in Figure 2-1. It is very likely
that multiple enteric, respiratory, bloodborne, vectorborne, and waterborne and
foodborne agents may induce immunologic changes (even in asymptomatic in-
fections) that promote infection, transmission, and clinical manifestation of ill-
nesses of the co-infectious pathogens. It is therefore important to capture all mor-
bidity data and conduct extensive diagnostic work in future studies so that the
analysis can be controlled for the effect of different co-infectious agents.
In the context of HIV/AIDS and malaria, the available data should be consid-
ered in:
REFERENCES
Bentwich Z, Maartens G, Torten D, Lal AA, Lal RB. 2000. Concurrent infections and HIV pathogen-
esis. AIDS 14:2071–2081.
Biggar RJ, Miotti PG, Taha TE, Mtimavalye L, Broadhead R, Justesen A, Yellin F, Liomba G, Miley
W, Waters D, Chiphangwi JD, Goedert JJ. 1996. Perinatal intervention trial in Africa: effect of
a birth canal cleansing intervention to prevent HIV transmission. Lancet 347:1647–1650.
Chandramohan D and Greenwood BM. 1998. Is there an interaction between human immunodefi-
ciency virus and Plasmodium falciparum? International Journal of Epidemiology 27:296–301.
Chattopadhya D, Kumari S, Chatterjee R, Verghese T. 1991. Antimalarial antibody in relation to
seroreactivity for HIV infection in sera from blood donors. Journal of Communicable Diseases
23:195–198.
FIGURE 2-1 Current knowledge and likely outcomes of the interaction between HIV and
malaria.
Likely outcome
Mother 1. Increased HIV viral load
2. Increased vertical transmission of HIV
3. Lower birthweight of infants born from dually infected
mothers
Corbett EL, Steketee RW, ter Kuile FO, Latif AS, Kamali A, Hayes RJ. 2002. HIV-1/AIDS and the
control of other infectious diseases in Africa. Lancet 359:2177–2187.
Gilks CF. 1993. The clinical challenge of the HIV epidemic in the developing world. Lancet 342:1037–
1039.
Grant AD, Djomand G, De Cock KM. 1997. Natural history and spectrum of disease in adults with
HIV/AIDS in Africa. AIDS 11:S43–S54.
Greenberg AE. 1992. Pp. 143–148 in AIDS in the World, TW Netter, J Mann, DJM Tarantola, eds.
Boston: Harvard University Press.
Hoffman IF, Jere CS, Taylor TE, Munthali P, Dyer JR, Wirima JJ, Rogerson SJ, Kumwenda N, Eron
JJ, Fiscus SA, Chakraborty H, Taha TE, Cohen MS, Molyneux ME. 1999. The effect of Plasmo-
dium falciparum malaria on HIV-1 RNA blood plasma concentration. AIDS 13:487–494.
Lachgar A, Jaureguiberry G, Le Buenac H, Bizzini B, Zagury JF, Rappaport J, Zagury D. 1998.
Binding of HIV-1 to RBCs involves the Duffy antigen receptors for chemokines (DARC). Bio-
medicine and Pharmacotherapy 52:436–439.
Lal RB, Rudolph D, Alpers MP, Sulzer AJ, Shi YP, Lal AA. 1994. Immunologic cross-reactivity
between structural proteins of human T-cell lymphotropic virus type I and the blood stage of
Plasmodium falciparum. Clinical and Diagnostic Laboratory Immunology 1:5–10.
Migot F, Ouedraogo JB, Diallo J, Zampan H, Dubois B, Scott-Finnigan T, Sanou PT, Deloron P.
1996. Selected P. falciparum specific immune responses are maintained in AIDS adults in
Burkina Faso. Parasite Immunology 18:333–339.
Moore JM, Ayisi J, Nahlen BL, Misore A, Lal AA, Udhayakumar V. 2000. Immunity to placental
malaria. II. Placental antigen-specific cytokine responses are impaired in human immunodefi-
ciency virus-infected women. Journal of Infectious Diseases 182:960–964.
Mulder DW, Nunn AJ, Wagner HU, Kamali A, Kengeya-Kayondo JF. 1994. HIV-1 incidence and
HIV-1-associated mortality in a rural Ugandan population cohort. AIDS 8:87–92.
Nwanyanwu OC, Kumwenda N, Kazembe PN, Jemu S, Ziba C, Nkhoma WC, Redd SC. 1997. Ma-
laria and human immunodeficiency virus infection among male employees of a sugar estate in
Malawi. Transactions of the Royal Society of Tropical Medicine and Hygiene 91:567–569.
Simooya OO, Mwendapole RM, Siziya S, Fleming AF. 1988. Relation between falciparum malaria
and HIV seropositivity in Ndola, Zambia. British Medical Journal 297:30–31.
Wabwire-Mangen F, Shiff CJ, Vlahov D, Kline R, Serwadda D, Sewankambo NK, Mugerwa RD,
Quinn TC. 1989. Immunological effects of HIV-1 infection on the humoral response to malaria
in an African population. The American Journal of Tropical Medicine and Hygiene 41:504–511.
Whittle HC, Brown J, Marsh K, Greenwood BM, Seidelin P, Tighe H, Wedderburn L. 1984. T-cell
control of Epstein-Barr virus-infected B cells is lost during P. falciparum malaria. Nature
312:449–450.
Xiao L, Owen SM, Rudolph DL, Lal RB, Lal AA. 1998. Plasmodium falciparum antigen-induced
human immunodeficiency virus type 1 replication is mediated through induction of tumor ne-
crosis factor-alpha. Journal of Infectious Diseases 177:437–445.
OVERVIEW
Humans exist in complex milieus, and their association with disease is af-
fected both by the environment in which they live and by their genetic suscepti-
bility to particular diseases. People live in concert with microbial agents that may
or may not cause disease in particular individuals, depending on their environ-
ment and their genetics.
There are substantial obstacles to identifying organisms associated with a
particular chronic disease. First, organisms can act in a “hit and run” manner, in
which they cause disease initially but then are either resolved due to natural im-
munity or are successfully eliminated with antibiotics. The damage has been done,
however, resulting in chronic disease. For some chronic diseases of this type,
such as Reiter’s syndrome, Guillain-Barré syndrome, or rheumatic heart disease,
it is very difficult to find a fingerprint of the organism in the disease tissue. Sec-
ond, organisms can be latent at the time of diagnosis. They may not be actively
replicating, so there is no active RNA transcription. Third, chronic latent or recur-
rent infection may be involved in the pathogenesis so that, again, the organism
may not be active at the time of diagnosis. Fourth, organisms may need a particu-
lar predisposing environment or a host with a particular genetic susceptibility, so
the simple presence or absence of the organism may be misleading.
To address these problems, evidence is assembled in a number of ways: from
epidemiological studies, from microbiological assessment of pathogenesis and
etiology, from studies that mimic the disease process in vitro or in animals, and
from clinical treatment trials.
135
Patrick Moore described how the discovery of new pathogens will require
the talents of multiple disciplines, including epidemiology, clinical medicine,
molecular biology, and pathology. Moore used the example of the identification
of the virus that causes Kaposi’s sarcoma, which often strikes gay men, to discuss
general issues in causality and to illustrate the limits of current approaches for
determining causality for a newly discovered agent and disease. The causative
agent, named Kaposi’s sarcoma-associated herpesvirus (KSHV), was identified
using a genetic technique called representational difference analysis. Once the
virus was identified, in 1994, events moved rather quickly—a fact that speaks to
the importance of new pathogen discovery. The virus’s genome has been se-
quenced, serologic tests have been developed, and studies have been initiated to
understand its epidemiology and to test possible treatments. Moreover, the virus
has since been found to cause at least two other types of disease. Based on this
experience, Moore pointed to the need for researchers to move beyond Koch’s
postulates or other traditional guidelines in their efforts to determine disease cau-
sality for suspect microbes. Researchers are attempting to do this by applying
various new techniques emerging from molecular biology and biotechnology. It
seems clear as well that epidemiologists developing new criteria for causality will
have to incorporate new pathogenic mechanisms that are not accounted for in
current disease models.
Mikhail Pletnikov discussed the importance of expanding research to better
understand the interplay of genetic and environmental factors in the causation of
a number of important developmental behavioral disorders. Among methodologi-
cal problems of studying the gene-environment interplay is the difficulty in firmly
defining environmental factors and making them quantifiable. In this context,
virus infections provide a promising research avenue, because of their etiologic
connection to several neurodevelopmental disorders, including autism and schizo-
phrenia, and because of the reliability of quantification of viral effects on brain
and behavior. In particular, Pletnikov described work using an animal model to
study gene-environmental interactions that occur during neonatal infection with
Borna disease virus (BDV). Neonatal BDV infection in rats has been shown to
produce distinct neuroanatomical, neurochemical, and behavioral abnormalities
that resemble pathological and clinical features of some human developmental
disorders. The significance of studying neonatal exposure derives from the fact
that the effects of many genetic and environmental risk factors are evident either
prior to or around the time of birth, and the interaction between them often is
apparent well before the onset or diagnosis of the chronic disease condition. Thus,
studying the effects of neonatal BDV infection across the entire postnatal period
in genetically different strains of rats will aid in understanding the course and
time-dependent character of the interaction of genetic background features and
the virus infection. In this way, the model system may allow study of some tre-
mendously complex mechanisms relevant to developmental disorders.
David Persing provided an overview of recent research in the area of infec-
tion, cancer, and the immune response. Current evidence suggests that inherited
predisposition to cancer probably accounts for only a subset of total cancer pa-
tients, and in most models of the development of neoplasia, an underlying as-
sumption is the contribution of an array of intrinsic and extrinsic factors within a
multistep process. A basic prerequisite of many models is an increase in the
baseline proliferation rates of essentially normal cell populations that leads to
dysregulation of normal growth control mechanisms. Since many infectious pro-
cesses often lead, directly or indirectly, to increased cell turnover and prolifera-
tion, certain agents are now widely regarded as carcinogens. Some of the patho-
gens that have been linked to cancer include human papillomaviruses (cervical
cancer and other skin cancers), human T-cell leukemia viruses (adult T-cell leu-
kemias and lymphomas in endemic areas), hepatitis B virus (liver cancer),
Epstein-Barr virus (Burkitt’s lymphoma and nasopharyngeal carcinoma), and
Helicobacter pylori infection (gastric carcinoma and MALT lymphoma).In addi-
tion, new disease associations are being made with respect to previously known
pathogens, such as the association of chronic hepatitis C virus infection with non-
Hodgkin’s lymphoma in certain populations.
In a separate presentation, Persing described recent and continuing advances
in the development and application of techniques for identifying pathogens that
cause chronic diseases. Although a paper on these subjects does not appear in this
chapter, the following paragraph notes the highlights.
The ability to detect and manipulate nucleic acid molecules in microorgan-
isms has created a powerful means for identifying previously unknown microbial
pathogens and for studying the host-pathogen relationship. Although a paper on
these subjects does not appear in the ensuing text, the highlights of his presenta-
tion are discussed here. Among the new technologies that Persing described is
broad-range polymerase chain reaction, which has proved instrumental in linking
a growing number of pathogens with chronic diseases, and representational dif-
ference analysis, which is an efficient means for finding differences between com-
plex genomes and for identifying specific DNA sequences from the genomes of
unknown pathogens. Researchers also are making use of sophisticated new DNA
microarrays and biosensors that, among other things, can monitor host response
as an indicator of the presence of infection or inflammation. In addition, new
methods for generating “libraries” of genetic information from very small
amounts of material are making it easier to conduct very specific and sensitive
serologic tests. Equipped with these and other advanced tools, researchers are
becoming better able to move beyond the limitations of Koch’s postulates and to
link infectious agents with chronic diseases more precisely and with greater con-
fidence than ever before.
In the ensuing discussions, participants began to sketch in some of the char-
acteristics of a comprehensive and coordinated effort that would enhance efforts
both to identify links between infectious microorganisms and chronic diseases
and to develop and implement interventions to minimize their health conse-
quences. The goal was not to set specific priorities, but to identify opportunities.
Highlighting a selection of the traits identified may provide a glimpse of the
overall picture envisioned.
Participants agreed, for example, on the need to develop standardized defini-
tions of infections and disease, to enable comparisons across studies and conclu-
sions about causality, and on the need to ensure that laboratory assays maintain
universally high standards of specificity, sensitivity, and reproducibility. New
laboratory technology also is needed that can meet such performance standards
while handling high throughput rates, in order to handle analyses of large cohorts
in a reasonable amount of time, a trait that likely will be required in many future
projects. Comparable efforts are needed to ensure that epidemiological studies
are conducted with vigor and in an appropriate manner. One step will involve
linking of databases that are designed (or modified) to be compatible. Peer review
journals can reinforce performance standards if publication depends on the use of
sound laboratory assays and epidemiologic design capable of supporting the con-
clusions.
Continued studies are needed to define temporal relationships between infec-
tions and disease—that is, what stage of infection determines outcome (e.g., first
infection, reinfection, persistent infection, coinfection, or subsequent cross-react-
ing infection). Studies also are needed to clarify at which stage infection must be
prevented or treated in order to minimize or eliminate chronic sequelae. It will be
important to determine the expected benefit of actions, to ensure that the benefits
will outweigh any possible risks. In other words, intervention should decrease
chronic disease burden without unduly endangering the people who receive care.
There is a need to better understand the natural history, especially the earliest
stages, of chronic diseases of unknown or incompletely known origins. What
makes this task especially important is the hit-and-run nature of some diseases in
which microbes set adverse events in motion and then disappear, the increased
difficulty of imputing causation to microbes detected late in the course of disease,
and the increased ease of treating or preventing disease at early time points. To-
ward this aim, clinicians should be increasingly encouraged to identify patients
who have recently developed or seem to be developing various suspect chronic
diseases, to collect in an orderly manner a range of clinical specimens, and then to
follow the course of the disease in order to identify tell-tale early clinical features.
Calls were made for more effort devoted to developing animal models of
chronic diseases, and to teaching health professionals about their value and their
limitations. Animal models can be powerful tools when the etiology or pathogen-
esis of a disorder is unknown. Psychiatric modeling with animals may present an
especially ripe area for probing a variety of important questions, yet many practi-
tioners in the field are not accustomed to working with such models.
Increased emphasis should be placed on longitudinal studies, as well as on
follow-up studies and “look back” studies of cohorts and surveillance results that
have been generated in the past. Longitudinal studies may prove particularly valu-
able given that rapid advances in the field may mean that we might not know
today which pieces of evidence will be needed in the future. Human specimen
collections, such as the National Children’s Study that will begin in 2004, may be
especially important for longitudinal research.
Participants identified a number of specific populations that should receive
additional attention. One such group includes people who move from rural areas
into cities, both in the developing and the developed world. Studies are needed to
see whether they bring new infections with them, or whether they prove to be
susceptible to new infections that they previously had not encountered. With the
world’s changing demographics, gathering such information may provide a win-
dow into pathogenesis of a number of chronic diseases.
Efforts are needed to address problems related to informed consent. Many
workshop participants expressed concern that current regulations and guidelines
are too complex, too uncertain, or too restrictive to allow for meaningful sharing
of data—and sometimes all three. There was general agreement that informed
consent is and must remain an important part of research involving human sub-
jects. But participants also agreed that all parties—from government, academia,
and private funding agencies—need to work together to develop a more standard-
ized method for gaining patient consent, for gathering identifying information,
and for being able to use this identifying information in the future. This may be an
opportunity for multiple institutions and multiple governments, domestic and for-
eign, to cooperate in devising a system of patient consent that operates more
smoothly, protects patient rights, and allows for expanded research on infections
and chronic diseases.
Given the magnitude of the outstanding scientific questions, and of the health
consequences at stake, an increasing share of future research likely will involve
groups of investigators representing a variety of disciplines, or groups of institu-
tions working collaboratively. Although there remains a clear role for individual
investigators, it is becoming apparent that large multidisciplinary projects often
can best marshal the critical mass needed to address the thorniest biological prob-
lems. In many cases, these large projects will include a multinational component,
in order to ensure that sufficient attention is paid to multiracial, multiethnic, and
multicultural differences.
Participants called on the overall scientific community to evaluate whether it
is organized and structured properly to address these issues, and whether its vari-
ous components communicate effectively. The community also should mount a
concerted effort to identify gaps in current knowledge about the etiology of
chronic diseases, pinpoint what needs to be done to close those gaps, chart the
obstacles that stand in the way, and then identify and provide the necessary finan-
cial resources (monetary and human) to drive progress.
Government can play an important role by reorienting its funding priorities.
Indeed, the time is ripe. The government is now investing nearly $1 billion in
rebuilding the nation’s public health system, and part of the money will go to-
ward linking state health departments more closely with local health departments
than has historically been the case. At the same time, government research cen-
ters are launching major new interdisciplinary projects, and universities, which
often have been in competition with one another, are beginning to join in collabo-
rations. Thus, foundations are beginning to be built for bridges linking public
health, clinical medicine, and research. But these promising efforts need to be
nurtured to ensure continued cooperation.
FIGURE 3-1 Representational differential analysis comparing DNA from a Kaposi’s sarcoma lesion to sterile-site tissue from the same patient.
141
The Infectious Etiology of Chronic Diseases: Defining the Relationship, Enhancing the Research, and Mitigating the Effec
The Infectious Etiology of Chronic Diseases: Defining the Relationship, Enhancing the Research, and Mitigating the Effec
http://www.nap.edu/catalog/11026.html
rehybridized again to the adapter-less healthy tissue DNA and the process is re-
peated, each time selectively enriching for the unique sequences found only in the
KS lesion (Gao and Moore, 1996).
Four RDA fragments were generated by this process, two of which were
found to be specific for the KS agent. Although these two fragments account for
less than 1 percent of the entire 145-kilobase viral genome, the few base-pairs
worth of unique information they provided made it possible to develop enough
tools to identify the agent.
The two fragments were used as Southern hybridization probes and tested
against KS lesions, showing that about three-quarters of the KS lesions were
positive for viral DNA. Using internal specific primers from the KS 330 band, a
PCR assay was developed that showed 25 out of 27, or 93 percent, of the initial
KS lesions tested positive. Moreover, the negative samples were equally telling:
one of the two negatives had degraded DNA and was not amplifiable by using
cellular primers, and the other one was mislabeled normal human kidney.
KSHV is a gamma herpesvirus belonging to the same class as Epstein-Barr
virus (EBV). It is associated with three different major proliferative diseases:
Kaposi’s sarcoma, primary effusion lymphoma (PEL) (Cesarman et al., 1995a), a
monoclonal B cell lymphoma, and multicentric Castleman’s disease (Soulier et
al., 1995), which is a polyclonal hyperplasia caused by a virus-encoded cytokine
expressed by KSHV (Parravicini et al., 1997). Nearly all KS and PEL patients
have KSHV infection, but only about half of HIV-negative, multicentric
Castleman’s disease patients are positive for KSHV infection indicating that this
disease has a heterogeneous pathogenesis.
The two aforementioned RDA fragments of the KSHV genome facilitated
the identification of infected cell lines to serve as source material for viral DNA
and as a reagent for biologic studies (Cesarman et al., 1995b). Genomic library
walking was performed using cosmid and lambda libraries from one of these cell
lines allowing sequencing of the remainder of the genome (Russo et al., 1996).
Using this information, various techniques were used to identify likely antigens
and generate serologic tests (Gao et al., 1996a,b; Kedes et al., 1996; Simpson et
al., 1996). While identification of high-titered infected cell lines sped up this
process, isolating the agent was not essential for developing tools to detect it.
Molecular biology has reached the point where it is straightforward to identify a
new agent, sequence its genome and develop serologic tests for it without ever
having actually purified, living sample of the agent. The virus does not have to be
grown in order to apply traditional techniques for determining whether or not an
agent is present.
The virus itself is a tremendously interesting scientific problem. It has a long
unique coding region containing all of the viral open reading frames. Unlike most
viruses, KSHV has pirated cellular genes over its evolution and the viral genes
are recognizable homologues to cellular genes of known function. Many of these
genes provide new insights into tumor virology through their control of the cell
cycle, prevention of apoptosis, or immune evasion properties.
One might conclude that this virus is completely different from other viruses
and not much can be learned from it to extend to other viruses. In fact, the oppo-
site is true. EBV, for example, induces cellular cyclin D2 to drive the cell through
the G1/S cell cycle checkpoint; KSHV encodes its own version of a cyclin D with
an analogous function. Other examples of functional correspondence between the
KSHV homologues and viral genes encoded by even distantly related viruses can
be readily seen (Moore and Chang, 1998a, 2001). For this reason KSHV might be
considered something like a molecular Rosetta stone because by using it, we can
begin to interpret the language of molecular virology in terms of cell biology for
many different viruses.
The importance of new pathogen discovery is illustrated by a timeline of KS
research. This would be equally true also for hepatocellular carcinoma and hepa-
titis C or a wide range of diseases where a new pathogen has been found. The
point is that things change quite rapidly once the agent is finally found. Moritz
Kaposi initially described the disorder in 1873, but not until 70 years later was
there a suggestion of an infectious etiology. In 1981, the onset of the AIDS epi-
demic brought a tremendous increase in scientific interest in this cancer. There
was still, however, little known about the pathogenesis of this disorder in 1993
when there were over 200,000 cases of AIDS in the United States. At that time
over 20 different agents had been proposed at one time or another as the causal
agent for Kaposi’s sarcoma.
The description of KSHV was first published in 1994 and within two years
its viral genome was completely sequenced (Neipel et al., 1997; Russo et al.,
1996). By that time it was known that the virus was found in all forms of Kaposi’s
sarcoma (Boshoff et al., 1995; Chang et al., 1996; Moore and Chang, 1995),
serologic tests had been developed (Gao et al., 1996a,b; Kedes et al., 1996; Miller
et al., 1996; Simpson et al., 1996) and studies initiated to understand the epidemi-
ology of this virus in KS (Moore et al., 1996; Whitby et al., 1995). Shortly there-
after, studies were performed to see whether ganciclovir, a specific antiviral agent,
could be used to treat KS (Martin et al., 1999). At the present, there have been
over 2,000 papers published on KSHV and its role in malignancy.
Finding a new pathogen also can benefit other fields. When KSHV was first
described, only two other related rhadinoviruses had been described in new world
primates. Although we live in North America, humans are still considered old
world apes. One was herpesvirus saimiri from squirrel monkeys and the other
herpesvirus ateles from spider monkeys. Researchers at the University of Wash-
ington began to look for other primate KSHV-like viruses (Rose et al., 1997).
Using consensus PCR, two were found in rhesus macaques, and subsequently in
all the various branches of the primates, both lower and higher primates. This
suggests that the viral ancestor of KSHV evolved with us over time. Even more
These were postulates that Koch developed for determining the cause of tu-
berculosis at a time when not much was known of viruses or the carrier state. This
was a brilliant attempt to develop a scientific rationale for determining whether
an agent is causal for disease or not.
Bradford Hill also developed epidemiologic criteria for causality which are
shown here for KSHV and KS (Hill, 1965). Though developed specifically for
cigarette smoking, most epidemiologists now use these criteria to determine
causality:
• Is the infection present in cases; do all types of the disease involve infec-
tion? Is it reproducible in multiple settings?
• Does infection precede disease?
• Is the infection specific to the disease or is it ubiquitous infection among
humans?
• Is the virus localized to the tumor (one interpretation of a biologic gradient)?
• Do the epidemiologic studies make sense (are they coherent?)?
• Is it biologically reasonable and do experiments confirm the relationship?
With regard to KSHV, the answers to these questions are largely true. KSHV
is present in more than 95 percent of KS lesions. It can be said that the remaining
negative 5 percent is probably spurious due to technical difficulties in detection
or misdiagnosis, and in fact the virus is absolutely necessary for disease. Though
this cannot be proven at present, it can be argued that the situation is very similar
to that of papillomavirus and cervical cancer 5 years ago. Is it generalizable? Yes.
All types of KS are infected as far as is known. It also appears to have the correct
temporal association in that cohort studies show that patients are infected before
developing disease, and not afterwards.
But specificity is an important question. KSHV is not singly associated with
Kaposi’s sarcoma. It is also associated with two other diseases. However, the
epidemiology of these two diseases makes some sense in terms of Kaposi’s sar-
coma, so multiple outcomes are not too worrisome. Depending on the assay that
is used, some researchers suggest that the infection rate in the general population
for this virus is much higher than alluded to here, but careful studies suggest that
less that 5 percent of Americans are infected with KSHV.
Is there a biologic gradient? Yes, there is. Are the epidemiologic findings
coherent? Yes, a wide range of epidemiologic studies seem to come to exactly the
same set of conclusions. Is it biologically plausible? Yes, there are multiple
oncogenes in this virus, related viruses cause cancers, and there are blinded clini-
cal trials which seem to suggest that treatment with ganciclovir prevents the de-
velopment of Kaposi’s sarcoma.
KSHV and KS was a relatively easy case even though it took two years and
seven or eight different studies before these conclusions could be reached. None-
theless, the case for causality was relatively easy.
Now let’s consider issues where causality is more problematic. First, KSHV
has been claimed not only to cause Kaposi’s sarcoma, but also a wide variety of
diseases that don’t fit its epidemiologic pattern, such as multiple myeloma and
sarcoidosis. Although studies supporting these associations were published in
reputable journals, they were based on PCR or had other problems and remain
questionable in terms of contemporary epidemiological knowledge. In the age of
PCR, it is difficult for the casual observer to sort out what is true and what is not.
Assuming that the problem of poor laboratory technique can be solved, there
are three more fundamental problems in determining causality. First, causality is
relative and should not be thought of as being cast in stone. Causality depends on
pathogenic assumptions. That is where Koch’s postulates fall down and also
where Hill’s criteria fall down as well.
For example, if a virus is associated with autoimmune disorders, it can be
assumed that one would have an immune response against that virus. In that case
the individual may actually clear the virus, so a reverse association would be seen
from what would normally be expected following Hill’s criteria. The criteria sim-
ply do not apply in this case, even though it is a reasonable possibility.
Second, causality is normative. Researchers can get together and study the
data but only a few agree to particular conclusions. When the studies describing
KSHV as the cause of KS were completed, it was thought that the issue of causal-
ity would be resolved. However, it still required a great deal of interpretation.
There were many contradictory studies that were ignored because they were not
considered valid. But others might disagree, and this is true for just about any
contentious issue. An agent is only considered causal for a disease when a major-
sality argument becomes circular even though we now have good reasons for
splitting up a disease manifestation into different diseases with different manifes-
tations.
It is likely that in the future, improved knowledge of pathogenic mechanisms
will reveal novel causal relationships. For example, not too long ago the idea that
a bacteria could cause stomach ulcers would have been considered laughable.
Helicobacter pylori and peptic ulcer disease had a pathogenic mechanism that
was poorly understood and thus there was no framework to gauge whether or not
a bacteria was the possible cause. In fact, pathologists had seen bacteria associ-
ated with ulcers for decades but didn’t remark on them because there was no way
to measure their significance.
New ways of determining causality that go beyond Hill’s criteria and Koch’s
postulates need to be developed if new and complex mechanisms for disease are
to be understood. Researchers have attempted to do this by taking into consider-
ation new techniques of molecular biology (Fredericks and Relman, 1996). It
seems clear that epidemiologists developing new criteria for causality will have
to incorporate new pathogenic mechanisms that are not currently accounted for.
Unfortunately, no one can predict what new pathogenic mechanisms will be
discovered and therefore there are no universal criteria for causality that will not
need future revisions. In the end, it cannot be absolutely proved that an agent
causes disease, only that it does not. Instead, while criteria such as Hill’s or Koch’s
postulates are enormously helpful in guiding our thinking, we should not be con-
strained by them as has happened in cases like EBV and nasopharyngeal carci-
noma. In this case, both science and public health have suffered from rigid adher-
ence to abstract criteria. For cases where established criteria break down, all that
can be done is to develop a detailed pathogenic model which can be tested using
epidemiologic studies and further modified. In essence, to use the scientific
method which is employed by scientists every day.
REFERENCES
Beral V, Peterman TA, Berkelman RL, Jaffe HW. 1990. Kaposi’s sarcoma among persons with AIDS:
a sexually transmitted infection? Lancet 335:123–128.
Boshoff C, Whitby D, Hatziioannou T, Fisher C, van der Walt J, Hatzakis A, Weiss R, Schulz T.
1995. Kaposi’s sarcoma-associated herpesvirus in HIV-negative Kaposi’s sarcoma. Lancet
345:1043–1044
Cesarman E, Chang Y, Moore PS, Said JW, Knowles DM. 1995a. Kaposi’s sarcoma-associated her-
pesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomas. New England
Journal of Medicine 332:1186–1191.
Cesarman E, Moore PS, Rao PH, Inghirami G, Knowles DM, Chang Y. 1995b. In vitro establishment
and characterization of two acquired immunodeficiency syndrome-related lymphoma cell lines
(BC-1 and BC-2) containing Kaposi’s sarcoma-associated herpesvirus-like (KSHV) DNA se-
quences. Blood 86:2708–2714.
Chang Y, Cesarman E, Pessin MS, Lee F, Culpepper J, Knowles DM, Moore PS. 1994. Identification
of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 265:1865–
1869.
Chang Y, Ziegler JL, Wabinga H, Katongole-Mbidde E, Boshoff C, Schulz T, Whitby D, Maddalena
D, Jaffe HW, Weiss RA, Moore PS. 1996. Kaposi’s sarcoma-associated herpesvirus and
Kaposi’s sarcoma in Africa. Archives of Internal Medicine 156:202–204.
Desrosiers RC, Sasseville VG, Czajak SC, Zhang X, Mansfield KG, Kaur A, Johnson RP, Lackner
AA, Jung JU. 1997. A herpesvirus of rhesus monkeys related to the human Kaposi sarcoma-
associated herpesvirus. Journal of Virology 71:9764–9769.
Fredericks DN and Relman DA. 1996. Sequence-based identification of microbial pathogens: A re-
consideration of Koch’s postulates. Clinical Microbiology Reviews 9:18–33.
Gao SJ and Moore PS. 1996. Molecular approaches to the identification of unculturable infectious
agents. Emerging Infectious Diseases 2:159–167.
Gao SJ, Kingsley L, Hoover DR, Spira TJ, Rinaldo CR, Saah A, Phair J, Detels R, Parry P, Chang Y,
Moore PS. 1996a. Seroconversion to antibodies against Kaposi’s sarcoma-associated herpesvi-
rus-related latent nuclear antigens before the development of Kaposi’s sarcoma. New England
Journal of Medicine 335:233–241.
Gao SJ, Kingsley L, Li M, Zheng W, Parravicini C, Ziegler J, Newton R, Rinaldo CR, Saah A, Phair
J, Detels R, Chang Y, Moore PS. 1996b. KSHV antibodies among Americans, Italians and
Ugandans with and without Kaposi’s sarcoma. Nature Medicine 2:925–928.
Greensill J, Sheldon JA, Murthy KK, Bessonette JS, Beer BE, Schulz TF. 2000. A chimpanzee
rhadinovirus sequence related to Kaposi’s sarcoma-associated herpesvirus/human herpesvirus
8: increased detection after HIV-1 infection in the absence of disease. AIDS 14:F129–135.
Hill AB. 1965. Environment and disease: association or causation? Proceedings of the Royal Society
of Medicine 58:295–300.
Kedes DH, Operskalski E, Busch M, Kohn R, Flood J, Ganem D. 1996. The seroepidemiology of
human herpesvirus 8 (Kaposi’s sarcoma-associated herpesvirus): distribution of infection in KS
risk groups and evidence for sexual transmission. Nature Medicine 2:918–924.
Koch R. 1942. The aetiology of tuberculosis (translation of Die Aetiologie der Tuberculose [1882]).
Pp. 392–406 in Source Book of Medical History, DH Clark, ed. New York: Dover Publications,
Inc.
Lacoste V, Mauclere P, Dubreuil G, Lewis J, Georges-Courbot MC, Gessain A. 2000. KSHV-like
herpesviruses in chimps and gorillas. Nature 407:151–152.
Lacoste V, Mauclere P, Dubreuil G, Lewis J, Georges-Courbot MC, Gessain A. 2001. A novel gamma
2-herpesvirus of the Rhadinovirus 2 lineage in chimpanzees. Genome Research 11:1511–1519.
Lisitsyn NA, Rosenberg MV, Launer GA, Wagner LL, Potapov VK, Kolesnik TB, Sverdlov ED.
1993. A method for isolation of sequences missing in one of two related genomes.
Molekuliarnaia Genetika, Mikrobiologiia, i Virusologiia 3:26–9
Martin DF, Kuppermann BD, Wolitz RA, Palestine AG, Li H, Robinson CA. 1999. Oral ganciclovir
for patients with cytomegalovirus retinitis treated with a ganciclovir implant. New England
Journal of Medicine 340:1063–1070.
Miller G, Rigsby MO, Heston L, Grogan E, Sun R, Metroka C, Levy JA, Gao SJ, Chang Y, Moore P.
1996. Antibodies to butyrate-inducible antigens of Kaposi’s sarcoma-associated herpesvirus in
patients with HIV-1 infection. New England Journal of Medicine 334:1292–1297.
Moore PS and Chang Y. 1995. Detection of herpesvirus-like DNA sequences in Kaposi’s sarcoma
lesions from persons with and without HIV infection. New England Journal of Medicine
332:1181–1185.
Moore PS and Chang Y. 1998a. Antiviral activity of tumor-suppressor pathways: clues from molecu-
lar piracy by KSHV. Trends in Genetics 14:144–150.
Moore PS and Chang Y. 1998b. The discovery of KSHV (HHV 8). Epstein-Barr Virus Report 5:1–3.
Moore PS and Chang Y. 2001. Kaposi’s sarcoma-associated herpesvirus. Pp. 2803–2833 in Fields
Virology, DM Knipe and P Howley, eds. Philadelphia: Lippincott, Williams & Wilkins.
Moore PS, Kingsley LA, Holmberg SD, Spira T, Gupta P, Hoover DR, Parry JP, Conley LJ, Jaffe
HW, Chang Y. 1996. Kaposi’s sarcoma-associated herpesvirus infection prior to onset of
Kaposi’s sarcoma. AIDS 10:175–180.
Neipel F, Albrecht JC, Fleckenstein B. 1997. Cell-homologous genes in the Kaposi’s sarcoma-asso-
ciated rhadinovirus human herpesvirus 8: determinants of its pathogenicity?. [Review]. Journal
of Virology 71:4187–4192.
O’Brien TR, Kedes D, Ganem D, Macrae DR, Rosenberg PS, Molden J, Goedert JJ. 1999. Evidence
for concurrent epidemics of human herpesvirus 8 and human immunodeficiency virus type 1 in
US homosexual men: rates, risk factors, and relationship to Kaposi’s sarcoma. Journal of Infec-
tious Diseases 180:1010–1017.
Parravinci C, Corbellino M, Paulli M, Magrini U, Lazzarino M, Moore PS, Chang Y. 1997. Expres-
sion of a virus-derived cytokine, KSHV vIL-6, in HIV-seronegative Castleman’s disease. Ameri-
can Journal of Pathology 151:1517–1522.
Raab-Traub N and Flynn K. 1986. The structure of the termini of the Epstein-Barr virus as a marker
of clonal cellular proliferation. Cell 47:883–889.
Rose TM, Strand KB, Schultz ER, Schaefer G, Rankin GW Jr, Thouless ME, Tsai CC, Bosch ML.
1997. Identification of two homologs of the Kaposi’s sarcoma-associated herpesvirus (human
herpesvirus 8) in retroperitoneal fibromatosis of different macaque species. Journal of Virology
71:4138–4144.
Russo JJ, Bohenzky RA, Chien MC, Chen J, Yan M, Maddalena D, Parry JP, Peruzzi D, Edelman IS,
Chang Y, Moore PS. 1996. Nucleotide sequence of the Kaposi sarcoma-associated herpesvirus
(HHV8). Proceedings of the National Academy of Sciences 93:14862–14867.
Simpson GR, Schulz TF, Whitby D, Cook PM, Boshoff C, Rainbow L, Howard MR, Gao SJ,
Bohenzky RA, Simmonds P, Lee C, de Ruiter A, Hatzakis A, Tedder RS, Weller IV, Weiss RA,
Moore PS. 1996. Prevalence of Kaposi’s sarcoma associated herpesvirus infection measured by
antibodies to recombinant capsid protein and latent immunofluorescence antigen. Lancet
348:1133–1138.
Soulier J, Grollet L, Oksenhendler E, Cacoub P, Cazals-Hatem D, Babinet P, d’Agay MF, Clauvel JP,
Raphael M, Degos L. 1995. Kaposi’s sarcoma-associated herpesvirus-like DNA sequences in
multicentric Castleman’s disease. Blood 86:1276–1280.
Whitby D, Howard MR, Tenant-Flowers M, Brink NS, Copas A, Boshoff C, Hatzioannou T, Suggett
FE, Aldam DM, Denton AS, et al. 1995. Detection of Kaposi’s sarcoma-associated herpesvirus
(KSHV) in peripheral blood of HIV-infected individuals predicts progression to Kaposi’s sar-
coma. Lancet 364:799–802.
Data from family, twin, and adoption studies convincingly show evidence of
a substantial genetic contribution to most neurodevelopmental disorders in hu-
mans. Moreover, recent improvements in molecular and genetic technologies have
resulted in the implication of genes at several chromosomal loci and a search for
candidate genes continues. However, multiple examples of deviation of complex
developmental disorders from clear-cut Mendelian transmission cannot be fully
explained by incomplete penetrance, variable expressivity, or polygenic etiology.
A growing body of evidence suggests an important role of environmental factors
in the causation of some developmental behavioral disorders. Unfortunately, en-
vironmental studies have been carried out with the same concept in mind, i.e., a
search for relevant risk factors that would be self-sufficient to explain the patho-
genesis of human conditions. Very little, if any, theoretical or methodological
interaction between genetic linkage analysis and environmental (e.g., toxicology
or teratology) studies has been undertaken. However, it is the gene-environment
interaction that determines variable disease outcomes and responses to treatment
and must be addressed in future research approaches.
Although it is clear that there are critical interactions between genes and
environment to produce disease phenotypes, this concept has not been a focus of
extensive consideration that the important role of environmental factors becomes
more apparent in the setting of interaction with genetic determinants. Separating
the search for genetic determinants vs. environmental disease etiologies was, in
part, based on the assumption that genes and environmental sources are mainly
additive in their effects, with the outcome reflecting the sum of their influences.
However, the evidence is now clear that genes and environment are interactive as
well, and several important issues of the gene-environment interaction are illus-
trated here with data obtained on the animal model of neurodevelopmental dam-
age in rats neonatally infected with an experimental teratogen, Borna disease
virus (BDV).
Among methodological problems of studying the gene-environment inter-
play is the difficulty in firmly defining environmental factors and making them
*This work was supported by the National Institutes of Health, grant 2RO1 MH 48948-08A1.
the gene-environmental interaction are discussed and illustrated with the data
from the analysis of effects of different genetic background on neurodevelop-
mental damage and responses to treatment in two rat strains following the neona-
tal BDV infection.
REFERENCES
Bautista JR, Schwartz GJ, de la Torre JC, Moran TH, Carbone KM. 1994. Early and persistent abnor-
malities in rats with neonatally acquired Borna disease virus infection. Brain Research Bulletin
34:31–40.
Bautista JR, Rubin SA, Moran TH, Schwartz GJ, Carbone KM. 1995. Developmental injury to the
cerebellum following perinatal Borna disease virus infection. Brain Research. Developmental
Brain Research 90:45–53.
Briese T, Schneemann A, Lewis AJ, Park YS, Kim S, Ludwig H, Lipkin WI. 1994. Genomic organi-
zation of Borna disease virus. Proceedings of the National Academy of Sciences 91:4362–4366.
Carbone K and Pletnikov M. 2000. Borna again, starting from the beginning. Molecular Psychiatry
5:577.
Carbone KM, Park SW, Rubin SA, Waltrip RW II, Vogelsang GB. 1991. Borna disease: association
with a maturation defect in the cellular immune response. Journal of Virology 65:6154–6164.
Cubitt B, Oldstone M, de la Torre JC. 1994. Sequence and genome organization of Borna disease
virus. Journal of Virology 68:1382–1396.
Dittrich W, Bode L, Kao M, Schneider K. 1989. Learning deficiencies in Borna disease virus-infected
but clinically healthy rats. Biological Psychiatry 26:818–828.
Eisenman LM, Brothers R, Tran MH, Kean RB, Dickson GM, Dietzschold B, Hooper DC. 1999.
Neonatal Borna disease virus infection in the rat causes a loss of Purkinje cells in the cerebel-
lum. Journal of Neurovirology 5:181–189.
Gonzalez-Dunia DM, Watanabe S, Syan M, Mallory E, de la Torre JC. 2000. Synaptic pathology in
Borna disease virus persistent infection. Journal of Virology 74:3341–3448.
Herzog S, Frese K, Rott R. 1991. Studies on the genetic control of resistance of black hooded rats to
Borna disease. Journal of General Virology 72:535–540.
Hirano N, Kao M, Ludwig H. 1983. Persistent, tolerant or subacute infection in Borna disease virus-
infected rats. Journal of General Virology 64:1521–1530.
Hornig M, Weissenbock H, Horscroft N, Lipkin WI. 1999. An infection-based model of
neurodevelopmental damage. Proceedings of the National Academy of Sciences 96:12102–
12107.
Johnson RT. 1998. Viral infections of the nervous system. Philadelphia: Lippincott-Raven.
Narayan OS, Herzog K, Frese H, Rott R. 1983. Behavioral disease in rats caused by immunopatho-
logical responses to persistent Borna virus in the brain. Science 220:1401–1403.
Pletnikov MV, Rubin SA, Vasudevan K, Moran TH, Carbone KM. 1999. Developmental brain injury
associated with abnormal play behavior in neonatally Borna Disease Virus (BDV)-infected
Lewis rats: A model of autism. Behavorial Brain Research 100:30–45.
Pletnikov MV, Rubin SA, Schwartz GJ, Carbone KM, Moran TH. 2000. Effects of neonatal rat Borna
disease virus (BDV) infection on the postnatal development of monoaminergic brain systems.
Brain Research. Developmental Brain Research 119:179–185.
Pletnikov MV, Rubin SA, Carbone KM, Moran TH, Schwartz GJ. 2001. Neonatal Borna disease virus
infection (BDV)-induced damage to the cerebellum is associated with sensorimotor deficits in
developing Lewis rats infection on the postnatal development of monoaminergic brain systems.
Brain Research. Developmental Brain Research 126:1–12.
Pletnikov MV, Rubin SA, Vogel MW, Moran TH, Carbone KM. 2002a. Effects of genetic back-
ground on neonatal Borna disease virus infection-induced neurodevelopmental damage. I. Brain
pathology and behavioral deficits. Brain Research 944:97–107.
Pletnikov MV, Rubin SA, Vogel MW, Moran TH, Carbone KM. 2002b. Effects of genetic back-
ground on neonatal Borna disease virus infection-induced neurodevelopmental damage. II. Neu-
rochemical alterations and responses to pharmacological treatments. Brain Research 944:108–
123.
Sprankel H, Richard K, Ludwig H, Rott R. 1978. Behavior alterations in tree shrews (Tupaia glis,
Diard 1820) induced by Borna disease virus. Medical Microbiology and Immunology 26:1–18.
Weissenbock H, Hornig M, Hickey WF, Lipkin WI. 2000. Microglia activation and neuronal apoptosis
in bornavirus infected neonatal Lewis rats. Brain Pathology 10:260–272.
During the past decade, the scientific community has witnessed a virtual
explosion of information regarding the genetic basis of disease, especially of in-
herited disorders and human cancers. Much of the effort of the Human Genome
Initiative has been focused on genetic abnormalities that arise during the develop-
ment of neoplasia and upon congenital predispositions to cancer that are associ-
ated with the inheritance of mutations within tumor suppressor genes and other
loci. These studies are of critical importance to our understanding of genetic and
cellular processes contributing to neoplasia. However, to date the evidence sug-
gests that inherited predisposition to cancer probably accounts for only a subset
of total cancer patients and appears to be insufficient to explain the sporadic cases
currently comprising the majority.
In most models of the development of neoplasia, an underlying assumption
is the contribution of an array of intrinsic and extrinsic factors within a multi-step
process. A basic prerequisite of many models is an increase in the baseline prolif-
eration rates of essentially normal cell populations, accompanied by genotypic
and phenotypic alterations leading to dysregulation of normal growth control
mechanisms. Accordingly, preneoplastic conditions are often associated with an
increase in the proliferation of tissues, and some cancer predisposing conditions
result from inherited predispositions toward increased mitotic rate (e.g., familial
polyposis). However, virtually any condition leading to increased cellular prolif-
eration, whether by a direct or indirect mechanism, might potentiate the develop-
ment of malignancy. Since many infectious processes often lead, directly or indi-
rectly, to increased cell turnover and proliferation, certain agents are now widely
regarded as carcinogens (Rosenthal and Purtilo, 1997).
In 1991, zur Hausen estimated that a significant fraction of all human can-
cers worldwide are associated with infections due to viruses, including human
papillomaviruses (cervical cancer and other skin cancers), human T-cell leuke-
mia viruses (adult T-cell leukemias and lymphomas in endemic areas), hepatitis
B virus (liver cancer), and Epstein-Barr virus (Burkitt’s lymphoma and nasopha-
ryngeal carcinoma) (zur Hausen, 1991). The estimate of the influence of infection
may now need to be revised in light of the fact that new viral associations have
been discovered and that other, nonviral associations have been uncovered
(Rosenthal and Purtilo, 1997). These include a common bacterial pathogen
(Helicobacter pylori infection with gastric carcinoma and MALT lymphoma),
and new viruses (hepatitis C virus with liver cancer, HHV-6 with non-Hodgkin’s
lymphoma, HHV-8 [a.k.a. KSHV]) with Kaposi’s sarcoma, Castleman’s disease,
and body cavity lymphomas (Mueller, 1995). In addition, new disease associa-
tions are being made with respect to previously known pathogens, such as the
association of chronic hepatitis C virus infection with non-Hodgkin’s lymphoma
in certain populations (Luppi et al., 1996). The following sections will summa-
rize briefly some of the established and emerging associations between chronic
infections and human cancer, as reflected in Table 3-1.
Helicobacter pylori
After many years of unwarranted skepticism, the medical establishment in
the United States and worldwide generally now recognizes Helicobacter pylori
as the most common cause of diffuse superficial gastritis and gastric and duode-
nal ulcers (McGowan et al., 1996). Infections caused by H. pylori persist within
the gastric mucosa for many years, and the incidence of infection is associated
with lower economic status and increasing age. Consistent with the step-wise
elucidation of the role of the etiologic agent, the treatment for gastric and duode-
nal ulcer has evolved from a surgical approach (gastrectomy), to medical man-
agement of gastric hyperacidity (H2 blockers), and most recently toward antibi-
otic therapy directed against H. pylori (McGowan et al., 1996).
Inflammation associated with H. pylori infection may progress to chronic
atrophic gastritis, which is a known predisposing condition for the development
of gastric carcinoma. Accordingly, H. pylori infection has been linked epidemio-
logically to gastric adenocarcinoma; many studies involving thousands of partici-
pants have now shown an increased risk of gastric cancer in persons with elevated
antibodies to Helicobacter pylori and in known H. pylori carriers (Uemura et al.,
2001). It is probable that host genetic factors (blood type, HLA type, other immu-
nogenetically determined factors) as well as microbial virulence factors (particu-
larly the presence of the cagA virulence factor) contribute to tissue burden of
organisms, persistence of infection, and the nature of the inflammatory response,
VIRUSES
the small bowel in association with the disease, converting to malignant lym-
phoma at later stages. Consistent with the experience of regression of MALT
lymphoma in H. pylori-infected patients, tetracycline treatment sometimes causes
regression of IPSID lesions in patients with early lesions, and is often used in
conjunction with chemotherapy in later stage lesions (Trotman et al., 1999). Fi-
nally, tropical phagedenic ulcer is a chronic persistent dermatological infection of
developing countries caused by coinfection with Fusobacterium and a spirochete,
Borrelia vincentii (Robinson et al., 1988). This infection leads to the develop-
ment squamous cell carcinoma within the depigmented margins of the ulcerative
lesion. Antibiotic treatment is effective for eliminating the bacterial infection and
presumably also for reducing cancer risk.
Taken together, it seems that chronic bacterial infections other than H. pylori
may also predispose to the development of malignancy, especially non-Hodgkins
lymphoma, by virtue of direct or indirect stimuli of target cell proliferation. Given
the increasing incidence of non-Hodgkin’s lymphoma in the US, and the fact that
the inciting organisms might be detectable in cancer patients at the time of pre-
sentation suggests that pathogen discovery efforts aimed at well-defined patient
populations might well be productive. More importantly, effective chemopre-
vention strategies may depend upon the identification of microbial, environmen-
tal, and host determinants in the development of neoplasia.
chronic biliary tract infection with Clonorchis sinensis and Opisthorchis viverrini
(Shin et al., 1996). Infection with both of these organisms is associated with
chronic inflammation and proliferation of the biliary epithelium. Chronic inflam-
mation of the bile duct epithelium due to infection with the newly recognized
protozoa Septata intestinalis and Cryptosporidium parvum have been recognized
in this country, but it is not yet known whether these infections are associated
with malignancy.
demic for malaria (the “lymphoma belt”). To this day, Burkitt’s lymphoma re-
mains one of the most dreaded diseases in sub-Saharan Africa. EBV can be found
in nearly 100 percent of endemic lymphomas, in contrast to its lower prevalence
in non-endemic Burkitt-type malignancies. In both endemic and non-endemic
tumor types, translocation of the c-myc cellular oncogene to an expressed locus
downstream from the immunoglobulin G promoter (IgG-P) occurs by gene rear-
rangement. However, the IgG-P/c-myc breakpoints in endemic cases occur within
a more tightly clustered region, suggesting stereotypic recombinational patterns
in the malaria-associated cases. Whether this pattern of recombination occurs
directly in response to malarial infection is currently unknown (Facer and Playfair,
1989), but an attractive hypothesis is that immunoglobulin gene rearrangement
driven by expansion of Plasmodium-specific B-cells participates in the develop-
ment of malignant clones. Differences in EBV subtypes have been observed in
endemic and non-endemic cases; endemic cases contain EBV subtype 2, in con-
trast to non-endemic cases which usually harbor EBV subtype 1 (Magrath et al.,
1992). Mechanisms for the possible pathogen interaction between EBV and Plas-
modium are poorly understood; the well known immunosuppressive effects of
malarial infection have been proposed by several investigators to activate EBV-
associated lymphoproliferation (Lam et al., 1991). The combination of immuno-
suppression with antigen-specific proliferation may, in this model, lead to the
development of a unique type of cancer.
EBV is also associated with nasopharyngeal carcinoma (NPC), a malignancy
that represents the most common tumor of males in southern China. Additional
environmental exposures to salted fish containing nitrosodimethylamines, and
perhaps inhaled herbal extracts have been implicated as possible cofactors in the
development of this malignancy. Latent forms of the EBV genome can be de-
tected by in situ hybridization. Immunologic predisposition may also contribute,
as reflected in relative overrepresentation of certain HLA types in patients with
NPC along with evidence of intrafamilial case clustering. In addition, an atypical
immune response as determined by the presence of IgA to certain EBV proteins
suggests that an aberrant immune response to the virus may underlie cancer risk
(Hsu et al., 2001). This EBV-specific IgA response may be an indirect, albeit
diagnostically important, indication of EBV infection on mucosal surfaces which
itself serves as a proferative stimulus of epithelial cell populations accompanied
by lymphocytic infiltration (lymphoepithelioma).
A recent report implicated EBV in the development of human breast cancer
(Magrath and Bhatia, 1999); various studies have detected up to a 51 percent
prevalence of EBV in breast cancer tissues, depending on the methods used. Since
the known EBV-associated carcinomas are lymphoepitheliomas, and since most
breast carcinomas are not lymphoepitheliomas, these findings have been contro-
versial and indeed many studies fail to confirm the initial findings (Chu et al.,
2001). A critical link to be established here would be the demonstration of un-
usual immunological responses to EBV proteins as demonstrated for NPC.
Human Retroviruses
The human retroviruses, human T cell leukemia viruses type I and II, infect
millions of persons worldwide and are associated with various neoplastic mani-
Hepatitis B Virus
Primary infection with hepatitis B virus usually follows an acute and conva-
lescent course in immunocompetent hosts with ultimate resolution of disease.
However, in approximately 5 to 10 percent of adults, and in most infants born to
infected carrier mothers, primary infection leads to chronic active hepatitis which
may progress to cirrhosis of the liver and hepatocellular carcinoma (Kew, 1996).
Continuous proliferation of hepatocytes, which may be present for the life of the
host, is a prominent feature of chronic active hepatitis; in such patients, the rela-
tive risk of developing hepatocelluar carcinoma is increased 20–40 fold. Because
of the large number of carriers of hepatitis B virus (HBV), approximately 200
million persons worldwide, hepatocellular carcinoma is one of the most common
cancers in the world and is the most common cancer in HBV-endemic areas of
the Far East and sub-Saharan Africa. The development of hepatocellular carci-
noma probably depends upon direct effects of certain viral determinants along
with chronic persistent proliferation of hepatocytes. HBV-related hepatocellular
carcinomas often contain defective HBV genomes expressing one or more viral
open reading frames; the viral X gene, which is often expressed in malignant
tissues, is a transcriptional transactivator of host promoter sequences including
those associated with oncogene expression and integrated viral genomes can acti-
vate host proto-oncogenes. There is little doubt that continuous proliferation of
hepatocytes is also an important contributor to the development of neoplastic
disease, since other disorders associated with chronic hepatitis and hepatocyte
turnover (such as chronic alcoholism or hereditary hemochromatosis) may also
lead to an increased predisposition to liver cancer, and the effects of alcohol in-
gestion and viral infection may well be synergistic in this regard (Brechot et al.,
1996).
Hepatitis C Virus
A rapidly emerging association of chronic viral infection with malignancy is
the development of hepatocellular carcinoma associated with chronic hepatitis C
virus infection. In the United States, HCV infection is projected to become the
major cause of liver cancer, due in part the large number of chronic carriers in this
country, currently estimated at 3.5 million persons. In the case of HCV infection,
a major determinant of development of hepatocellular cancer appears to be dura-
tion of infection and the degree of persistent liver injury (Zein and Persing, 1996).
Several studies have associated certain HCV subtypes with differences in liver
disease severity, interferon responsiveness, and duration of infection. Viral sub-
types that appear to have been in the U.S. population for longer periods of time
are dramatically overrepresented among patients with liver cancer. Specifically,
subtype 1b, which is present in 15–20 percent of cases nationwide, is present in
90 percent or more of liver cancer patients in several studies conducted at differ-
ent centers (Zein et al., 1996). Since presence of genotype 1b appears to be a
marker of longer duration of infection in U.S. patients, it is possible that other
genotypes will be more commonly implicated in cases of hepatocellular carci-
noma in other countries, and that the overrepresentation of genotype 1b in the
United States will decline over time. Additional factors such as alcohol consump-
tion may contribute independently to risk of neoplasia, even in patients with more
recent infections (Brechot et al., 1996). As for HPV infection, host immunoge-
netic or other factors may play a role in susceptibility to viral infection as well as
in determining the severity of infection (see below).
Recently, chronic HCV infection has been associated with the development
of B-cell non-Hodgkin’s lymphomas in an Italian population (Mele et al., 2003)
and with the development of cryoglobulinemia and monoclonal gammopathy in
the United States (Cacoub et al., 1994). The development monoclonal
gammopathy is thought to presage the development of hematologic malignancies
in a significant subset of cases; it is not yet known whether HCV-associated mono-
clonal gammopathy or mixed cryoglobulinemia is a predisposing variable for the
development of such malignancies in the United States. One recent study sug-
gests that HCV infection-associated lymphoproliferative disease might include
multiple myeloma (Montella et al. 2001). It is possible that MGUS is a marker of
an underlying benign, perhaps antigen driven lymphoproliferative process, which
may after many years convert to a malignant process via somatic mutation. In this
respect, the genesis of neoplasia may be similar to that described for Helicobacter
pylori-related MALT lymphoma. The relative disparity in the frequency of non-
Hodgkin’s lymphoma associated with HCV infection in the United States com-
pared to that in Europe may conceivably reflect differences in duration of infec-
tion within the population, immunogenetic differences, or exposure to other
oncoviruses including KHSV.
al., 1965). Inoculation of BPV into newborn hamsters or mice has also been dem-
onstrated to induce tumors (zur Hausen, 1996). Some human studies in recent
years have suggested that relatively common HPV types can be detected in hu-
man transitional cell carcinoma (TCC) of the bladder in up to 34 percent of cases
in certain patient populations (Smetana et al., 1995), but the presence of HPV in
the neoplastic tissue has been difficult to demonstrate with consistency (Chetsanga
et al., 1992). Interestingly, cases of rapidly progressive multifocal TCC of the
bladder have been reported in patients following renal and cardiac transplanta-
tion, suggesting that HPV may play a more significant role in TCC of the bladder
in immunosuppressed patients (Noel et al., 1994). Although smoking history is
the leading risk factor in the development of squamous cell carcinoma of the
lung, a recent study implicated the presence of HPV type 16 in an unexpectedly
high number of cases of well-differentiated squamous cell carcinoma of the lung
on the island of Okinawa (Kinoshita et al., 1995). However, although some stud-
ies have supported this finding (Hirayasu et al., 1996), others have failed to impli-
cate HPV in lung cancer (Szabo et al., 1994). In this regard, it is interesting to
note that risk of cervical cancer is linked epidemiologically to risk of carcinoma
of the lung but not uterine or ovarian cancers, and that both of the former are
linked to smoking (Anderson et al., 1997). Clearly, additional studies will be
necessary which are designed to rule out the effects of incidental virus and DNA
contamination, yet also designed to be capable of detection of the widest range of
HPV types.
Recent studies of the natural history of HPV infection have suggested that
the ability of the genetically heterogeneous papillomaviruses to exploit relative
deficiencies in the immune response may represent an important determinant of
the risk of developing chronic infection and subsequent neoplastic disease. Atten-
tion has focused recently on cytokine production in HPV-specific helper T lym-
phocyte populations in women with cervical cancer compared to women in whom
the disease regresses spontaneously. Several studies have now indicated that fail-
ure to mount cytotoxic T cell (CTL) responses to human papillomavirus-infected
cells may significantly predispose to the development of cervical cancer (Tsukui
et al., 1996); this failure may be virus type-specific, such that the types repre-
sented most often in cervical cancer are those most successful at avoidance of
CTL activity (Ellis et al., 1995). Conditions associated with reduced CTL activ-
ity, such as HIV and HTLV infection, oral contraceptive use, pregnancy, and
immune suppression associated with transplantation have likewise been associ-
ated with increased HPV viral burden and acceleration of disease progression
(Sun et al., 1995). Twin studies have suggested that risk factors for cervical can-
cer, as for another HPV-associated lesion, epidermoplasia verruciformis, may be
inherited (Ahlbom et al., 1997). More recent studies have failed to detect an asso-
ciation with somatic mutations in the gene encoding p53, and have further ex-
plored associations with the immunoglobulin gene cluster (Cuzick et al., 2000).
Understanding the contribution of the environmental, host somatic, and host im-
Conclusions
In purely reductionistic terms, infectious diseases can be viewed as horizon-
tally acquired genetic disorders, in which exogenously acquired nucleic acids of a
pathogen integrate, either chromosomally, episomally, or extracellularly, with
those of the host to disrupt normal cellular processes or produce inflammation.
Developing a better understanding of the interactions of human microbial flora
with their hosts, along with an understanding of other host and environmental
determinants of pathogenicity, represents an increasingly important intersection
of infectious disease research with the Human Genome Project. Illustrations of
the latter concept are provided by the discovery of resistance to HIV infection by
virtue of mutations within a gene encoding an HIV coreceptor (CCR-5) (Samson
et al., 1996). Another important example is the inherited susceptibility to Myco-
bacterium avium-intracellulare infections in families carrying a mutant allele of
the interferon-gamma (IFN-γ) receptor (Newport et al., 1996). The identification
of such an allele within the human population has far-reaching implications, since
it may be associated with a general reduction in the ability to mount cytotoxic T-
cell responses during infections of many types. Genetic predisposition to
REFERENCES
Ablashi DV, Chatlynne L, Thomas D, Bourboulia D, Rettig MB, Vescio RA, Viza D, Gill P, Kyle
RA, Berenson JR, Whitman JE. 2000. Lack of serologic association of human herpesvirus-8
(KSHV) in patients with monoclonal gammopathy of undetermined significance with and with-
out progression to multiple myeloma. Blood 96:2304–2306.
Ahlbom A, Lichtenstein P, Malmstrom H, Feychting M, Hemminki K, Pedersen NL. 1997. Cancer in
twins: genetic and nongenetic familial risk factors. Journal of the National Cancer Institute
89:287–293.
Ambinder RF. 1990. Human lymphotropic viruses associated with lymphoid malignancy: Epstein-
Barr and HTLV-1. Hematology Oncology Clinics of North America 4:821–833.
Anderson KE, Woo C, Olson JE, Sellers TA, Zheng W, Kushi LH, Folsom AR. 1997. Association of
family history of cervical, ovarian, and uterine cancer with histological categories of lung can-
cer—the Iowa women’s health study. Cancer Epidemiology, Biomarkers & Prevention 6:401–
405.
Beksac M, Ma M, Akyerli C, DerDanielian M, Zhang L, Liu J, Arat M, Konuk N, Koc H, Ozcelik T,
Vescio R, Berenson JR. 2001. Frequent demonstration of human herpesvirus 8 (HHV-8) in bone
marrow biopsy samples from Turkish patients with multiple myeloma (MM). Leukemia
15:1268–1273.
Brechot C, Nalpas B, Feitelson MA. 1996. Interactions between alcohol and hepatitis viruses in the
liver. Clinics in Laboratory Medicine 16:273–287.
Buchanan J and Nieland-Fisher NS. 2001. Role of immune function in human papillomavirus infec-
tion. Journal of the American Medical Association 286:1173–1174.
Cacoub P, Fabiani FL, Musset L, Perrin M, Frangeul L, Leger JM, Huraux JM, Piette JC, Godeau P.
1994. Mixed cryoglobulinemia and hepatitis C virus. American Journal of Medicine 96:124–
132.
Chang Y and Moore PS. 1996. Kaposi’s Sarcoma (KS)-associated herpesvirus and its role in KS.
Infectious Agents & Disease 5:215–222.
Chen YT. 2000. Cancer vaccine: identification of human tumor antigens by SEREX. Cancer Journal
6 (Suppl 3):S208–217.
Chetsanga C, Malmstrom PU, Gyllensten U, Morenolopez J, Dinter Z, Peterson U. 1992. Low inci-
dence of human papillomavirus type 16 DNA in bladder tumour detected by polymerase chain
reaction. Cancer 69:1208–1211.
Chu PG, Chang KL, Chen YY, Chen WG, Weiss LM. 2001. No significant association of Epstein-
Barr virus infection with invasive breast carcinoma. American Journal of Pathology 159:571–
578.
Magrath I, Jain V, Bhatia K. 1992. Epstein-Barr virus and Burkitt’s lymphoma. Seminars in Cancer
Biology 3:285–295.
Majewski S and Jablonska S. 1995. Epidermodysplasia verruciformis as a model of human
papillomavirus-induced genetic cancer of the skin. Archives of Dermatology 131:1312–1318.
McGowan CC, Cover TL, Blaser MJ. 1996. Helicobacter pylori and gastric acid: biological and thera-
peutic implications. Gastroenterology 110:926–938.
Mele A, Pulsoni A, Bianco E, Musto P, Szklo A, Sanpaolo MG, Iannitto E, De RenzoA, Martino B,
Liso V, Andrizzi C, Pusterla S, Dore F, Maresca M, Rapicetta M, Marcucci F, Mandelli F,
Franceschi S. 2003. Hepatitis C virus and B-cell non-Hodgkin lymphomas: an Italian multicenter
case-control study. Blood. 102(3):996–999.
Montella M, Crispo A, Frigeri F, Ronga D, Tridente V, De Marco M, Fabbrocini G, Spada O, Mettivier
V, Tamburini M. 2001. HCV and tumors correlated with immune system: a case-control study
in an area of hyperendemicity. Leukemia Research 25:775–781.
Moore PS and Chang Y. 1995. Detection of herpesvirus-like DNA sequences in Kaposi’s sarcoma in
patients with and without HIV infection [see comments]. New England Journal of Medicine
332:1181–1185.
Moro MH, Bjornsson J, Marietta EV, Hofmeister EK, Germer JJ, Bruinsma E, David CS, Persing DH.
2001. Gestational attenuation of Lyme arthritis is mediated by progesterone and IL-4. Journal of
Immunology 166:7404–7409.
Mueller N. 1995. Overview: viral agents and cancer. Environmental Health Perspectives 103(Suppl
8):259–261.
Newport MJ, Huxley CM, Huston S, Hawrylowicz CM, Oostra BA, Williamson R, Levin M. 1996. A
mutation in the interferon-gamma-receptor gene and susceptibility to mycobacterial infection.
New England Journal of Medicine 335:1941–1949.
Noel JC, Thiry L, Verhest A, Deschepper N, Peny MO, Sattar AA, Schulman CC, Haot J. 1994.
Transitional cell carcinoma of the bladder: evaluation of the role of human papillomaviruses.
Urology 44:671–675.
O’Connor F, Buckley M, O’Morain C. 1996. Helicobacter pylori: the cancer link. Journal of the
Royal Society of Medicine 89:674–678.
Olson C, Pamucku AM, Brobst DF. 1965. Papillomalike virus from bovine urinary tumours. Cancer
Research 25:840–847.
Palmarini M, Sharp JM, de las Heras M, Fan H. 1999. Jaagsiekte sheep retrovirus is necessary and
sufficient to induce a contagious lung cancer in sheep. Journal of Virology 73:6964–6972.
Persing DH. 1997. The cold zone: a curious convergence of tick-transmitted diseases. Clinical Infec-
tious Diseases 25(Suppl 1):S35–42.
Plumelle Y, Gonin C, Edouard A, Bucher BJ, Thomas L, Brebion A, Panelatti G. 1997. Effect of
Strongyloides stercoralis infection and eosinophilia on age at onset and prognosis of adult T-cell
leukemia. American Journal of Clinical Pathology 107:81–87.
Ramzan NN, Loftus E, Burgart LJ, Rooney M, Batts KP, Wiesner RH, Fredricks DN, Relman DA,
Persing DH. 1997. Diagnosis and monitoring of Whipple disease by polymerase chain reaction.
Annals of Internal Medicine 126(7).
Rettig MB, Ma HJ, Vescio RA, Pold M, Schiller G, Belson D, Savage A, Nishikubo C, Wu C, Fraser
J, Said JW, Berenson JR. 1997. Kaposi’s sarcoma-associated herpesvirus infection of bone
marrow dendritic cells from multiple myeloma patients. Science 276:1851–1854.
Robinson DC, Adriaans B, Hay RJ, Yesudian P. 1988. The clinical and epidemiologic features of
tropical ulcer (tropical phagedenic ulcer). International Journal of Dermatology 27:49–53.
Rosenthal LJ and Purtilo DT. 1997. Neoplasms associated with infectious agents. P. 1707. In Pathol-
ogy of Infectious Diseases, vol. II, DH Connor, FW Chandler, HJ Manz, DA Schwartz, EE
Lack, eds. Stamford, CT: Appleton and Lange.
Rosin MP, Anwar WA, Ward AJ. 1994. Inflammation, chromosomal instability, and cancer: the schis-
tosomiasis model. Cancer Research 54:1929s–1933s.
Samson M, Libert F, Doranz BJ, Rucker J, Liesnard C, Farber CM, Saragosti S, Lapoumeroulie C,
Cognaux J, Forceille C, Muyldermans G, Verhofstede C, Burtonboy G, Georges M, Imai T,
Rana S, Yi Y, Smyth RJ, Collman RG, Doms RW, Vassart G, Parmentier M. 1996. Resistance
to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine
receptor gene [see comments]. Nature 382:722–725.
Sauter M, Schommer S, Kremmer E, Remberger K, Dolken G, Lemm I, Buck M, Best B, Neumann-
Haefelin D, Mueller-Lantzsch N. 1995. Human endogenous retrovirus K10: expression of Gag
protein and detection of antibodies in patients with seminomas. Journal of Virology 69:414–
421.
Shamanin V, zur Hausen H, Lavergne D, Proby CM, Leigh IM, Neumann C, Hamm H, Goos M,
Haustein UF, Jung EG et al. 1996. Human papillomavirus infections in nonmelanoma skin
cancers from renal transplant recipients and nonimmunosuppressed patients [see comments].
Journal of the National Cancer Institute 88:802–811.
Shin HR, Lee CU, Park HJ, Seol SY, JM Chung, Choi HC, Ahn YO, Shigemastu T. 1996. Hepatitis
B and C virus, Clonorchis sinensis for the risk of liver cancer: a case-control study in Pusan,
Korea. International Journal of Epidemiology 25:933–940.
Smetana Z, Keller T, Leventon-Kriss S, Huszar M, Lindner A, Mitrani-Rosenbaum S, Mendelson E,
Smetana S. 1995. Presence of human papilloma virus in transitional cell carcinoma in Jewish
population in Israel. Cellular & Molecular Biology 41:1017–1023.
Spina M and Tirelli U. 1992. Human immunodeficiency virus as a risk factor in miscellaneous can-
cers. Current Opinion in Oncology 4:907–910.
Stoler MH. 2001. HPV for cervical cancer screening: is the era of the molecular Pap smear upon us?
Journal of Histochemistry and Cytochemistry 49:1197–1198.
Strickler HD, Rattray C, Escoffery C, Manns A, Schiffman MH, Brown C, Cranston B, Hanchard B,
Palefsky JM, Blattner WA. 1995. Human T-cell lymphotropic virus type I and severe neoplasia
of the cervix in Jamaica. International Journal of Cancer 61:23–26.
Sun XW, Ellerbrock TV, Lungu O, Chiasson MA, Bush TJ, Wright T Jr. 1995. Human papillomavirus
infection in human immunodeficiency virus-seropositive women. Obstetrics & Gynecology
85:680–686.
Szabo I, Sepp R, Nakamoto K, Maeda M, Sakamoto H, Uda H. 1994. Human papillomavirus not
found in squamous and large cell lung carcinomas by polymerase chain reaction [see com-
ments]. Cancer 73:2740–2744.
Thijs JC, Kuipers EJ, van Zwet AA, Pena AS, de Graaff J. 1995. Treatment of Helicobacter pylori
infections. QJM 88:369–389.
Trimble JJ and Desrosiers RC. 1991. Transformation by herpesvirus saimiri. Advances in Cancer
Research 56:335–355.
Trotman BW, Pavlick AC, Igwegbe IC, Goldstein MM. 1999. Immunoproliferative small intestinal
disease: case report and literature review. Journal of the Association for Academic Minority
Physicians 10:88–93.
Tsukui T, Hildesheim A, Schiffman MH, Lucci JR, Contois D, Lawler P, Rush BB, Lorincz AT,
Corrigan A, Burk RD, Qu W, Marshall MA, Mann D, Carrington M, Clerici M, Shearer GM,
Carbone DP, Scott DR, Houghten RA, Berzofsky JA. 1996. Interleukin 2 production in vitro by
peripheral lymphocytes in response to human papillomavirus-derived peptides: correlation with
cervical pathology. Cancer Research 56:3967–3974.
Uemura N, Okamoto S, Yamamoto S, Matsumura N, Yamaguchi S, Yamakido M, Taniyama K,
Sasaki N, Schlemper RJ. 2001. Helicobacter pylori infection and the development of gastric
cancer. New England Journal of Medicine 345:784–789.
Weber DM, Dimopoulos MA, Anandu DP, Pugh WC, Steinbach G. 1994. Regression of gastric lym-
phoma of mucosa-associated lymphoid tissue with antibiotic therapy for Helicobacter pylori.
Gastroenterology 107:1835–1838.
Zein NN and Persing DH. 1996. Hepatitis C genotypes: current trends and future implications. [Re-
view] [50 refs]. Mayo Clinic Proceedings 71:458–462.
Zein NN, Poterucha JJ, Gross J Jr, Wiesner RH, Therneau TM, Gossard AA, Wendt NK, Mitchell PS,
Germer JJ, Persing DH. 1996. Increased risk of hepatocellular carcinoma in patients infected
with hepatitis C genotype 1b. American Journal of Gastroenterology 91:2560–2562.
zur Hausen H. 1991. Viruses in human cancers. Science 254:1167–1173.
zur Hausen H. 1996. Roots and perspectives of contemporary papillomavirus research. Journal of
Cancer Research and Clinical Oncology 122:3–13.
zur Hausen H and Rosl F. 1994. Pathogenesis of cancer of the cervix. Cold Spring Harbor Symposia
on Quantitative Biology 59:623–628.
OVERVIEW
The ultimate goal in identifying connections between infectious agents and
chronic diseases is to find better ways to cure, or even prevent, those diseases.
Various therapeutic approaches may prove fruitful and deserve continued atten-
tion. Several of these areas were explored during the workshop, along with ways
to most effectively move therapeutic methods from the laboratory into widespread
practice. Discussion was somewhat limited, however, by time constraints and the
availability of speakers.
Among therapeutic possibilities, vaccines hold a special attraction, given their
unique record of totally eliminating or eradicating several target diseases. Recent
advances in molecular biology and genetics have provided powerful new meth-
ods for vaccine development. Of particular importance is the possibility offered
by the new sciences of genomics and proteomics to detect the relevant antigens
and to identify specific aspects of immunity that should be enhanced by the vac-
cine. There also have been important advances made in understanding the pro-
cesses of acquired immunity and the pathogenesis of various diseases (including
the interplay between microbe and host). Collectively, these efforts may point the
way to the eventual development of effective means to control at least some
chronic diseases.
To underpin scientific efforts, sound planning and program management is a
must. In order to most effectively develop and implement new prevention and
intervention efforts—encompassing vaccines and a host of other means—there
will need to be in place an overarching strategy to guide efforts across a range of
fronts. This strategy will rely heavily on collaborations to enhance the productiv-
174
*I wish to thank warmly colleagues whose contribution to this paper have been essential: Jenni M.
Vuola, Mirja Puolakkainen, Tuula Penttilä, Anne Sarén, Anu Haveri, and Laura Mannonen from the
Institute or Department of Virology, Haartman Institute, University of Helsinki.
ence and persistence, of the microbe (as seems to be the case with gastric ulcer
associated with Helicobacter pylori infection) or mediated by a process once set
in motion or fully carried out by the microbe that then disappears (the latter may
be the case with juvenile diabetes due to destruction of islet cells associated with
a viral infection).
3. How does acquired immunity affect the condition? In case of persistent
infection we would need to know the characteristics of the immunity that allows
the infection to persist—which component is missing and why, how could we
change the situation to convert the persistence-associated immunity to a protec-
tive immunity? Would it be possible to affect this during the persistent state so
that the persistence could be cured? For vaccine development, the characteristics
of protective immunity for each infection would need to be known. Questions
that can be asked to help this characterization include the role of antibodies (if
yes, to which antigens? is high affinity essential? would some isotype be specifi-
cally needed?) and the role of T cells (and further, whether it is the CD4+ cells
stimulated by antigenic peptide bound to the MCH class II molecules on the
surface of antigen-presenting cells or the CD8+ cells stimulated by peptide bound
to class I molecules, and whether it should be a Th 1- or Th 2-type response, each
associated with its separate sets of cytokines).
4. Do we have an experimental animal model? The use of such a model is
normally an essential part of vaccine development that helps to convert the theo-
retical findings at the laboratory bench to a form on which we can predict what
can be expected of the vaccine’s performance in humans. The better the model,
the more we can learn about the candidate vaccine, and more importantly, about
the pathogenesis of the infection, the disease, and the immunity. Nevertheless, it
is also true that no animal experiments can replace final clinical trials in which
the vaccine is evaluated in its real target population.
though one is currently needed for efficient delivery of the annual influenza vac-
cine. However, the development of a vaccine capable of curing an established
persistent infection is a formidable challenge, and no such vaccine exists as a
model.
ing for the importance of the Type III system also in Chlamydiae (Fields and
Hackstadt, 2000; Bavoil et al., 2000).
In addition to hiding from the immune system by their intracellular lifestyle,
Chlamydiae have also developed a sophisticated mechanism for thwarting the
MHC-based immune recognition. This takes place by downregulation of the tran-
scription of both MHC class I and class II molecules; the downregulation is medi-
ated by specific degradation by C. pneumoniae-coded proteins of host cell factors
promoting the transcription (Zhong et al., 1999, 2000). This is an entirely novel
mechanism of immune evasion by pathogenic microbes, and certainly presents a
formidable challenge for vaccine development.
The immune recognition and subsequent killing of the infected cells is not
necessarily uniform for all cells; thus the infection may proceed or become per-
sistent in only a part of the initially infected cells. Then we are faced with the
likely possibility that antigen presentation in the persistently infected cells will
differ from that seen during the acute phase. Very likely, therefore, a vaccine
capable of preventing and curing the persistent infection will be different from
the one preventing the acute infection. The mouse model may be suitable for
studying the persistent infection, too. C. pneumoniae-specific DNA can be de-
tected by PCR for at least 2 months after cultures have turned negative, and the
dormant infection can be reactivated by immunosuppressive treatment of the mice
(Malinverni et al., 1995; Laitinen et al., 1996). Our experience with both methods
of detection suggests that persistence develops in part of the animals only, a fur-
ther point of resemblance to the human C. pneumoniae infection. Persistent infec-
tion can also be detected in vitro in cell cultures of C. pneumoniae (Byrne et al.,
2001; Pantoja et al., 2001). Such a mode of growth can be induced by treatment
of the culture with IFN-γ or sublethal doses of antibiotics or by depletion of
tryptophan. The morphology of the inclusion bodies in which C. pneumoniae
normally multiply and mature to the infectious EB forms changes and no EBs are
seen.
Crucial questions on the path to the development of a vaccine aimed at cur-
ing the persistent C. pneumoniae infection then include:
REFERENCES
Bavoil PM, Hsia RC, Ojcius DM. 2000. Closing in on Chlamydia and its intracellular bag of tricks.
Microbiology 146:2713–2731.
Byrne GI, Ouellette SP, Wang Z, Rao JP, Lu L, Beatty WL, Hudson AP. 2001. Chlamydia pneumoniae
expresses genes required for DNA replication but not cytokinesis during persistent infection of
HEp-2 cells. Infection and Immunity 69:5423–5429.
Fields KA and Hackstadt T. 2000. Evidence for the secretion of Chlamydia trachomatis CopN by a
type III secretion mechanism. Molecular Microbiology 38:1048–1060.
Grayston JT. 2000. Background and current knowledge of Chlamydia pneumoniae and atherosclerosis.
The Journal of Infectious Diseases 181:S402–410.
Igietseme JU, Black CM, Caldwell HD. 2002. Chlamydia vaccines. Strategies and status. Biodrugs
16:19–35.
Kaukoranta-Tolvanen S-SE, Laurila AL, Saikku P, Leinonen M, Liesirova L, Laitinen K. 1993.
Experimental infection of Chlamydia pneumoniae in mice. Microbial Pathogenesis 15:293.
Laitinen K, Laurila AL, Leinonen M, Saikku P. 1996. Reactivation of Chlamydia pneumoniae infec-
tion in mice by cortisone treatment. Infection and Immunity 64:1488–1490.
Lee CL and Ko YC. 1997. Hepatitis B vaccination and hepatocellular carcinoma in Taiwan. Pediat-
rics 99:351–353.
Malinverni R, Kuo CC, Campbell LA, Grayston JT. 1995. Reactivation of Chlamydia pneumoniae
lung infection in mice by cortisone. The Journal of Infectious Diseases 172:593–594.
Murdin AD, Dunn P, Sodoyer R, Wang J, Caterini J, Brunham RC, Aujame L, Oomen R. 2000. Use
of a mouse lung challenge model to identify antigens protective against Chlamydia pneumoniae
lung infection. The Journal of Infectious Diseases 181(Suppl 3):S544–551.
O’Connor S, Taylor C, Lee AC, Epstein S, Libby P. 2001. Potential infectious etiologies of athero-
sclerosis: A multifactorial perspective. Emerging Infectious Diseases 7:780–788.
Pantoja LG, Miller RD, Ramirez JA, Molestina RE, Summersgill JT. 2001. Characterization of
Chlamydia pneumoniae persistence in HEp-2 cells treated with gamma interferon. Infection and
Immunity 69:7927–7932.
Penttilä JM, Anttila M, Puolakkainen M, Laurila A, Varkila K, Sarvas M, Mäkelä PH, Rautonen N.
1998. Local immune responses to Chlamydia pneumoniae in the lungs of BALB/c mice during
primary infection and reinfection. Infection and Immunity 66:5113–5118.
Penttilä JM, Anttila M, Varkkila K, Puolakkainen M, Sarvas M, Mäkelä PH, Rautonen N. 1999.
Depletion of CD8+ cells abolishes memory in acquired immunity against Chlamydia
pneumoniae in BALB/c mice. Immunology 97:490–496.
Penttilä T, Vuola JM, Puurula V, Anttila M, Sarvas M, Rautonen N, Mäkelä PH, Puolakkainen M.
2000. Immunity to Chlamydia pneumoniae induced by vaccination with DNA vectors express-
ing a cytoplasmic protein (Hsp60) or outer membrane proteins (MOMP and Omp2). Vaccine
19:1256–1265.
Rottenberg ME, Rotfuchs ACG, Gigliotti D, Svanholm C, Bandholtz L, Wigzell HJ. 1999. Role of
innate and adaptive immunity in the outcome of primary infection with Chlamydia pneumoniae,
as analyzed in genetically modified mice. The Journal of Immunology 162:2829–2836.
Saikku P. 1999. Epidemiology of Chlamydia pneumoniae in atherosclerosis. American Heart Journal
138:S500–503.
Svanholm C, Bandholtz L, Castanos-Velez E, Wigzell H, Rottenberg ME. 2000. Protective DNA
immunization against Chlamydia pneumoniae. Scandinavian Journal of Immunology 51:345–
353.
Zhong G, Fan T, Liu L. 1999. Chlamydia inhibits interferon γ-inducible major histocompatibility
complex class II expression by degradation of upstream stimulatory factor 1. The Journal of
Experimental Medicine 189:1931–1938.
Zhong G, Liu L, Fan T, Fan P, Ji H. 2000. Degradation of transcription factor RFX5 during the
inhibition of both constitutive and interferon γ-inducible major histocompatibility complex class
I expression in Chlamydia-infected cells. The Journal of Experimental Medicine 191:1525–
1534.
Evidence that microbes are at the root of chronic conditions such as peptic
ulcer disease, Whipple’s disease, hepatocellular carcinoma, and cervical cancer
has transformed medicine. These examples underscore the plausibility that infec-
tious agents might be linked to numerous other non-communicable chronic con-
ditions. Indeed, research into speculative and as yet to be proposed associations
can no longer be viewed as “fishing expeditions,” despite the investigative chal-
lenges. Strategies that use collaborations to enhance the productivity of indepen-
dent investigators, integrate rigorously executed laboratory techniques into well-
designed epidemiologic studies and surveillance systems, and complement
short-term studies with long-term follow-up can overcome the hurdles to create
new prevention and intervention opportunities. Balancing research on potential
infectious links for common chronic conditions, in which the contribution of mi-
crobes to overall burden could be minor, with that on less common diseases,
perhaps likely to have a primary infectious cause, could benefit many.
The Basics
Certain basic tenets, however, are crucial to successfully defining, character-
izing, and mitigating the burden of chronic diseases that is induced by infectious
diseases. Widespread use of standardized or comparable case definitions—for the
infection and the outcome—is needed for comparisons across studies and conclu-
sions on causality. So, too, are universally high standards of specificity, sensitiv-
ity, and reproducibility in laboratory assays, applied to appropriate specimens
and controls. Minimum performance criteria are feasible, even when investigator
creativity enters uncharted territory or population and exposure differences im-
pact reproducibility. Peer review journals can reinforce these standards if publi-
cation depends upon the use of sound epidemiologic design and laboratory assays
capable of supporting the conclusions.
• Pathogen discovery activities that: identify novel agents, define which species
of known agents impart chronic sequelae, and detect agents in alternative tissues;
• Development of new and improved laboratory technologies to advance
pathogen discovery and the detection of known agents;
• Expansion of viral screening methodology; and
• Continued development of improved, more sensitive and specific labora-
tory diagnostic assays that can identify an infectious root of disease at the site of
pathology or a distant site, and distinguish active from latent or past infection.
Overarching Issues
Strategies that build on fundamentals of sound science are likely to identify
and clarify additional infectious disease-chronic disease links, confirming this
arena to be both our challenge and our future. Some investments of today may
yield early answers, while other returns may take years. That is the nature of the
unknown and of chronic conditions. However, the potential benefits of these in-
vestments to populations and to individuals are great—greatest if investment be-
gins now. As the field expands two additional, crosscutting issues call for con-
tinuous consideration:
Although the links between infectious diseases and chronic diseases can be
complex and multifactorial, they can be characterized, and beneficial interven-
tions against infection designed. Most fruitful is an approach that increases in-
vestments in the laboratory, epidemiology, and surveillance, emphasizing inte-
gration of these elements and collaborations that can increase the yield of research
and medical science.
Appendix
A
Workshop Agenda
AGENDA
Session I
Case Studies of Infectious Agents Associated with Chronic Diseases
Evidence continues to mount implicating microorganisms as etiologic agents of
chronic diseases that contribute to substantial mortality and morbidity. This ses-
sion will examine definitive and emerging associations between infectious agents
and chronic diseases with a range of pathogenic mechanisms and diversity in
etiologic microbes. The review will explore advances in research, detection, and
187
screening that have contributed to these discoveries and some of the challenges
that remain.
11:15 BREAK
1:00 LUNCH
Session II
Challenges in Framing the Research
Identification and confirmation of the infectious causation of chronic dis-
eases are complicated by several factors, which include detection of microbes at
the time of diagnosis of the chronic condition, the lack of adequate methods to
identify novel or rare microorganisms, and the influence of environmental and
genetic factors on the etiology of the chronic diseases. This session will examine
these challenges and identify existing and potential methods and technologies for
overcoming these obstacles.
APPENDIX A 189
3:30 BREAK
Session III
Discussion Panel: Shaping the Research and Development Agenda
3:45 Panel members, Forum members, and the audience will comment on and
respond to considerations such as the role of industry in developing diagnostics;
possibilities for the coordination between basic and clinical scientists, patholo-
gists, and epidemiologists in developing standardized specific case definitions
and specimens and the development of comparable methods of analysis; the les-
sons that can be learned about the microbes from the chronic sequelae they pro-
duce; and methods for funding the research.
Session IV
Implications for Developing Countries
As researchers, clinicians, and policymakers have recognized the growing
disease burden from chronic diseases in developing countries, understanding of
the infectious etiology of these diseases becomes increasingly important in these
areas where many infectious diseases still remain endemic. This session will
review the consequences of highly prevalent infectious diseases linked to chronic
diseases and explore the global and local response needed to combat these out-
comes in resource-limited environments.
Session V
Barriers and Opportunities to Detect, Prevent, and Mitigate the Impact of
Chronic Diseases Caused by Infectious Agents
The complexity of the relationship between infectious agents and chronic
diseases requires a multi-disciplinary approach to reveal the implications of early
detection and prevention of chronic diseases caused by infectious agents. This
session will summarize the advances and gaps in collaborative research on detec-
tion and diagnostic technologies, their integration with epidemiological studies
and surveillance that can forward the efforts in this important area, and the impli-
cations for clinical management practices and priorities.
APPENDIX A 191
3:00 BREAK
Session VI
Discussion Panel: The Next Steps for the Healthcare Community
Panel members, Forum members, and the audience will comment on and
respond to considerations such as the role of industry and academic research in
developing treatments; the implications for the health care and prevention com-
munity in detecting and treating these diseases; and the benefits of managing
acute infections vs. chronic diseases—the argument for vaccines and antimicro-
bials.
Appendix
B
Information Resources
OVERVIEW
Danesh J, Newton R, Beral V. 1997. A human germ project? Nature 389:21–24.
Mueller N. 1995. Overview: viral agents and cancer. Environmental Health Perspectives 103:259–
261.
Parsonnet J. 1995. Bacterial infection as a cause of cancer. Environmental Health Perspectives 103
Suppl 8:263–268.
Persing DH and Prendergast FG. 1999. Infection, immunity, and cancer. Archives of Pathology &
Laboratory Medicine 123:1015–1022.
CAUSAL ASSOCIATIONS
Commission on Tropical Diseases of the International League Against Epilepsy. 1994. Relationship
between epilepsy and tropical diseases. Epilepsia 35:89–93.
Durkin MS, Khan NZ, Davidson LL, Huq S, Munir S, Rasul E, Zaman SS. 2000. Prenatal and post-
natal risk factors for mental retardation among children in Bangladesh. American Journal of
Epidemiology 152:1024–1033.
El-Serag HB and Mason AC. 2000. Risk factors for the rising rates of primary liver cancer in the
United States. Archives of Internal Medicine 160:3227–3230.
Epstein SE, Zhou YF, Zhu J. 1999. Infection and atherosclerosis: emerging mechanistic paradigms.
Circulation 100:E20–E28.
Franco EL, Duarte-Franco E, Ferenczy A. 2001. Cervical cancer: epidemiology, prevention, and the
role of human papillomavirus infection. Canadian Medical Association Journal 164:1017–1025.
Gilden DH, Burgoon MP, Kleinschmidt-DeMasters BK, Williamson RA, Ghausi O, Burton DR,
Owens GP. 2001. Molecular immunologic strategies to identify antigens and β-cell repsonses
unique to multiple sclerosis. Archives of Neurology 58:43–48.
Johnson RT. 1994. The virology of demyelinating diseases. Annals of Neurology 36 Suppl:S54–S60.
192
APPENDIX B 193
Lipkin WI, Hornig M, Briese T. 2001. Borna disease virus and neuropsychiatric disease—a reappraisal.
Trends in Microbiology 9:295–298.
Maeda N, Palmarini M, Murgia C, Fan H. 2001. Direct transformation of rodent fibroblasts by
jaaagsiekte sheep retrovirus DNA. Proceedings of the National Academy of Sciences 98:4449–
4454.
Morris JA. 1995. Schizophrenia, bacterial toxins and the genetics of redundancy. Medical Hypotheses
46:362–366.
Roivainen M, Rasilainen S, Ylipaasto P, Nissinen R, Ustinov J, Bouwens L, Eizirik DL, Hovi T,
Otonkoski T. 2000. Mechanism of coxsackievirus-induced damage to human pancreatic β-cells.
The Journal of Clinical Endocrinology and Metabolism 85:432–440.
Scott MR, Will R, Ironside J, Nguyen HO, Tremblay P, DeArmond SJ, Prusiner SB. 1999. Compel-
ling transgenetic evidence for transmission of bovine spongiform encephalopathy prions to
humans. Proceedings of the National Academy of Sciences 96:15137–15142.
Stuver S. 1998. Towards global control of liver cancer? Seminars in Cancer Biology 8:299–306.
Swedo SE, Leonard HL, Garvey M, Mittleman B, Allen AJ, Perlmutter S, Lougee L, Dow S, Zamkoff
J, Dubbert BK. 1998. Pediatric autoimmune neuropsychiatric disorders associated with strepto-
coccal infections: clinical description of the first 50 cases. American Journal of Psychiatry
155:264–271.
Yolken RH, Karlsson H, Yee F, Johnston-Wilson NL, Torrey EF. 2000. Endogenous retroviruses and
schizophrenia. Brain Research Reviews 31:193–199.
Appendix
C
Biosketches
194
APPENDIX C 195
DAVID ACHESON, M.D., is Chief Medical Officer at the Center for Food
Safety and Applied Nutrition, U.S. Food and Drug Administration. He received
his medical degree at the University of London. After completing internships in
general surgery and medicine, he continued his postdoctoral training in Manches-
ter, England, as a Wellcome Trust Research Fellow. He subsequently was a
Wellcome Trust Training Fellow in Infectious Diseases at the New England Medi-
cal Center and at the Wellcome Research Unit in Vellore, India. Dr. Acheson was
Associate Professor of Medicine, Division of Geographic Medicine and Infec-
tious Diseases, New England Medical Center until 2001. He then joined the fac-
ulties of the Department of Epidemiology and Preventive Medicine and Depart-
ment of Microbiology and Immunology at the University of Maryland Medical
School. Currently at the FDA, his research concentration is on foodborne patho-
APPENDIX C 197
World Bank, Asia Technical Environment Division, Ms. Carter-Foster has worked
on a wide variety of health, trade and environmental issues amassing in-depth
knowledge and experience in policy development and program implementation.
APPENDIX C 199
JAMES M. HUGHES, M.D., is the Director of the National Center for Infec-
tious Diseases at the Centers for Disease Control and Prevention (CDC) and an
Assistant Surgeon General in the Public Health Service. A board-certified physi-
cian in internal medicine, infectious diseases, and preventive medicine, Dr.
Hughes received his B.A. and M.D. from Stanford University in 1966 and 1971,
respectively. He completed his residency in internal medicine at the University of
Washington and a fellowship in infectious diseases at the University of Virginia.
Since joining CDC in 1973 as an Epidemic Intelligence Service officer, he has
worked primarily on foodborne disease and infection control in health care set-
tings. In 1992, Dr. Hughes became Director of the National Center for Infectious
Diseases, which is addressing domestic and global challenges posed by emerging
infectious diseases and the threat of bioterrorism. He is a member of the Institute
of Medicine and a fellow of the American College of Physicians, the Infectious
Diseases Society of America, and the American Association for the Advance-
ment of Science.
APPENDIX C 201
Emerging Infectious Diseases. He is also codeveloper and course leader for Sci-
ence, Politics, and Animal Health Policy. Dr. King received his B.S. and D.V.M.
degrees from The Ohio State University, and his M.S. degree in epidemiology
from the University of Minnesota. He has also completed the Senior Executive
Program at Harvard University, and received a M.P.A. from American Univer-
sity. Dr. King previously served on the Committee for Opportunities in Agricul-
ture, the Steering Committee for a Workshop on the Control and Prevention of
Animal Diseases, and the Committee to Ensure Safe Food from Production to
Consumption.
STEPHEN S. MORSE, Ph.D., is Director of the Center for Public Health Pre-
paredness at the Mailman School of Public Health of Columbia University, and a
faculty member in the Epidemiology Department. Dr. Morse recently returned to
Columbia after four years in government service as Program Manager at the De-
fense Advanced Research Projects Agency, where he co-directed the Pathogen
Countermeasures program and subsequently directed the Advanced Diagnostics
program. Before coming to Columbia, he was Assistant Professor of Virology at
The Rockefeller University in New York, where he remains an adjunct faculty
member. Dr. Morse is the editor of two books, Emerging Viruses (Oxford Uni-
versity Press, 1993; paperback, 1996) and The Evolutionary Biology of Viruses
(Raven Press, 1994); the former was selected by American Scientist as one of the
“100 Top Science Books of the 20th Century.” Dr. Morse serves as a Section
Editor of the CDC journal Emerging Infectious Diseases and was formerly an
Editor-in-Chief of the Pasteur Institute’s journal Research in Virology. As the
chair and principal organizer of the 1989 Conference on Emerging Viruses held
by the National Institute for Allergy and Infectious Disease, National Institutes of
Health, he coined the term and concept of emerging viruses and infections. He
was a member of the joint Institute of Medicine (IOM)–National Academy of
Sciences’ Committee on Emerging Microbial Threats to Health, chaired its task
force on viruses, and contributed the committee’s report, Emerging Infections
(1992). He also was a member of the IOM Committee on Xenograft Transplanta-
tion. Dr. Morse has been an adviser to the World Health Organization, the Pan-
American Health Organization, the U.S. Food and Drug Administration, the De-
fense Threat Reduction Agency, and other federal agencies. He is a Fellow of the
New York Academy of Sciences and a past Chair of its Microbiology Section. He
was the founding Chair of ProMED, the nonprofit international Program to Moni-
tor Emerging Diseases, and was an originator of ProMED-mail, an international
network inaugurated by ProMED in 1994 for outbreak reporting and disease
monitoring using the Internet. At present, he serves on the Steering Committee of
the Institute of Medicine’s Forum on Microbial Threats. Dr. Morse received his
Ph.D. from the University of Wisconsin–Madison.
APPENDIX C 203
with three children. His personal interests are in military history, photography,
automobile racing and exploring the wilderness of the American West.
GARY A. ROSELLE, M.D., received his M.D. from Ohio State University
School of Medicine in 1973. He served his residency at Northwestern University
School of Medicine and his Infectious Diseases fellowship at the University of
Cincinnati School of Medicine. Dr. Roselle is the Program Director for Infectious
Diseases for the Department of Veterans Affairs Central Office in Washington,
D.C., as well as the Chief of the Medical Service at the Cincinnati Veterans Affairs
Medical Center. He is a professor of medicine in the Department of Internal Medi-
cine, Division of Infectious Diseases at the University of Cincinnati College of
Medicine. Dr. Roselle serves on several national advisory committees. In addi-
tion, he is currently heading the Emerging Pathogens Initiative for the Depart-
ment of Veterans Affairs. Dr. Roselle has received commendations from the
Cincinnati Medical Center Director, the Under Secretary for Health for the
Department of Veterans Affairs, and the Secretary of Veterans Affairs for his
work in the infectious diseases program for the Department of Veterans Affairs.
He has been an invited speaker at several national and international meetings, and
has published more than 80 papers and several book chapters.
APPENDIX C 205
SPEAKERS
KATHRYN M. CARBONE, M.D., is the Acting Associate Director for Re-
search at FDA’s Center for Biologics Evaluation and Research (CBER) and leads
Virus Vaccine Neurovirulence Test Development in CBER’s Laboratory of Pedi-
atric and Respiratory Viral Diseases. She is also an Associate Profesor at Johns
Hopkins University School of Medicine and an Adjunct Professor of Medicine at
George Washington University. Dr. Carbone graduated magna cum laude from
Harvard–Radcliffe College in 1979 and graduated with honors from the Univer-
sity of Wisconsin School of Medicine in 1983. She completed internal medicine
and subspecialty training in infectious diseases at Johns Hopkins Hospital, re-
ceiving her board certifications in 1986 and 1988. Upon joining the faculty of the
Johns Hopkins School of Medicine in 1988, she continued studying virus infec-
tions of the brain with a special focus on multidisciplinary studies of the develop-
ing rat nervous system with Borna disease virus. She has continued these studies
and also has investigated the neurovirulence of vaccines for viral diseases such as
mumps since joining CBER in 1996 as Chief of the Laboratory of Pediatric and
Respiratory Viral Diseases.
APPENDIX C 207
was Henderson Inventor of the Year in 1997 for his new glutamine derivative-
based ORNT (oral rehydration and nutrition therapy). Guerrant was named Pro-
fessor Honoris Causa at UFC and received the Emilio Ribas Medal of the Brazil-
ian Society of Infectious Diseases in 1997. He has served on several editorial and
USDA and WHO advisory boards, VA and NIH Study Sections, Clark and Child
Health Foundation Boards, chaired the U.S. Cholera Panel of the U.S.–Japan
Cooperative Medical Science Program, and the International Affairs Committee
of the Infectious Diseases Society of America.
APPENDIX C 209
WILLIAM MASON, Ph.D., Senior Member at the Fox Chase Cancer Center,
joined in 1973 following a postdoctoral fellowship in retrovirology in the labora-
tory of Dr. Peter K. Vogt, at the University of Southern California. He began
working on hepatitis B in 1980 following the discovery of duck hepatitis B virus
by Dr. Jesse Summers. This early work, carried out in collaboration with Sum-
mers and with John Taylor, led to the widespread adoption of this animal model
as the system for studying how these viruses replicate. One of the immediate
consequences of this work was the discovery by Summers and Mason that hepa-
titis B viruses replicate by reverse transcription, like the retroviruses, and the
development of a detailed model for this process, again with Summers and Tay-
lor. The duck virus also served as an early and continuing model for the evalua-
tion of antiviral therapies. Since 1990, Dr. Mason’s lab has studied how hepatitis
B viruses maintain a chronic infection, and the effects of antiviral agents such as
lamivudine and L-FMAU on this process. This work has employed the duck
model of chronic infection, as well as the woodchuck model, which had been
discovered by Summers in the 1970s. Current work in Mason’s laboratory is
focused on the consequences of combining drug therapy with immunotherapy to
stimulate the host’s defenses against infected liver cells, and microarray technol-
ogy to evaluate the progression of chronic infections.
APPENDIX C 211
lion, two year research program on innate immunity funded by the Defense Ad-
vanced Research Projects Agency (DARPA), and has a long track record of ex-
tramural funding from the National Institutes of Health. David serves on a num-
ber of corporate boards and advisory councils including the Boards of Directors
for Virologic and ASM resources, and science advisory boards for IDI, the Burrill
and Company Life Sciences Investment Funds, and the Mayo Clinic Clinical
Research Center. He has authored 213 peer-reviewed articles and book chapters,
has served as Editor-in-Chief of 2 books, and is listed as an inventor on 21 issued
or pending U.S. patents.
THOMAS C. QUINN, M.D., M.Sc., is Senior Investigator and Head of the Sec-
tion on International AIDS Research in the Laboratory of Immunoregulation at
the National Institute of Allergy and Infectious Diseases. Since 1981, he has been
assigned to the Division of Infectious Diseases at Johns Hopkins University
School of Medicine where he is a Professor of Medicine. He also has adjunct
appointments in the Department of International Health, and the Department of
Immunology and Molecular Microbiology in The Johns Hopkins School of Hy-
giene and Public Health. He currently directs the Johns Hopkins School of Medi-
cine P3 HIV/AIDS Research Facility and the International STD Research Labo-
ratory. Dr. Quinn’s investigations have involved the study of the epidemiologic,
virologic, immunologic features of HIV infection in Africa, the Caribbean, South
America and Asia. In 1984, he helped establish the interagency project called
“Project SIDA” in Kinshasa, Zaire which was the largest AIDS investigative
project in sub-Saharan Africa. Dr. Quinn has been involved in laboratory investi-
gations which have helped define the biological factors involved in heterosexual
transmission and perinatal transmission, the natural history of HIV infections in
APPENDIX C 213
ROBERT YOLKEN, M.D., graduated from Harvard College and Harvard Medi-
cal School and did a residence in Pediatrics at Yale. He received Fellowship
training at Cornell–New York Hospital and at the Laboratory of Infectious Dis-
eases at the National Institutes of Health. He joined the faculty in the Department
of Pediatrics in 1979 and is currently the Ted and Vada Stanley Distinguished
Professor of Developmental Neurovirology in that Department. His research in-
terests include diagnostic virology and the identification of infectious causes of
APPENDIX C 215
chronic diseases. Since 1995 he has worked extensively on studies related to the
etiology of human neuropsychiatric diseases such as schizophrenia and bipolar
disorder. He is the author or coauthor of more than 200 publications in peer re-
viewed journals and is one of the coeditors of the Manual of Clinical Microbiol-
ogy. He has received numerous awards including the Abbott Award for the Rapid
Diagnosis of Human Diseases, the Wellcome Diagnostics Award, and the Mead
Johnson Award for Pediatric Research.
FORUM STAFF
STACEY L. KNOBLER, is Director of the Forum on Microbial Threats at the
Institute of Medicine (IOM). She previously served as the codirector of the IOM
Board on Global Health’s study, Neurological, Psychiatric, and Developmental
Disorders in Developing Countries (2001), and as the research associate for the
Assessment of Future Scientific Needs for Live Variola Virus (1999). Ms. Knobler
is actively involved in program research and development for the Board on Global
Health. Previously, she held positions as a Research Associate at the Brookings
Institution’s Foreign Policy Studies Program and as an Arms Control and
Democratization Consultant for the Organization for Security and Cooperation in
Europe in Vienna and Bosnia-Herzegovina. Ms. Knobler has also worked as a
research and negotiations analyst in Israel and Palestine. She is currently a mem-
ber of the CBACI Senior Working Group for Health, Security, and U.S. Global
Leadership. Ms. Knobler has conducted research and coauthored published
articles on biological and nuclear weapons control, foreign aid, health in develop-
ing countries, poverty and public assistance, and the Arab–Israeli peace process.
MARJAN NAJAFI, M.P.H., was the research associate for the Forum on
Microbial Threats in the Board on Global Health from March 2001 to November
2003. She also worked with the IOM committee that produced Veterans and Agent
Orange: Update 2000. Ms. Najafi received her undergraduate degrees in chemi-
cal engineering and applied mathematics from the University of Rhode Island.
Subsequently, she served as a public health engineer with the Maryland Depart-
ment of Environment and, later, with the Research Triangle Institute in North
Carolina. After earning a master’s of public health from the Bloomberg School of
Public Health at Johns Hopkins University, she managed a lead-poisoning pre-
vention program in Micronesia, funded by a grant from the U.S. Department of
Health and Human Services. She also studied the effects of cellular phone radia-
tion on human health.