You are on page 1of 11

REVIEWS

EPITOPE SPREADING IN IMMUNE-


MEDIATED DISEASES: IMPLICATIONS
FOR IMMUNOTHERAPY
Carol L. Vanderlugt and Stephen D. Miller
Evidence continues to accumulate supporting the hypothesis that tissue damage during an
immune response can lead to the priming of self-reactive T and/or B lymphocytes, regardless
of the specificity of the initial insult. This review will focus primarily on epitope spreading at the
T-cell level. Understanding the cellular and molecular basis of epitope spreading in various
chronic immune-mediated human diseases and their animal models is crucial to
understanding the pathogenesis of these diseases and to the ultimate goal of designing
antigen-specific treatments.

CRYPTIC EPITOPE A typical immune response against a self or foreign pro- In most of the models to be discussed, epitope
A cryptic epitope is defined as a tein is usually focused on one or two epitopes within spreading initiated as a result of tissue damage is
hidden or sequestered epitope that protein, which are termed dominant. Epitope thought to play an active role in ongoing disease
that is processed and presented spreading was initially defined as the diversification of pathology. However, there is some evidence that
more efficiently as a result of an
inflammatory immune response
epitope specificity from the initial focused, dominant epitope spreading is an important component of
initiated by either a dominant epitope-specific immune response to subdominant protective immune responses, acting to enhance the
epitope, as in a response to an epitopes on that protein1,2. The hierarchy of dominant efficiency of the immune response, as seen in tumour
infectious agent, or revealed as a and CRYPTIC EPITOPES in this scenario is thought to be due clearance, and as a mechanism to downregulate
result of the diversification of
the response secondary to self
to a combination of differential protein processing and immune responses, such as those occurring in
tissue damage, as in an presentation by various antigen-presenting cell popula- autoimmunity.
autoimmune response. tions, and also to the availability of epitope-specific At present, most treatment regimens for autoim-
T cells, taking into consideration central and peripheral mune diseases like multiple sclerosis, type-1 diabetes
tolerance mechanisms3. and myasthenia gravis rely on generalized suppression
Our laboratory and others, extending this view, of the immune system and/or neutralization of
have studied epitope spreading specifically in the con- inflammatory mediators. It is, however, desirable to
text of chronic tissue damage, which can lead to design antigen-specific forms of immunotherapy
autoimmunity. In these studies, regardless of the initial that leave the immune system intact and able to neu-
Department of antigenic stimulus — be it viral, graft rejection or tralize opportunistic pathogens. Mounting evidence
Microbiology-Immunology autoimmune — the specificity of the immune for a pathological role for epitope spreading in
and Interdepartmental response spreads to include self epitopes other than chronic autoimmune disease makes the development
Immunobiology Center,
Northwestern University that which initiated the inflammatory process (FIG. 1). of epitope-specific therapies problematic4. Therefore,
Medical School, In this scenario, infection, organ transplantation or a more complete understanding of the cellular and
303 E. Chicago Avenue, autoimmunity (organ-specific or systemic) leads to tis- molecular basis of the epitope spreading phenomenon
Chicago, IL 60611, USA sue damage. The resulting inflammation and tissue is required to promote the development of antigen-
Correspondence to S.D.M.
e-mail: s-d-miller@
damage then primes a hierarchical cascade of autoreac- specific treatments for human autoimmune diseases,
northwestern.edu tive T-cell specificities, even allowing cryptic or more efficient vaccines and more effective methods to
DOI: 10.1038/nri724 sequestered epitopes to be processed and presented. control transplant rejection.

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | FEBRUARY 2002 | 8 5


© 2002 Macmillan Magazines Ltd
REVIEWS

a Primary epitope Secondary epitope


MHC II TCR MHC II TCR

APC TH 1 APC TH 1

CD28
Peripheral CD80/86
lymphoid tissue b Proliferation and differentiation j

Blood
vessel
Mono TH 1 TH 1

c Adhesion and penetration

Target
d i
tissue Chemokines
IFN-γ
MIP-1α Tissue
TH 1 APC debris

h
e
LT/TNF-β
Cytokines Tissue
IFN-γ g destruction
Mono
IFN-γ
MIP-1α Phagocytosis
O2 radicals, NO
Proteolytic enzymes
TNF-α
f Activation
Macrophage

Figure 1 | Epitope spreading in autoimmune and virus-induced tissue immunopathology. Presentation of the primary
epitope (the immunodominant self or viral epitope) occurs in peripheral lymphoid tissue (a), resulting in activation and differentiation
of autoreactive TH1 cells (b). The activated TH1 cells migrate (c) into the target tissue, where they encounter antigen presented by
resident APCs. (d). After antigen restimulation, the pathologic TH1 cells release a cascade of chemokines and cytokines (e), leading
to recruitment of additional mononuclear phagocytes from the peripheral blood, which are activated along with resident APCs (f).
Activated mononuclear cells then lead to bystander tissue destruction (g) via phagocytic mechanisms and release of TNF-α,
proteolytic enzymes, NO and O2 radicals. The tissue debris (h) is processed and presented on resident and peripheral APCs (i),
leading to the activation and differentiation of a second wave of TH1 cells (j), which can re-enter the tissue and cause additional
tissue destruction. APC, antigen presenting cell; IFN-γ, interferon-γ; LT/TNF-β, lymphotoxin/tumour necrosis factor β; MIP-1α,
macrophage inflammatory protein 1α; MHC II, major histocompatibility complex class II; Mono, monocyte; TCR, T-cell receptor;
TH1, T helper cells type 1 (see glossary).

Epitope spreading in autoimmune disease define. Animal models have therefore been useful
Characterization of the cellular and molecular basis of because the peptide specificity of the initial immune
epitope spreading in various chronic immune-mediated response can be manipulated, genetically identical ani-
diseases and disease models is extremely important both mals can be used and the immune response can be
to understanding the pathogenesis of those diseases and serially assessed in different peripheral lymphoid
to achieving the ultimate goal of designing antigen- organs, as well as in the target tissue of the disease.
specific treatments for human autoimmune diseases.
Although immune responses are complex, involving Autoimmune models of multiple sclerosis. Multiple scle-
both humoral and cellular immune components, rosis is a human autoimmune demyelinating disease
some autoimmune diseases are predominately CD4+ thought to be CD4+ T-cell mediated and to be charac-
T-cell mediated, whereas others seem to be primarily terized by mononuclear cell infiltration into the central
antibody mediated. We will focus on the role of epi- nervous system (CNS). Autoimmune and virus-induced
tope spreading in T-cell-mediated autoimmune dis- animal models of multiple sclerosis have yielded many
eases. Although we present some intriguing human insights into the possible mechanisms involved in the
studies, a pathological role for epitope spreading is human disease. The study of experimental autoimmune
very difficult to verify in human disease because the encephalomyelitis (EAE), which is induced by priming
initiating antigenic specificity is usually impossible to the animal with myelin antigens — including myelin

86 | FEBRUARY 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd
REVIEWS

T HELPER TYPE 1 (TH1) basic protein (MBP), proteolipid protein (PLP), myelin It is also interesting that the hierarchical order of
CD4+ T cells have been divided oligodendrocyte glycoprotein (MOG) and myelin pep- epitope spreading during relapsing EAE in the SJL
into at least two distinct types. tides — led to the initial description of epitope spread- mouse correlates with the size of the T-cell repertoire,
TH1 cells produce IFN-γ,
ing1,2. EAE has been an excellent model in which to with the pattern of spreading proceeding from the
lymphotoxin and TNF-α, and
mediate macrophage study this process because in this model disease is easily most to the least immunodominant epitope (that is,
inflammatory responses such as induced by a defined peptide, and the changing speci- PLP139–151>PLP178–191>MBP84–104)10. In the SJL mouse, it
delayed-type hypersensitivity ficity of the immune response to endogenous myelin has long been known that the response to the PLP139–151
(DTH). Demyelination in epitopes after disease initiation and subsequent tissue epitope is dominant in EAE induced by mouse spinal
multiple sclerosis models is
thought to be due to TH1 cells.
damage can be followed over time. In addition, EAE cord homogenate11. This is in agreement with recent
TH2 cells produce IL-4, IL-10 presents as either an acute, relapsing–remitting or findings of a very high precursor frequency of
and/or TGF-β, and can chronic disease (as does multiple sclerosis), depending PLP139–151-reactive T cells in the periphery of naive SJL
downregulate TH1 responses. on the mouse strain used and the priming myelin epi- mice. This high precursor frequency is partly due to
topes (TABLE 1). Multiple investigators have characterized lack of thymic deletion to PLP139–151, because an alter-
INTRAMOLECULAR EPITOPE
SPREADING a hierarchical order of epitope spreading in various EAE native isoform of PLP (DM20, which lacks residues
Spreading from one epitope to systems and have shown a pathological role for this 116–150) is more abundantly expressed in the thymus
another on the same molecule, process in chronic disease progression3,5–9. than is full-length PLP12. Even though other encephali-
for example, from PLP139–151 to Our laboratory has extensively characterized the role togenic epitopes such as PLP178–191 and MBP84–104 bind
PLP178–191.
of epitope spreading in relapsing EAE initiated by prim- the SJL major histocompatibility complex (MHC) class
INTERMOLECULAR EPITOPE ing SJL mice with the immunodominant PLP epitope, II molecule with efficiency close to that of PLP139–151,
SPREADING PLP139–151. In this model, PLP139–151-specific CD4+ T-cell induction of EAE with any myelin epitope other than
Spreading of the specificity of an reactivity is induced within 3 days of priming and is PLP139–151 invariably leads to spreading first to
immune response from an
maintained throughout the disease course. Immediately PLP139–151-specific T cells9,10.
epitope on one molecule to one
on a different molecule is termed before, and continuing during, the first relapse, Some circumstantial evidence that epitope spreading
intermolecular epitope PLP178–191 reactivity (INTRAMOLECULAR EPITOPE SPREADING) is occurs in human autoimmune disease is presented later
spreading. An example would be detected by T-cell proliferation and delayed-type hyper- in this review. If epitope spreading proves to be an inte-
the spread in EAE induced with sensitivity (DTH) assays, and during the second relapse, gral part of chronic human autoimmune disease, pep-
PLP139–151, an epitope on
proteolipid protein, to an
MBP84–104 responses (INTERMOLECULAR EPITOPE SPREADING) tide-specific therapies may be problematic, as the speci-
epitope on myelin basic protein, are identified (FIG. 2a). The development of these ficity of the pathogenic response would change over
such as MBP84–104. responses has been shown to correlate with the extent time. However, full T-cell activation, and therefore func-
of myelin destruction during the acute disease phase, tional epitope spreading, requires co-stimulatory signals
and T cells isolated from the CNS of sick mice prolifer- such as those provided by CD28–CD80/86 and
ate to the spread epitopes PLP178–191 and MBP84–104. CD154–CD40 interactions. In this regard, short-term
These T cells can also transfer disease to naive recipi- blockade of CD28–CD80 co-stimulation using anti-
ents9. More convincingly, peptide-specific tolerance to CD80 F(ab) fragments administered after recovery
the relapse-associated epitopes blocks disease progres- from acute PLP139–151-induced relapsing EAE resulted in
sion, even though peripheral T-cell responses against a striking reduction in relapse incidence13. This protec-
the disease-inducing peptide PLP139–151 remain intact10. tion was long lasting, and worked efficiently even when
For example, PLP178–191-specific tolerance induction treatment began after the first relapse. Importantly,
during remission from PLP139–151-induced EAE reduced CD28 blockade seemed to lead to specific tolerance of
the relapse incidence from 80% in controls to 20% in T-cell responses to the relapse-associated PLP178–191 epi-
tolerant mice. tope10. For example, when administered before the first
relapse, anti-CD80 F(ab) therapy resulted in PLP178–191-
specific tolerance and protection from disease relapse,
Table 1 | Clinical disease in induced mouse models of multiple sclerosis even though peripheral T HELPER TYPE 1 (T 1) responses to
H

the initiating PLP139–151 epitope were unaffected.


Mouse strain Inducing epitope Disease pattern
Similarly, we have shown that short-term blockade of
SJL PLP139–151 Relapsing–remitting*
the CD40–CD154 interaction using a monoclonal anti-
PLP178–191 Relapsing–remitting* CD154 antibody administered either at the peak of
MBP84–104 Relapsing–remitting* acute disease, or during remission from acute disease,
(SJLxSWR)F1 PLP139–151 Relapsing–remitting* significantly inhibited disease relapse concomitant with
B10.PL MBP1–11 Monophasic‡ suppression of T-cell reactivity to the relapse-associated
PLP178–191 Relapsing-remitting
myelin epitopes14. These results indicate that epitope
spreading plays a major role in EAE progression in SJL
C57BL/B6 MOG35–55 Monophasic/chronic§
mice, and short term co-stimulatory blockade can
PLP178–191 Monophasic‡ specifically inhibit initiation of T-cell responses to the
*Relapsing–remitting disease is characterized by an acute paralytic episode, followed by a significant spread epitope and expression of clinical relapses.
clinical recovery (remission) period and, later, a significant worsening of clinical symptoms. There can
be multiple relapses. Interestingly, treatment of SJL mice during ongoing

Monophasic disease is characterized by an acute paralytic episode followed by complete recovery relapsing EAE using intact antibodies or F(ab) frag-
from clinical symptoms. ments specific for the downregulatory co-stimulatory
§
Chronic disease is characterized by an acute paralytic episode followed by stabilization or, often, a
gradual worsening of disease symptoms. molecule cytotoxic T-lymphocyte-associated protein 4
MBP, myelin basic protein; MOG, myelin oligodendrocyte glycoprotein; PLP, proteolipid protein. (CTLA-4) led to exacerbated disease progression and

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | FEBRUARY 2002 | 8 7


© 2002 Macmillan Magazines Ltd
REVIEWS

a Acute phase Primary relapse Secondary relapse b Disease onset Early chronic Late chronic

Anti-PLP139–151 Anti-PLP178–191 Anti-MBP84–104 Anti-TMEV Anti-TMEV Anti-TMEV


Anti-PLP139–151 Anti-PLP139–151
Anti-PLP178–191
Anti-PLP56–70

Clinical severity

Clinical severity
Anti-MOG92–106
Anti-MBP84–104

10 20 30 40 50 60 70 20 40 60 80 100 120 140


Days post-induction Days post-infection
PLP139–151 TMEV
Primed

Figure 2 | Hierarchical pattern of intramolecular and intermolecular epitope spreading in PLP139–151-induced relapsing
EAE and Theiler’s virus-induced demyelinating disease (TMEV-IDD). a | The relapsing–remitting clinical course and pattern
of epitope spreading in relapsing-EAE in the SJL mouse induced by priming with the immunodominant PLP139–151 epitope. Acute
disease is mediated by PLP139–151-specific TH1 cells, which are then downregulated, resulting in disease remission. Myelin
destruction during the acute clinical episode leads to the priming of T cells to a secondary epitope, PLP178–191 (intramolecular
epitope spreading), which mediates the primary relapse. After regulation of these cells, a third wave of TH1 cells is induced to the
lesser immunodominant MBP84–104 epitope (intermolecular epitope spreading), which mediates the secondary relapse. b | The
clinical course and pattern of epitope spreading in TMEV-IDD in the SJL mouse. Disease onset is initiated by TMEV-specific CD4+
T cells, which arise several days after intracerebral infection and release proinflammatory cytokines upon encountering virus
epitopes presented by persistently infected antigen-presenting cells in the CNS. These virus-specific responses persist throughout
the chronic disease course. Myelin debris released as a result of the initial tissue damage leads to the induction of CD4+ TH1
responses against myelin epitopes, which occurs in a hierarchical order, beginning with responses to the immunodominant
PLP139–151 epitope and then progressing to less-dominant myelin epitopes. MBP, myelin basic protein; MOG, myelin
oligodendrocyte glycoprotein; PLP, proteolipid protein; TMEV, Theiler’s murine encephalomyelitis virus.

enhanced T-cell reactivity to both inducing and relapse- was found to be associated with a humoral response to
associated epitopes15,16. This indicates that normally, MOG, a myelin protein not included in the inducing
CTLA-4-mediated signalling negatively regulates the MP4 fusion protein25.
dynamic spread of autoreactive T-cell responses during Although more difficult to prove, there is evidence
the course of autoimmune disease. that epitope spreading occurs in human multiple sclero-
Relapsing EAE induced in (SWR×SJL)F1 mice has sis. Recently, Tuohy et al. followed T-cell responses over
also been extensively characterized. The immune several years in three IMDS (ISOLATED MONOSYMPTOMATIC
response to the inducing peptide PLP139–151 declines with DEMYELINATING SYNDROME) patients, showing that T-cell
time, whereas responses to new myelin epitopes autoreactivity to particular PLP epitopes decreased with
ISOLATED MONOSYMPTOMATIC
DEMYELINATING SYNDROME
emerge5,17. Interestingly, interferon-β (IFN-β) treat- time after diagnosis. Interestingly, when two of these
(IMDS). IMDS is a group of ment, which reduces disease relapses in these mice and patients progressed to clinically definite multiple sclero-
distinct clinical disorders often in multiple sclerosis patients18,19, inhibits epitope spread- sis, autoreactivity to PLP peptides other than those first
associated with eventual ing in the mouse model20. Responses to PLP139–151 after observed appeared17,26,27. Goebeles et al. studied five
progression toward clinically
IFN-β treatment are essentially unchanged, with one multiple sclerosis patients by isolating MBP-specific
definite multiple sclerosis.
important exception. Splenocytes from mice treated T-cell lines at five time points. Specificity was deter-
TCR Vβ CDR3 SPECTRATYPING with IFN-β and activated ex vivo with PLP139–151 pro- mined using a panel of overlapping peptides and rep-
Polymerase chain reaction- duced significantly more interleukin-10 (IL-10) and less ertoire analysis was done by TCR Vβ CDR3 SPECTRATYPING.
based method of identifying IL-12 than control mice, indicating that IFN-β may It was concluded that a pattern of a focused, stable epi-
pseudoclonal TCR usage by
analyzing Vβ family gene usage.
reduce the relapse rate by creating a TH2-like environ- tope response is the exception rather than the rule in
In independent reactions, ment in which epitope spreading is blocked20. IL-10 progressive multiple sclerosis28.
Vβ–Cβ products across the inhibits expression of proinflammatory cytokines21,22,
CDR3 region are amplified from MHC class II molecules23 and co-stimulatory24 mole- Virally induced models of multiple sclerosis. Viruses
cDNA, tagged with a
cules by antigen-presenting cells (APCs). These proper- and other infectious insults are implicated in the aeti-
fluorochrome, and resolved on a
polyacrylamide gel ties would probably limit tissue destruction and the abil- ology of many human autoimmune diseases, includ-
electrophoresis gel. Expanded ity of APCs to activate T cells that are specific to new ing multiple sclerosis 29. There are various mecha-
pseudoclonal Vβ–Cβ products antigens; that is, epitope spreading. nisms by which infection can lead to the initiation of
of a single length are Epitope spreading has also been described by an autoimmune response. These include molecular
distinguished from other
Vβ–Cβ products by size
McFarland et al. in the nonhuman primate (marmoset) mimicry, bystander activation, release of cryptic epi-
differences introduced at the MP4-induced EAE model. MP4 is a fusion protein con- topes and epitope spreading (FIG. 3). Spreading from
coding junction. sisting of PLP and MBP. In this model, demyelination viral to self epitopes has been shown or suggested to

88 | FEBRUARY 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd
REVIEWS

play a pathological role in several virus-induced NOD mouse model of type 1 diabetes. Type 1 diabetes is
autoimmune disease models, whereas bystander acti- characterized by a chronic autoimmune response
vation and molecular mimicry are thought to be against islet β-cells, which begins early in life. Clinical
involved in other diseases (reviewed in REF. 30) (TABLE 2). diabetes, which is the result of the subsequent loss of
We have examined the role of epitope spreading in these cells and their ability to produce insulin, occurs
initiating anti-myelin autoimmune responses in the several years later37. INSULITIS — and subsequently dia-
chronic stages of Theiler’s murine encephalitogenic betes — develops spontaneously in the non-obese dia-
virus-induced demyelinating disease (TMEV-IDD). betic (NOD) mouse and is thought to be primarily
TMEV, a picornavirus and natural mouse pathogen, dependent on CD4+ TH1 responses. Much work has
induces a life-long persistent infection of CNS-resi- been done to characterize the specificity of T-cell
dent APCs, leading to a chronic–progressive CD4+ responses in NOD mice as diabetes develops38–44. The
T-cell-mediated demyelinating disease in susceptible 45-kd isoform of glutamic acid decarboxylase (GAD45)
mouse strains such as SJL/J. Myelin destruction is ini- is considered by many (although not all45) investigators
tiated by virus-specific CD4 + T cells, which target to be the initial target of CD4+ T-cell autoimmunity in
CNS-persistent virus31. Peripheral T-cell responses to this model of diabetes and early tolerance to this
myelin epitopes develop during progressive disease in response has been shown to block epitope spreading
a hierarchical order, beginning with the dominant and disease39.
PLP139–151 peptide (FIG. 2b), but only after the onset of Recent work in human diabetes has focused mainly
demyelination32,33. As disease progresses, responses to on humoral responses. Bonifacio et al. examined serum
various other less-dominant encephalitogenic myelin antibodies to insulin, GAD and protein tyrosine phos-
epitopes (such as PLP178–191, PLP56–70 and MOG92–106) phatase (islet antigen 2, IA-2) in offspring of diabetic
develop. Autoreactivity is initiated in part by the parents at various times, starting at birth. Two distinct
in vivo processing of myelin debris and subsequent autoimmune profiles were observed: a rapid disease
presentation of myelin epitopes by CNS-resident onset at the first appearance of an autoantibody
APCs34. CNS APCs harvested before any signs of response and a slower epitope spreading stage with a
demyelination and disease (<40 days after infection) later onset of disease. The early response in both sub-
present viral peptides, but not myelin peptides34. sets (at ~ 2–3 yr of age) was typically immunoglobulin
However, by 90 days post-infection, microglia and G1 (IgG1) and most commonly to insulin. IgG1 anti-
macrophages isolated from the spinal cords of diseased bodies to GAD and IA-2 often appeared simultane-
mice endogenously present both viral and myelin epi- ously or soon after 2–3 yr of age. In the group with
topes to T cell lines and hybridomas34. Borrow et al. also slower disease onset, these initial responses declined
showed that DTH responses to various myelin epitopes (due to antigen-specific regulation) and responses to
were activated during ongoing TMEV-IDD35. other antigens appeared46. These investigators also
Significantly, induction of tolerance to multiple examined epitope spreading, specifically in the anti-
encephalitogenic myelin epitopes, using the MP-4 GAD antibody response, from birth to disease onset in
fusion protein in SJL mice with ongoing TMEV-IDD, offspring of diabetic parents. The initial response usu-
attenuated disease progression and resulted in signifi- ally involved anti-GAD64 residues 96–444 and epitope
cantly less demyelination and decreased inflammatory spreading was evident in most offspring47. GAD-spe-
cell infiltration in the CNS36. These results support the cific epitope spreading was also seen in another large
hypothesis that tissue damage initiated by virus-specific study group of diabetic patients48.
T cells results in presentation of self antigens and acti- Interestingly, pancreas graft rejection in diabetic
vation of autoreactive T cells in the inflammatory CNS recipients (under generalized immune suppression)
environment, indicating a pathological role for epitope was associated with increased or sustained high levels
spreading in virus-induced autoimmune disease. of humoral islet autoimmunity and with epitope

Table 2 | Proposed mechanisms for virus-induced autoimmunity


Animal virus model Human autoimmune Proposed mechanism for References
disease virus-induced autoimmunity
Theiler’s murine encephalomyelitis Multiple sclerosis Epitope spreading 32
virus (TMEV)
Recombinant TMEV* Multiple sclerosis Molecular mimicry 84
Semliki forest virus (SFV) Multiple sclerosis Bystander activation, 85,86
molecular mimicry
Coxsackie B3 virus Myocarditis Bystander activation, 87,88
molecular mimicry
INSULITIS Coxsackie B4 Virus Diabetes Bystander activation 89
Inflammation surrounding the Mouse cytomegalovirus (MCMV) Myocarditis Bystander activation 87,88
insulin-producing β-cells in the
pancreas. Diabetes occurs when
Herpes simplex virus type 1 (HSV-1) Herpes stromal keratitis Bystander activation, 90,91
molecular mimicry
β-cells can no longer produce
adequate amounts of insulin. *Induced by infection with a TMEV variant engineered to express an epitope mimic of proteolipid protein residues 139–151.

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | FEBRUARY 2002 | 8 9


© 2002 Macmillan Magazines Ltd
REVIEWS

A Molecular mimicry B Epitope spreading

a Persistent
microbial infection

c Self-tissue d Self-tissue b Microbial


destruction e Self-tissue peptide
d Self-tissue destruction peptides
peptide mimic
Inflammatory
cytokines
Macrophage Macrophage sTH1
b Inflammatory TCR MHC II
MHC II TCR cytokines

APC mTH1 mTH1 APC


c
a

f Epitope spread from


Infectious microbial to self-peptides sTH1
agent
Peptide
MHC II TCR

mTH1

Macrophage
a Microbial
C Bystander activation D Cryptic antigens infection of
tissue

d IFN-γ

Increased Self-tissue f a Microbial


self-antigen destruction infection e Activation
presentation
f APC engulfs b Microbial
of APC self peptides peptides i Self-tissue
destruction
b Microbial
APC peptide
mTH1 APC
c Inflammatory c
cytokines
and chemokines
d Activation
of APC

h Inflammatory
mTH1 APC sTH1 cytokines

g Cryptic antigen
processed and
sTH1 e Recruitment presented
of self-reactive
T cells

Figure 3 | Mechanisms of infection-induced autoimmunity. After a microbial infection, activated microbe-specific TH1 (mTH1)
cells migrate to the infected organ. A | Molecular mimicry describes the activation of crossreactive TH1 cells that recognize both the
microbial epitope (mTH1) and the self epitope (sTH1) (a). Activation of the crossreactive T cells results in the release of cytokines and
chemokines (b) that recruit and activate monocytes and macrophages, which mediate self-tissue damage (c). The subsequent
release of self-tissue antigens and their uptake by APCs perpetuates the autoimmune disease (d). B | Epitope spreading involves a
persistent microbial infection (a) that causes the activation of microorganism-specific TH1 cells (b,c), which mediate self-tissue
damage (d). This results in the release of self peptides (e), which are engulfed by APCs and presented to self-reactive TH1 cells (f).
Continual damage and release of self peptides results in the spread of the self-reactive immune response to multiple self-epitopes
(f). C | Bystander activation is the nonspecific activation of self-reactive TH1 cells. Activation of microorganism-specific TH1 cells
(a,b) leads to inflammation (c,d) and results in the increased infiltration of T cells at the site of infection and the activation of
self-reactive TH1 cells by TCR-dependent and -independent mechanisms (e) Self-reactive T cells activated in this manner mediate
self-tissue damage and perpetuate the autoimmune response (f). D | Cryptic antigen model describing the initiation of
autoimmunity by differential processing of self peptides. Following microbial infection (a) IFN-γ is secreted by both activated
microbe-specific TH1 cells (b,c) and microbe-infected tissue cells (d). This activates APCs (e) and can lead to APC engulfing
self-antigens (f). Cytokine activation of APCs can induce increased protease production and different processing of captured
self-antigens, resulting in presentation of cryptic epitopes. The presentation of these cryptic epitopes can activate self-reactive
TH1 cells (g), leading to self-tissue destruction (h,i). APC, antigen-presenting cell; MHC II, major histocompatibility complex class II;
TCR, T-cell receptor. Modified with permission from REF. 30.

90 | FEBRUARY 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd
REVIEWS

spreading, but not with the levels of GAD- or IA-2-spe- synovium- or cartilage-associated antigens has been
cific serum antibody seen before the allotransplanta- reported, along with reactivity against microbial and self
tion49. This implies that recurrent autoimmunity, along HEAT-SHOCK PROTEINS (HSPs), which are upregulated by
with epitope spreading, leads to transplant rejection in cells under stress and released by necrotic cell death.
diabetic patients. Adjuvant arthritis, a model of rheumatoid arthritis55,56,
is induced in the Lewis rat by injection of COMPLETE
Models of myasthenia gravis. Myasthenia gravis is char- FREUND’S ADJUVANT, which contains heat-killed
acterized by the presence of anti-acetylcholine receptor Mycobacterium tuberculosis. There is evidence to suggest
(AChR) antibodies, which leads to a decreased number that epitope spreading has a role in the pathogenesis of
of AChRs at the neuromuscular junction, impaired adjuvant arthritis. Moudgil et al. found that the initial
neuromuscular transmission and muscle weakness. T-cell response towards the 65-kDa HSP of M. tubercu-
Experimental autoimmune myasthenia gravis (EAMG) losis was focused on epitopes in the middle of this pro-
can be induced in animals by immunization with tein, with some reactivity near the N-terminus. By 8–10
AChR, leading to AChR-specific T-cell responses and weeks after disease onset, reactivity to peptides towards
antibody production50. In rabbits, after induction of the C-terminus of Mycobacterial hsp65 was evident,
disease by immunization with three human extracel- and tolerance induced by pretreatment with these
lular peptides, serum antibodies to native AChR are C-terminal peptides in synthetic adjuvant blocked
present and bind strongly to rabbit, rather than to arthritis induction57,58. Epitope spreading in this sce-
human, AChR50,51. This suggests that endogenous pre- nario may be secondary to tissue damage, as the spread
sentation of rabbit AChR — which has recently been epitopes represent conserved sequences shared with rat
reported to be mediated by myoblasts52 — during the Hsp60. Therefore, epitope spreading might have a pro-
initial anti-human AChR immune response leads to tective outcome in this setting. Similarly, synovial T cells
epitope spreading. taken just before disease remission from patients with
In another model of EAMG, in which C57BL/6 juvenile chronic arthritis or rheumatoid arthritis show
mice are immunized with AChR, anti-CTLA-4 anti- enhanced responses to self Hsp65 (REF. 59) and are a
body treatment resulted in severe EAMG — which had good indicator of a favourable prognosis60,61. Further
a rapid onset — enhanced T-cell responses to AChR evidence of epitope spreading in rheumatoid arthritis
and increased anti-AChR-antibody production. Anti- comes from a study by Alam et al., who sequenced more
CTLA-4 antibody is thought to block a downregulatory than 650 T-cell receptor (TCR) Vβ D–J junctional
signal and therefore increase any ongoing T-cell regions from synovial and peripheral blood mononu-
response. Interestingly, mice immunized with the clear cells of two patients with rheumatoid arthritis.
immunodominant peptide α(146–162) (an extracellu- Their results indicate that a dynamic TCR selection
lar sequence of the AchR) and treated with anti-CTLA-4 process takes place during disease progression62.
showed symptoms of clinical EAMG. Diversification of
the autoantibody repertoire and enhanced T-cell prolif- Epitope spreading in organ allograft rejection
eration against not only the immunizing α(146–162) Acute transplant rejection is thought to be the result of
peptide, but also other subdominant epitopes, was also direct cytotoxic T lymphocyte (CTL) recognition of
evident. Control mice did not show any signs of clinical MHC class I molecules expressed on donor APCs,
disease, indicating that treatment with anti-CTLA-4 because elimination of passenger dendritic cells from
antibody augmented the initial α(146–162)-specific the organ substantially reduces transplant rejection. By
immune response and led to epitope spreading in both contrast, indirect helper CD4+ T-cell recognition of
the T- and B-cell components of this pathological recipient MHC class II molecules plus alloantigenic
autoimmune response53. peptide (transplanted tissue processed and presented by
Human myasthenia gravis — both early and late host APCs) is most likely to be responsible for chronic
onset — is a complex disease that is often characterized rejection63. In human studies, onset of acute rejection in
by thymic pathologies such as hyperplasia, atrophy or heart transplant recipients was shown to correlate with
thymoma. Several studies have suggested heterogeneity T-cell responses to a single dominant epitope on one
HEAT-SHOCK PROTEIN of T-cell responses against epitopes on the α-, γ- and alloantigen64,65. However, in allograft recipients with
Heat-shock proteins are δ-subunits of AChR; however, much work remains to recurring episodes of rejection or in patients in which
expressed in all cells, including
microbes, when they are
be done50. In addition to T-cell and antibody epitopes chronic rejection was at its onset, recipient T-cell reac-
stressed; for example, when they on AChR, antibodies against striated muscle antigens tivity could spread to other epitopes within the allo-
experience high temperatures. such as titin are characteristic of generalized myasthe- geneic MHC molecule, as well as to other alloantigens
These proteins can then become nia gravis54. This suggests that the immune response expressed by graft tissue. In a recent study, recipients of
targeted by an immune
in myasthenia gravis is not focused on a dominant heart allografts were monitored for reactivity against
response.
epitope. However, studies in patients with the disease donor human leukocyte antigen (HLA)-DR peptides
COMPLETE FREUND’S ADJUVANT in which responses are monitored over time will be before and up to 36 months after transplantation64.
Used to trigger an immune necessary to determine whether there is evidence of In patients who received a doubly mismatched trans-
response to proteins or peptides epitope spreading. plant (HLA-DR disparity at both loci), chronic rejection
emulsified in the adjuvant; it
consists of freeze-dried
was significantly more likely when intermolecular epi-
Mycobacterium, emulsifying Models of arthritis. Arthritis is an inflammatory disease tope spreading occurred than if reactivity was directed
agents and mineral oil. of the joint synovium. T-cell reactivity towards various only to the initially targeted HLA-DR molecule65.

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | FEBRUARY 2002 | 9 1


© 2002 Macmillan Magazines Ltd
REVIEWS

Implications for immunotherapy is a surprising lack of reports in which bystander sup-


Substantial evidence from studies of numerous animal pression is used to treat established disease in animal
models and of various human diseases supports the models of autoimmunity, so the efficacy of this
hypothesis that tissue damage, regardless of the initiat- approach in the light of ongoing chronic inflammation
ing event, can lead to epitope spreading, which can then is not known. As a cautionary note, a recent clinical trial
contribute to ongoing disease. Chronic or persistent in multiple sclerosis that employed an APL of the HLA-
infections may trigger, sustain or exacerbate autoim- DR2-restricted MBP83–99 epitope led to disease exacerba-
mune disease through epitope spreading via antigen tions in a significant number of patients, in spite of evi-
presentation of epitopes released within injured tis- dence that TH2 responses had been induced73. This may
sue30,66,67. The initial focused immune attack on organ indicate that TH2 cells can contribute to organ-specific
allografts also spreads to include new T- and B-cell reac- autoimmunity in a pre-existing inflammatory setting in
tivity, leading to chronic graft rejection. However, which the blood–tissue barrier is compromised.
tumour vaccination studies suggest that epitope spread- Autoreactive TH2 cells have been shown to be capable of
ing may increase the efficacy of vaccination protocols. mediating diabetes74 and CNS demyelination75 when
The immune response therefore appears to be transferred to immunodeficient recipients.
extremely dynamic, with a constantly changing focus. As discussed above, our group and others have shown
Although the hierarchy and functional significance of that short-term blockade of either the CD28–CD80/86
responses to spread epitopes can be characterized in or CD40–CD154 co-stimulatory pathways in animals
defined animal models of immune-mediated disease, with established relapsing EAE specifically inhibits the
gathering this knowledge in human disease is difficult. initiation of responses to spread epitopes, resulting in
long-term protection from disease progression. Short-
Treatment of autoimmunity and transplant rejection. term co-stimulation blockade, therefore, has the net
What implications do the phenomena of epitope effect of inducing antigen-specific tolerance, in that
spreading have for the future of antigen-specific these reagents specifically target ongoing T-cell activa-
immunotherapies? Currently, it would be difficult to tion (epitope spreading), but only short-term treatment
define the pattern of epitope spreading in a chronic is required.
human autoimmune disease because the initiating epi- We have focused primarily on epitope spreading as a
tope specificity is not known and the epitope domi- mechanism involved in magnification of pathological
nance and hierarchy of immune responses to multiple immune responses. It is likely, however, that epitope
tissue antigens would vary depending on the individ- spreading is an intrinsic part of an immune reaction,
ual’s HLA genotype. Therefore, the use of peptide- both in magnifying the initial attack and in its subse-
specific tolerance to treat chronic autoimmune diseases quent downregulation. Therefore, initiation of TH2
in humans may not be practical at this time. However, as responses via epitope spreading may be an important
shown in the relapsing EAE models discussed previ- intrinsic immunoregulatory mechanism in TH1-
ously, it is theoretically possible to achieve long-term initiated diseases, geared to limit tissue destruction and
amelioration of autoimmune tissue destruction by promote re-establishment of tissue-specific immune
inducing peptide-specific tolerance to endogenous tis- tolerance. Interestingly, in the NOD model of type 1 dia-
sue epitopes in mice with ongoing disease10. In addition, betes, early induction of a TH2 (anti-inflammatory)
using an animal model of myasthenia gravis, Xu et al. response to one specific β-cell autoantigen (βCAA), by
developed a monoclonal antibody that recognizes the neonatal priming in incomplete freunds adjuvant, accel-
T-cell receptor (TCR) of α(100–116) AChR-specific erated epitope spreading of TH2 responses to other
T cells. Treatment with this mAb reversed ongoing βCAAs. This spreading of TH2 autoimmunity resulted
EAMG (when given early after disease onset) and in active tolerance and reduced disease incidence42,44.
reduced serum antibodies, including antibodies to the Early treatment with any one of several different
main immunogenic region, α(61–76), of the AChR in βCAAs prevents the development of diabetes in the
this rat model68. Although these examples show that, in NOD mice, presumably through TH2-mediated epitope
some disease models, peptide-specific treatments can spreading leading to the production of anti-inflamma-
ameliorate the ongoing disease, direct translation to tory cytokines. Although the efficiency of this treatment
treatment of human disease is currently difficult. in NOD mice declines when initiated at a later stage in
Another mechanism by which peptide-specific toler- the disease progression, the hope remains that early
ance may result in long-lasting disease inhibition, treatment of human diabetes might lead to therapeutic
despite epitope spreading, is via bystander suppression. epitope spreading and disease reduction.
Treatment of mice to induce TH2-directed immune Alloantigen-specific tolerance studies also provide
deviation before or shortly after priming for disease, evidence that epitope spreading may be important for
using therapies induced by self peptide administration immunoregulation. A study examining skin-graft toler-
by intravenous, oral or nasal routes69–71 or by altered ance after donor-specific transfusions found that toler-
peptide ligands (APLs)72, has been shown to inhibit ance induced by transfusions mismatched at one MHC
induction of EAE by other epitopes. However, therapy class-I locus actually spread to skin transplants that were
in all of these studies was started before the onset of mismatched at two MHC-class-I loci or at one MHC-
clinical disease and therefore induced a pre-established class-I locus plus multiple minor histocompatibility
anti-inflammatory TH2/regulatory environment. There antigens76. This data was interpreted to be the result of

92 | FEBRUARY 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd
REVIEWS

INFECTIOUS TOLERANCE epitope spreading of tolerance, but it might involve INFEC- Concluding remarks
+
Production of anti-inflammatory TIOUS TOLERANCE mediated by CD4 regulatory T cells77. The immune response is remarkably complex and
cytokines (e.g. IL-4, IL-10, strives to maintain a balance between pro- and anti-
TGF-β) by an antigen-specific
Implications for tumour and microbial vaccination. inflammatory responses. Understanding the tempo-
regulatory T cell, which suppress
immune responses to additional Epitope spreading has also been shown to be involved in ral dynamics of sequential immune responses to
epitopes in a non-specific the magnification of beneficial (infection and cancer) newly arising specificities and how these affect dis-
manner. immune responses. For example, El-Shami et al. have ease pathology and regulation remains an important
described epitope spreading after ovalbumin area of study in autoimmunity, transplantation and
(OVA)257–264 peptide immunization plus EG.70VA (thy- vaccination. The immune response consists of both
moma cell line of C57BL/6 transfected with the cDNA the initial magnification phase — which can be either
of chicken OVA) challenge in C57BL/6 mice78. Injection deleterious, as in autoimmune disease, or beneficial,
of EG.7OVA alone results in tumour growth and a CTL as in vaccinations or infectious disease — and a later
response restricted to the immunodominant OVA257–264 downregulatory phase to return the immune system
epitope. Mice immunized with peptide-pulsed irradi- to homeostasis. Epitope spreading may be an impor-
ated cells to induce an OVA257–264-specific CTL response tant component of both phases. Knowledge of the
and later challenged with a lethal dose of live EG.7OVA pattern of epitope spreading in human autoimmune
tumour cells were completely protected. In addition to disease or transplant rejection could be used to
OVA257–264, CTL responses in these mice targeted addi- design antigen-specific therapies that block ongoing
tional OVA peptides (intramolecular epitope spreading) tissue destruction or organ rejection, respectively.
and EL4 cells (intermolecular epitope spreading). Mice Alternatively, therapies could be designed to enhance
that underwent epitope spreading as a result of the natural downregulatory mechanisms. For exam-
OVA257–264 vaccination and EG.7OVA-tumour challenge ple, magnification of TH2 responses via epitope
were protected when challenged with live EL4 tumour spreading could be used to suppress pathogenic TH1
cells (no OVA antigen present), indicating that the inter- responses for protection against the development of
molecular epitope spreading induced during the vacci- diabetes, as discussed above.
nation process was protective. Similar results have been As discussed, identifying the hierarchy and func-
reported in immunity to P815 tumour cells79. tional significance of epitopes involved in human
Two recent studies in human cancer research vali- autoimmune diseases is difficult at present. Technical
date research into peptide-based vaccines and indicate limitations aside, short-term co-stimulatory blockade
that epitope spreading may play an important role in (CD28 or CD154) in animal models has been shown
their effectiveness. ERBB2 (HER-2/neu) is a self antigen to affect long-term antigen-specific regulation of
that is overexpressed in 15–30% of human adenocarci- responses to spread epitopes. It is also important to
nomas. Patients with ERBB2-overexpressing breast or note that epitope spreading during magnification of
ovarian tumours received ERBB2 peptides inoculated immune responses is not always pathological, but can
together with granulocyte–macrophage colony-stimu- be beneficial. For example, the use of peptide vaccines
lating factor (GM-CSF), which has been shown in rats for tumour or microbial vaccination may be more effi-
to elicit CD4+ T-cell responses to the intact protein. cient than once thought, given that responses to the ini-
Before vaccination, these patients had no ERBB2- tial peptide may be followed by responses to additional
specific responses80. After vaccination with specific pep- tumour or microbial peptides via epitope spreading,
tides, patients acquired immune responses not only to enhancing the efficacy of protection or therapy.
ERBB2 peptide but also to peptides not included in the Reagents like anti-CTLA-4, which lower the threshold
vaccine (epitope spreading). In another study of of T-cell activation and enhance the magnification of
breast/ovarian cancer, patients were injected with autol- T-cell responses, thereby accelerating epitope spread-
ogous dendritic cells pulsed with ERBB2- or mucin-1- ing, may improve the efficacy of tumour or microbial
derived peptides. Data from several patients in this peptide vaccines15,82,83.
study indicate that immunization with a single tumour As described in this review, attempts to manipulate
antigen may induce CTL reactivity towards several the pro-inflammatory/anti-inflammatory balance of
tumour antigens (that is, epitope spreading) in vivo81. the immune system can lead to unexpected conse-
These results indicate that epitope spreading may quences. Although the studies discussed here indicate
increase the efficiency of protein vaccination. Although that there are multiple possible therapeutic avenues to
the data described here were obtained from tumour explore, a much greater understanding of the regulation
immune responses, the concept may be applicable to of the immune system is required before effective and
microbial vaccination. safe therapies can be applied to human disease.

1. Lehmann, P. V., Forsthuber, T., Miller, A. & Sercarz, E. E. the autoimmune T-cell repertoire. Immunol. Today 14, immune therapy may still succeed [with a hedge on this bet].
Spreading of T-cell autoimmunity to cryptic determinants of 203–208 (1993). J. Autoimmun. 14, 278–282 (2000).
an autoantigen. Nature 358, 155–157 (1992). 3. Lehmann, P. V., Targoni, O. S. & Forsthuber, T. G. Shifting 5. Yu, M., Johnson, J. M. & Tuohy, V. K. A predictable sequential
The first description of epitope spreading in an T-cell activation thresholds in autoimmunity and determinant determinant spreading cascade invariably accompanies
autoimmune disease. spreading. Immunol. Rev. 164, 53–61 (1998). progression of experimental autoimmune encephalomyelitis:
2. Lehmann, P. V., Sercarz, E. E., Forsthuber, T., Dayan, C. M. 4. Steinman, L. Despite epitope spreading in the pathogenesis a basis for peptide-specific therapy after onset of clinical
& Gammon, G. Determinant spreading and the dynamics of of autoimmune disease, highly restricted approaches to disease. J. Exp. Med. 183, 1777–1788 (1996).

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | FEBRUARY 2002 | 9 3


© 2002 Macmillan Magazines Ltd
REVIEWS

6. Vanderlugt, C. L. et al. The functional significance of epitope 27. Tuohy, V. K., Yu, M., Yin, L., Kawczak, J. A. & Kinkel, R. P. 50. Vincent, A. et al. Determinant spreading and immune
spreading and its regulation by co-stimulatory molecules. Spontaneous regression of primary autoreactivity during responses to acetylcholine receptors in myasthenia gravis.
Immunol. Rev. 164, 63–72 (1998). chronic progression of experimental autoimmune Immunol. Rev. 164, 157–168 (1998).
7. Kumar, V. Determinant spreading during experimental encephalomyelitis and multiple sclerosis. J. Exp. Med. 189, 51. Vincent, A., Jacobson, L. & Shillito, P. Response to human
autoimmune encephalomyelitis: is it potentiating, protecting or 1033–1042 (1999). acetylcholine receptor α 138–199: determinant spreading
participating in the disease? Immunol. Rev. 164, 73–80 (1998). 28. Goebels, N. et al. Repertoire dynamics of autoreactive T initiates autoimmunity to self-antigen in rabbits. Immunol.
8. Tuohy, V. K. et al. The epitope spreading cascade during cells in multiple sclerosis patients and healthy subjects: Lett. 39, 269–275 (1994).
progression of experimental autoimmune encephalomyelitis epitope spreading versus clonal persistence. Brain 123, 52. Curnow, J., Corlett, L., Willcox, N. & Vincent, A.
and multiple sclerosis. Immunol. Rev. 164, 93–100 (1998). 508–518 (2000). Presentation by myoblasts of an epitope from endogenous
9. McRae, B. L., Vanderlugt, C. L., Dal Canto, M. C. & Miller, S. D. 29. Kurtzke, J. F. Epidemiologic evidence for multiple sclerosis acetylcholine receptor indicates a potential role in the
Functional evidence for epitope spreading in the relapsing as an infection. Clin. Microbiol. Rev. 6, 382–427 (1993). spreading of the immune response. J. Neuroimmunol. 115,
pathology of experimental autoimmune encephalomyelitis. 30. Olson, J. K., Croxford, J. L. & Miller, S. D. Virus-induced 127–134 (2001).
J. Exp. Med. 182, 75–85 (1995). autoimmunity: potential role of viruses in initiation, 53. Wang, H. B. et al. Anti-CTLA-4 antibody treatment triggers
The first demonstration that epitope spreading has perpetuation, and progression of T cell-mediated determinant spreading and enhances murine myasthenia
pathological significance in ongoing autoimmunity. autoimmune diseases. Viral Immunol. 14, 227–250 (2001). gravis. J. Immunol. 166, 6430–6436 (2001).
10. Vanderlugt, C. L. et al. Pathologic role and temporal 31. Karpus, W. J., Pope, J. G., Peterson, J. D., Dal Canto, M. C. 54. Yamamoto, A. M. et al. Anti-titin antibodies in myasthenia
appearance of newly emerging autoepitopes in relapsing & Miller, S. D. Inhibition of Theiler’s virus-mediated gravis: tight association with thymoma and heterogeneity of
experimental autoimmune encephalomyelitis. J. Immunol. demyelination by peripheral immune tolerance induction. nonthymoma patients. Arch. Neurol. 58, 885–890 (2001).
164, 670–678 (2000). J. Immunol. 155, 947–957 (1995). 55. Van Eden, W. et al. Heat-shock protein T-cell epitopes
A demonstration that ongoing autoimmunity and 32. Miller, S. D. et al. Persistent infection with Theiler’s virus trigger a spreading regulatory control in a diversified
epitope spreading can be specifically inhibited by leads to CNS autoimmunity via epitope spreading. Nature arthritogenic T-cell response. Immunol. Rev. 164, 169–174
peptide-specific tolerance or blockade of Med. 3, 1133–1136 (1997). (1998).
CD80/86–CD28 co-stimulation. The first description that a persistent virus infection 56. Sonderstrup, G. & McDevitt, H. Identification of autoantigen
11. Kennedy, M. K. et al. Inhibition of murine relapsing can lead to autoimmunity via epitope spreading. epitopes in MHC class II transgenic mice. Immunol. Rev.
experimental autoimmune encephalomyelitis by immune 33. Katz-Levy, Y. et al. Temporal development of autoreactive 164, 129–138 (1998).
tolerance to proteolipid protein and its encephalitogenic TH1 responses and endogenous antigen presentation of self 57. Moudgil, K. D. et al. Diversification of T cell responses to
peptides. J. Immunol. 144, 909–915 (1990). myelin epitopes by CNS-resident APCs in Theiler’s virus- carboxy-terminal determinants within the 65-kD heat-shock
12. Anderson, A. C. et al. High frequency of autoreactive myelin infected mice. J. Immunol. 165, 5304–5314 (2000). protein is involved in regulation of autoimmune arthritis.
proteolipid protein-specific T cells in the periphery of naive 34. Katz-Levy, Y. et al. Endogenous presentation of self myelin J. Exp. Med. 185, 1307–1316 (1997).
mice: mechanisms of selection of the self-reactive epitopes by CNS-resident APCs in Theiler’s virus-infected 58. Moudgil, K. D. Diversification of response to hsp65 during
repertoire. J. Exp. Med. 191, 761–770 (2000). mice. J. Clin. Invest. 104, 599–610 (1999). the course of autoimmune arthritis is regulatory rather than
13. Miller, S. D. et al. Blockade of CD28/B7-1 interaction Evidence of endogenous presentation of self epitopes pathogenic. Immunol. Rev. 164, 175–184 (1998).
prevents epitope spreading and clinical relapses of murine by resident APCs in the target tissue of the disease. 59. Prakken, A. B. et al. Autoreactivity to human heat-shock
EAE. Immunity 3, 739–745 (1995). 35. Borrow, P. et al. Investigation of the role of delayed-type- protein 60 predicts disease remission in oligoarticular
14. Howard, L. M. et al. Mechanisms of immunotherapeutic hypersensitivity responses to myelin in the pathogenesis of juvenile rheumatoid arthritis. Arthritis Rheum. 39,
intervention by anti-CD40L (CD154) antibody in an animal Theiler’s virus-induced demyelinating disease. Immunology 1826–1832 (1996).
model of multiple sclerosis. J. Clin. Invest. 103, 281–290 93, 478–484 (1998). 60. Prakken, A. B. et al. T-cell reactivity to human HSP60 in
(1999). 36. Neville, K. L., Padilla, J. & Miller, S. D. Myelin-specific oligo-articular juvenile chronic arthritis is associated with a
Evidence that blockade of CD40/CD154 co- tolerance attenuates the progression of a virus-induced favorable prognosis and the generation of regulatory
stimulation can ameliorate ongoing autoimmunity and demyelinating disease: implications for the treatment of MS. cytokines in the inflamed joint. Immunol. Lett. 57, 139–142
epitope spreading. J. Neuroimmunol. (In the press). (1997).
15. Karandikar, N. J., Eagar, T. A., Vanderlugt, C. L., Bluestone, 37. Eisenbarth, G. S. Type I diabetes mellitus. A chronic 61. deGraeff-Meeder, E. R. et al. Juvenile chronic arthritis: T cell
J. A. & Miller, S. D. CTLA-4 downregulates epitope autoimmune disease. N. Engl. J. Med. 314, 1360–1368 reactivity to human HSP60 in patients with a favorable
spreading and mediates remission in autoimmune disease. (1986). course of arthritis. J. Clin. Invest. 95, 934–940 (1995).
J. Neuroimmunol. 109, 173–180 (2000). 38. Delovitch, T. L. & Singh, B. The nonobese diabetic mouse as 62. Alam, A. et al. Persistence of dominant T cell clones in
16. Karandikar, N. J., Vanderlugt, C. L., Bluestone, J. A. & Miller, a model of autoimmune diabetes: immune dysregulation synovial tissues during rheumatoid arthritis. J. Immunol.
S. D. Targeting the B7/CD28:CTLA-4 costimulatory system gets the NOD. Immunity 7, 727–738 (1997). 156, 3480–3485 (1996).
in CNS autoimmune disease. J. Neuroimmunol. 89, 10–18 39. Kaufman, D. L. et al. Spontaneous loss of T cell tolerance to 63. Bradley, J. A. Indirect T cell recognition in allograft rejection.
(1998). glutamic acid decarboxylase in murine insulin-dependent Int. Rev. Immunol. 13, 245–255 (1996).
17. Tuohy, V. K., Yu, M., Yin, L., Kawczak, J. A. & Kinkel, P. R. diabetes. Nature 366, 69–72 (1993). 64. Suciu-Foca, N., Harris, P. E. & Cortesini, R. Intramolecular
Regression and spreading of self-recognition during the 40. Zechel, M. A., Elliott, J. F., Atkinson, M. A. & Singh, B. and intermolecular spreading during the course of organ
development of autoimmune demyelinating disease. Characterization of novel T-cell epitopes on 65 kDa and allograft rejection. Immunol. Rev. 164, 241–246 (1998).
J. Autoimmun. 13, 11–20 (1999). 67 kDa glutamic acid decarboxylase relevant in autoimmune 65. Ciubotariu, R. et al. Persistent allopeptide reactivity and
18. Rudick, R. A. Disease-modifying drugs for relapsing- responses in NOD mice. J. Autoimmun. 11, 83–95 (1998). epitope spreading in chronic rejection of organ allografts.
remitting multiple sclerosis and future directions for multiple 41. Zechel, M. A., Chaturvedi, P. & Singh, B. Characterization of J. Clin. Invest 101, 398–405 (1998).
sclerosis therapeutics. Arch. Neurol. 56, 1079–1084 (1999). immunodominant peptide determinants of IDDM-associated Evidence of epitope spreading in allograft rejection.
19. Arnason, B. G. Immunologic therapy of multiple sclerosis. autoantigens in the NOD mouse. Res. Immunol. 148, 66. Di Rosa, F. & Barnaba, V. Persisting viruses and chronic
Annu. Rev. Med. 50, 291–302 (1999). 338–348 (1997). inflammation: understanding their relation to autoimmunity.
20. Tuohy, V. K. et al. Modulation of the IL-10/IL-12 cytokine 42. Tian, J., Lehmann, P. V. & Kaufman, D. L. Determinant Immunol. Rev. 164, 17–27 (1998).
circuit by interferon-β inhibits the development of epitope spreading of T helper cell 2 (TH2) responses to pancreatic 67. Dorries, R. The role of T-cell-mediated mechanisms in virus
spreading and disease progression in murine autoimmune islet autoantigens. J. Exp. Med. 186, 2039–2043 (1997). infections of the nervous system. Curr. Top. Microbiol.
encephalomyelitis. J. Neuroimmunol. 111, 55–63 (2000). Evidence of protective epitope spreading. Immunol. 253, 219–245 (2001).
21. Fiorentino, D. F., Zlotnik, A., Mosman, T. R., Howard, M. H. 43. Zechel, M. A., Krawetz, M. D. & Singh, B. Epitope 68. Xu, L., Villain, M., Galin, F. S., Araga, S. & Blalock, J. E.
& O’Garra, A. IL-10 inhibits cytokine production by activated dominance: evidence for reciprocal determinant spreading Prevention and reversal of experimental autoimmune
macrophages. J. Immunol. 147, 3815–3822 (1991). to glutamic acid decarboxylase in non-obese diabetic mice. myasthenia gravis by a monoclonal antibody against
22. Fiorentino, D. F. et al. IL-10 acts on the antigen-presenting Immunol. Rev. 164, 111–118 (1998). acetylcholine receptor-specific T cells. Cell Immunol. 208,
cell to inhibit cytokine production by TH1 cells. J. Immunol. 44. Tian, J. et al. Infectious TH1 and TH2 autoimmunity in 107–114 (2001).
146, 3444–3451 (1991). diabetes-prone mice. Immunol. Rev. 164, 119–127 (1998). 69. Leadbetter, E. A. et al. Experimental autoimmune
23. De Waal Malefyt, R., Abrams, J., Bennett, B., Figdor, C. G. & 45. Durinovic-Bello, I. Autoimmune diabetes: the role of T cells, encephalomyelitis induced with a combination of myelin
De Vries, J. E. Interleukin 10 (IL-10) inhibits cytokine MHC molecules and autoantigens. Autoimmunity 27, basic protein and myelin oligodendrocyte glycoprotein is
synthesis by human monocytes: an autoregulatory role of 159–177 (1998). ameliorated by administration of a single myelin basic
IL-10 produced by monocytes. J. Exp. Med. 174, 46. Bonifacio, E., Scirpoli, M., Kredel, K., Fuchtenbusch, M. & protein peptide. J. Immunol. 161, 504–512 (1998).
1209–1220 (1991). Ziegler, A. G. Early autoantibody responses in prediabetes 70. Al-Sabbagh, A., Nelson, P. A., Akselband, Y., Sobel, R. A. &
24. Ding, L. & Shevach, E. M. IL-10 inhibits mitogen-induced are IgG1 dominated and suggest antigen-specific Weiner, H. L. Antigen-driven peripheral immune tolerance:
T cell proliferation by selectively inhibiting macrophage regulation. J. Immunol. 163, 525–532 (1999). suppression of experimental autoimmmune
costimulatory function. J. Immunol. 148, 3133–3139 (1992). 47. Bonifacio, E., Lampasona, V., Bernasconi, L. & Ziegler, A. G. encephalomyelitis and collagen-induced arthritis by aerosol
25. McFarland, H. I. et al. Determinant spreading associated Maturation of the humoral autoimmune response to administration of myelin basic protein or type II collagen.
with demyelination in a nonhuman primate model of multiple epitopes of GAD in preclinical childhood type 1 diabetes. Cell. Immunol. 171, 111–119 (1996).
sclerosis. J. Immunol. 162, 2384–2390 (1999). Diabetes 49, 202–208 (2000). 71. Anderton, S. M. & Wraith, D. C. Hierarchy in the ability of
26. Tuohy, V. K., Yu, M., Weinstock-Guttman, B. & Kinkel, R. P. 48. Sohnlein, P. et al. Epitope spreading and a varying but not T cell epitopes to induce peripheral tolerance to antigens
Diversity and plasticity of self recognition during the disease-specific GAD65 antibody response in type I from myelin. Eur. J. Immunol. 28, 1251–1261 (1998).
development of multiple sclerosis. J. Clin. Invest. 99, diabetes. The Childhood Diabetes in Finland Study Group. 72. Nicholson, L. B., Murtaza, A., Hafler, B. P., Sette, A. &
1682–1690 (1997). Diabetologia 43, 210–217 (2000). Kuchroo, V. K. A T cell receptor antagonist peptide induces
An initial demonstration of spreading and focusing of 49. Braghi, S. et al. Modulation of humoral islet autoimmunity by T cells that mediate bystander suppression and prevent
responses to PLP epitopes during the progression of pancreas allotransplantation influences allograft outcome in autoimmune encephalomyelitis induced with multiple myelin
multiple sclerosis. patients with type 1 diabetes. Diabetes 49, 218–224 (2000). antigens. Proc. Natl Acad. Sci. USA 94, 9279–9284 (1997).

94 | FEBRUARY 2002 | VOLUME 2 www.nature.com/reviews/immunol


© 2002 Macmillan Magazines Ltd
REVIEWS

73. Bielekova, B. et al. Encephalitogenic potential of the myelin 80. Disis, M. L., Grabstein, K. H., Sleath, P. R. & Cheever, M. A. 87. Lawson, C. M. Evidence for mimicry by viral antigens in
basic protein peptide (amino acids 83–99) in multiple Generation of immunity to the HER-2/neu oncogenic animal models of autoimmune disease including
sclerosis: results of a phase II clinical trial with an altered protein in patients with breast and ovarian cancer using a myocarditis. Cell Mol. Life Sci. 57, 552–560 (2000).
peptide ligand. Nature Med. 6, 1167–1175 (2000). peptide-based vaccine. Clin. Cancer Res. 5, 1289–1297 88. Fairweather, D., Kaya, Z., Shellam, G. R., Lawson, C. M. &
74. Pakala, S. V., Kurrer, M. O. & Katz, J. D. T helper 2 (TH2) (1999). Rose, N. R. From infection to autoimmunity. J. Autoimmun.
T cells induce acute pancreatitis and diabetes in immune- 81. Brossart, P. et al. Induction of cytotoxic T-lymphocyte 16, 175–186 (2001).
compromised nonobese diabetic (NOD) mice. J. Exp. Med. responses in vivo after vaccinations with peptide-pulsed 89. Horwitz, M. S. et al. Diabetes induced by Coxsackie virus:
186, 299–306 (1997). dendritic cells. Blood 96, 3102–3108 (2000). initiation by bystander damage and not molecular mimicry.
75. Lafaille, J. J. et al. Myelin basic protein-specific T helper 2 82. Leach, D. R., Krummel, M. F. & Allison, J. P. Enhancement of Nature Med. 4, 781–786 (1998).
(TH2) cells cause experimental autoimmune antitumor immunity by CTLA-4 blockade. Science 271, 90. Zhao, Z.-S., Granucci, F., Yeh, L., Schaffer, P. A. & Cantor, H.
encephalomyelitis in immunodeficient hosts rather than 1734–1736 (1996). Molecular mimicry by herpes simplex virus-type 1:
protect them from the disease. J. Exp. Med. 186, 307–312 83. Hurwitz, A. A., Yu, T. F., Leach, D. R. & Allison, J. P. CTLA-4 Autoimmune disease after viral infection. Science 279,
(1997). blockade synergizes with tumor-derived 1344–1347 (1998).
76. Yang, L., DuTemple, B., Gorczynski, R. M., Levy, G. & granulocyte–macrophage colony-stimulating factor for 91. Deshpande, S. P. et al. Herpes simplex virus-induced
Zhang, L. Evidence for epitope spreading and active treatment of an experimental mammary carcinoma. Proc. keratitis: evaluation of the role of molecular mimicry in lesion
suppression in skin graft tolerance after donor-specific Natl Acad. Sci. USA 95, 10067–10071 (1998). pathogenesis. J. Virol. 75, 3077–3088 (2001).
transfusion. Transplantation 67, 1404–1410 (1999). 84. Olson, J. K., Croxford, J. L., Calenoff, M., Dal Canto, M. C. &
77. Waldmann, H. & Cobbold, S. Regulating the immune Miller, S. D. A virus-induced molecular mimicry model of
response to transplants: a role for CD4+ regulatory cells? multiple sclerosis. J. Clin. Invest. 108, 311–318 (2001). Online links
Immunity 14, 399–406 (2001). Evidence that epitope spreading can be initiated after
78. el Shami, K. et al. MHC class I-restricted epitope spreading induction of autoimmunity via molecular mimicry. DATABASES
in the context of tumor rejection following vaccination with a 85. Mokhtarian, F., Shi, Y., Zhu, P. F. & Grob, D. Immune The following terms in this article are linked online to:
single immunodominant CTL epitope. Eur. J. Immunol. 29, responses, and autoimmune outcome, during virus infection LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink
3295–3301 (1999). of the central nervous system. Cell. Immunol. 157, 195–210 CD28 | CD40 | CD154 | CD80 | CD86 | CTLA-4 | GAD | GM-CSF |
Evidence of MHC-class-I–restricted epitope (1994). ERBB2 | HLA-DR | HLA-DR2 | IA-2 | IgG1 | IL-10 | IL-12 | IFN-β |
spreading in tumour immunity. 86. Mokhtarian, F., Zhang, Z., Shi, Y., Gonzales, E. & Sobel, R. A. IFN-γ | MBP | MOG | PLP | TNF-α
79. Markiewicz, M. A., Fallarino, F., Ashikari, A. & Gajewski, T. F. Molecular mimicry between a viral peptide and a myelin OMIM: http://www.ncbi.nlm.nih.gov/Omim
Epitope spreading upon P815 tumor rejection triggered by oligodendrocyte glycoprotein peptide induces autoimmune type-1 diabetes | multiple sclerosis | myasthenia gravis |
vaccination with the single class I MHC-restricted peptide demyelinating disease in mice. J. Neuroimmunol. 95, 43–54 rheumatoid arthritis
P1A. Int. Immunol. 13, 625–632 (2001). (1999). Access to this interactive links box is free online.

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | FEBRUARY 2002 | 9 5


© 2002 Macmillan Magazines Ltd

You might also like