Download Current Opinion in Immunology

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Phagocyte wikipedia , lookup

Duffy antigen system wikipedia , lookup

Monoclonal antibody wikipedia , lookup

Lymphopoiesis wikipedia , lookup

Immune system wikipedia , lookup

T cell wikipedia , lookup

Allergy wikipedia , lookup

DNA vaccination wikipedia , lookup

Innate immune system wikipedia , lookup

Adaptive immune system wikipedia , lookup

Multiple sclerosis research wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Hygiene hypothesis wikipedia , lookup

Autoimmunity wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Sjögren syndrome wikipedia , lookup

Immunomics wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Molecular mimicry wikipedia , lookup

Transcript
Sponsored document from
Current Opinion in Immunology
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sponsored Document
Antigen-specific immunotherapy of autoimmune and allergic
diseases
Catherine A Sabatos-Peyton*, Johan Verhagen*, and David C Wraith
David C Wraith: [email protected]
School of Cellular and Molecular Medicine, University of Bristol, Medical Sciences Building,
University Walk, Bristol, BS8 1TD, UK
Abstract
Sponsored Document
Nearly a century has passed since the first report describing antigen-specific immunotherapy
(antigen-SIT) was published. Research into the use of antigen-SIT in the treatment of both allergic
and autoimmune disease has increased dramatically since, although its mechanism of action is only
slowly being unravelled. It is clear though, from recent studies, that success of antigen-SIT depends
on the induction of regulatory T (T reg) cell subsets that recognise potentially disease-inducing
epitopes. The major challenge remaining for the widespread use of antigen-SIT is to safely administer
high doses of immunodominant and potentially pathogenic epitopes in a manner that induces T cell
tolerance rather than activation. This review illustrates that intelligent design of treatment agents and
strategies can lead to the development of safe and effective antigen-SIT.
Introduction
Sponsored Document
Current treatments for allergic and autoimmune disease treat disease symptoms or depend on
non-specific immune suppression. Treatment would be improved greatly by targeting the
fundamental cause of the disease, that is the loss of tolerance to an otherwise innocuous antigen
in allergy or self-antigen in autoimmune disease (AID). Much has been learned about the
mechanisms of peripheral tolerance in recent years. We now appreciate that antigen presenting
cells (APC) may be either immunogenic or tolerogenic, depending on their location,
environmental cues and activation state [1]. Furthermore, it is clear that both FoxP3+ and
FoxP3− cells with regulatory properties can be induced with specific antigen. This, therefore,
provides two guiding principles for the design of antigen-specific immunotherapeutic
strategies. First, antigen should be directed to tolerogenic rather than immunogenic APC;
secondly, administration of antigen should lead to the induction of regulatory T cells.
Allergen-specific immunotherapy
The application of allergen-SIT has become increasingly popular since first reported by
Leonard Noon in 1911. Subcutaneous (s.c.) or oral/sublingual administration of allergens has
been used for the successful treatment of a wide range of allergies including those to bee venom
[2], cow's milk [3], peanut [4] or birch pollen [5]. Typically, this starts with a build-up day
where the maximum tolerated dose is determined. This dose is then gradually escalated over
© 2010 Elsevier Ltd.
*These authors contributed equally to the writing of this review.
This document was posted here by permission of the publisher. At the time of deposit, it included all changes made during peer review,
copyediting, and publishing. The U.S. National Library of Medicine is responsible for all links within the document and for incorporating
any publisher-supplied amendments or retractions issued subsequently. The published journal article, guaranteed to be such by Elsevier,
is available for free, on ScienceDirect.
Sabatos-Peyton et al.
Page 2
Sponsored Document
a period of approximately two months to a high maintenance dose, which is administered
regularly for months to years. By escalating the treatment dose, a maintenance dose can be
reached that is far higher than the maximum tolerated dose at onset, with limited adverse effects.
Studies have shown that optimal results are achieved using the highest tolerable maintenance
dose [6] or a high cumulative dose, that is long-term treatment [7]. To further prevent adverse
effects and increase efficacy, a wide range of novel therapeutic strategies have been employed,
including the use of non-IgE binding allergen derivatives, adjuvants, alternative routes of
administration, fusion proteins, allergen-encoding cDNA and peptides that represent T cell
epitopes (reviewed in [8]). Hypoallergenic peptides, in particular, are an increasingly popular
alternative to whole proteins and have proven successful in animal models and human trials
[9,10,11••].
Sponsored Document
Progress in the development of allergen-SIT has been hindered by a lack of understanding of
the underlying immunological mechanisms. In recent years, it has become clear that the ratio
of allergen-specific T cells secreting distinct cytokines plays a crucial role in the onset and
cessation of allergic diseases. First, it is important to realise that allergic and non-allergic
individuals recognise the same T cell epitopes of common allergens [12] and that only the
frequency of different subsets of CD4+ T cells specific for these epitopes differs. It is now clear
that the balance between effector T cell populations on the one hand and IL-10-secreting,
suppressive T cells on the other makes the difference between an atopic or healthy immune
response. In atopic individuals, the highest proportion of T cells recognising common
environmental allergens are IL-4-secreting T helper (Th) 2 cells, whereas IL-10-secreting T
cells prevail in healthy individuals [13]. The importance of the dominance of the IL-10secreting T cell population for a healthy immune response to allergens is elegantly
demonstrated in the case of beekeepers [14••]. During the season, beekeepers are stung
frequently, thus receiving repetitive high doses of allergen. Remarkably, their immune
response to the venom skews rapidly, with a dramatic shift in the dominant T cell subtype
towards IL-10-secreting cells. This induction of IL-10-secreting cells has now also been shown
to be a dominant feature of successful allergen-SIT in a range of allergies (Table 1) [4,5,11••,
15,16]. Allergen-SIT is successful at any age but early treatment of a single allergy may prevent
epitope spreading and hence limit the atopic march in later life [17].
Autoantigen-specific immunotherapy
Sponsored Document
The use of SIT for AID has lagged behind SIT for allergy. This may be because AIDs are more
heterogeneous than allergic diseases; the disease-initiating or target antigen may not be known;
and/or the immune pathogenesis of AID is associated with epitope spreading [18] and
substantial tissue damage may have occurred before an effective diagnosis has been made.
Effective SIT for AID will, therefore, require the induction of cells capable of ‘bystander’
regulation or suppression at the earliest stage of disease [19,20].
First attempts at SIT, in diseases such as multiple sclerosis (MS), were not successful [21–
23]. Weiner and colleagues extended these studies by testing the phenomenon of mucosal
tolerisation in various experimental models [24]. This was universally effective and revealed
that a relatively low dose of antigen, delivered by the oral route, would induce ‘bystander
suppression’ whereby the administration of antigen A would induce cells capable of
suppressing responses to antigens B and C. Clinical trials proved that oral tolerance induction
is safe but not as effective as expected from studies in animal models.
The administration of self-antigen via plasmid DNA is an attractive approach since coexpression of cytokines and immune modulators can be used to enhance mechanisms of
tolerance induction. Early examples of DNA vaccination in AID models produced conflicting
results; disease could be either suppressed [25] or enhanced [26,27], depending on the disease
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 3
Sponsored Document
model or antigen expressed. Various approaches have been taken to enhance tolerance
induction. CpG motifs in plasmid DNA contribute to the Th1 response; this is reduced by coadministration of an oligonucleotide expressing GpG in place of CpG [28]. A DNA vaccine
encoding myelin basic protein, with CpG motifs replaced by GpG, was recently tested in MS
patients [29•]. Treatment with a 0.5 mg dose of DNA resulted in the reduction of new lesions
in the CNS, coinciding with a decrease in the Th1 response to myelin antigens. Co-expression
of cytokines designed to reduce the Th1 response to antigen enhanced the efficacy of
tolerogenic DNA vaccination in both EAE [30] and type I diabetes (T1D) models [31]. An
intriguing, novel approach involves the introduction of the microRNA miR-142 into the
antigen-expressing vector [32••]. The microRNA suppressed antigen expression in professional
APC and led to the induction of antigen-specific FoxP3+ cells in the liver. Expression of antigen
in the liver generally enhances the generation of induced FoxP3+ Treg cells and has proven
effective in models of uveitis and MS [33•,34•].
Sponsored Document
Although antigenic protein therapy has been successful in pre-clinical models, the approach
has not translated well into the clinic. Nevertheless, there has been success in the use of an
alum-based islet-antigen vaccine. GAD-alum treatment led to the preservation of residual
insulin secretion in patients with recent-onset T1D [35•]. One approach, designed to improve
the safety of self-antigen delivery in AID, involves the coupling of intact antigen to APC using
chemical fixatives [36]. This is based on the concept that fixation promotes a tolerogenic
response to the APC. A systematic study of this approach, however, revealed that efficacy is
not dependent on the delivery of antigen on autologous cells; in fact, cells may be replaced
with antigen-coated beads [37]. This implies that the fixed cells themselves are not directly
involved in tolerance induction; rather, they carry intact antigen to tolerogenic APC for
processing and presentation. A phase I trial of fixed autologous peripheral blood leukocytes
coupled with a cocktail of seven encephalitogenic myelin peptides is underway in early
relapsing-remitting MS patients [38].
Sponsored Document
The alternative to coupling antigen to APC is simply to administer soluble peptide via a
tolerogenic route. Much has been learned about the nature of the peptide, dose, route of delivery
and timing. The peptide must mimic the naturally processed antigen when bound to major
histocompatibility complex (MHC) [39]; post-translational modification of the peptide may
be required [40], and treatment must be initiated as soon as possible following definite
diagnosis (e.g., a single peptide could suppress diabetes at an early stage of disease while
treatment of late stage disease required administration of a combination of peptides [41]). One
approach to improving the safety and efficacy of peptide therapy has included the development
of recombinant MHC–peptide complexes. Treatment with sIAg7–pGAD65 complexes
effectively blocked the development of diabetes in the NOD mouse; suppression was dependent
on induction of islet-cell-specific IL-10-secreting CD4+ T cells [42]. Similarly, treatment with
a single recombinant MHC–peptide complex could reverse EAE through induction of IL-10secreting regulatory cells [43]. Clinical trials of peptide therapy have shown promising results
in a range of AID (Table 2) with various routes of administration and dosing schedules under
investigation. Importantly, as in many pre-clinical models, IL-10 is frequently associated with
effective peptide therapy.
The importance of antigen dose and IL-10 production for the success of
antigen-SIT
IL-10 secretion is a common self-regulatory property for the major CD4+ T helper subsets,
with Th1, Th2, Th9 and Th17 cells all shown to secrete IL-10 in the face of chronic exposure
to antigen (reviewed in [44•,45]). Both allergen-SIT and autoantigen-SIT exploit this natural
IL-10-secreting phenotype of highly differentiated effector cells with repeated exposure to
high-dose antigen converting effector T cells to IL-10-secreting regulatory populations (Figure
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 4
Sponsored Document
1). The greatest hazard of high-dose peptide-specific therapy, however, is a harmful immune
response due to the initial burst of cell activation with subsequent proliferation and excessive
cytokine release. This became evident in trials of altered peptide ligand (APL) therapy in MS.
Treatment was terminated when it became evident that an allergic response to the peptide had
been induced at the highest dose [46,47]. Importantly, this problem was not observed at lower
doses of peptide. We have recently shown, using peptide analogues, that anergy, suppression
and IL-10 secretion are dose or affinity dependent, with lower signal strength leading to anergy
and higher signal strength driving IL-10 secretion and effective regulation of the inflammatory
immune response [48•]. Combined with the body of knowledge from allergen-SIT, these data
suggest that initiating treatment with lower doses and building to the highest maintenance dose
allows the full benefit of tolerance induction while also protecting the recipient from harmful
side effects. We propose, therefore, that for antigen-SIT in both allergy and AID, a stepwise
approach, with dose escalation activating T cells through increasing strength of signal to a
higher maintenance dose, will induce tolerogenic IL-10-secreting cells, capable of suppressing
the effector properties of their initiating population (Figure 2).
Sponsored Document
Some studies suggest that IL-10 secretion is a transitory property of highly stimulated cells,
dependent on tissue-specific environmental cues for its maintenance, while others suggest that
the IL-10 locus can be genetically modified in terminal differentiation. Recent evidence has
shown that sustained high-dose TCR signalling and high levels of IL-12 were required for the
induction and maintenance of IL-10 secretion in Th1 cells, through sustained ERK1 and ERK2
MAP kinase phosphorylation [49••]. However, for Th2 cells, epigenetic modification of
chromatin at the IL-10 locus has been demonstrated [50,51]. Thus, the mechanistic details of
IL-10 regulation in T cells and the crucial question of whether IL-10 secretion can be imprinted
remain open.
Conclusion
Sponsored Document
This review has highlighted recent advances in specific immunotherapy for allergic and
autoimmune disease. One overriding conclusion is that regulatory mechanisms involving IL-10
are important for effective therapy. Our belief is that basic research should focus on means to
target tolerogenic APC; to promote, for example through the use of appropriate adjuvants,
secretion of IL-10 from both APC and T cells; and to investigate the mechanisms of dose
escalation tolerance. Future clinical trials should focus on patient groups that are most likely
to benefit from the treatment, for example major changes should not be expected in advanced
stages of disease [38]; no SIT trial should be undertaken without detailed investigation of
immunological changes arising from the treatment; and intervention should be undertaken as
early after diagnosis as possible. Finally, the dose of antigen required for SIT is the most critical
consideration; dose escalation should allow for a safe increase in dose until an effective,
tolerogenic dose is achieved.
References and recommended reading
Papers of particular interest, published within the period of review, have been highlighted as:
• of special interest
•• of outstanding interest
References
1. Reis e SousaC.Dendritic cells in a mature ageNat Rev Immunol6200647648316691244
2. AkdisC.A.AkdisM.BleskenT.WymannD.AlkanS.S.MullerU.BlaserK.Epitope-specific T cell
tolerance to phospholipase A2 in bee venom immunotherapy and recovery by IL-2 and IL-15 in vitroJ
Clin Invest981996167616838833918
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 5
Sponsored Document
Sponsored Document
Sponsored Document
3. SkripakJ.M.NashS.D.RowleyH.BreretonN.H.OhS.HamiltonR.G.MatsuiE.C.BurksA.W.WoodR.A.A
randomized, double-blind, placebo-controlled study of milk oral immunotherapy for cow's milk
allergyJ Allergy Clin Immunol12220081154116018951617
4.
JonesS.M.PonsL.RobertsJ.L.ScurlockA.M.PerryT.T.KulisM.ShrefflerW.G.SteeleP.HenryK.A.Adair
M.Clinical efficacy and immune regulation with peanut oral immunotherapyJ Allergy Clin
Immunol124200929230019577283
5. MobsC.SlotoschC.LofflerH.JakobT.HertlM.PfutznerW.Birch pollen immunotherapy leads to
differential induction of regulatory T cells and delayed helper T cell immune deviationJ
Immunol18420102194220320048125
6. FrewA.J.PowellR.J.CorriganC.J.DurhamS.R.Efficacy and safety of specific immunotherapy with SQ
allergen extract in treatment-resistant seasonal allergic rhinoconjunctivitisJ Allergy Clin
Immunol117200631932516461133
7. GadermaierE.FlickerS.AbererW.EggerC.ReiderN.FockeM.VrtalaS.KundiM.ValentaR.Analysis of
the antibody responses induced by subcutaneous injection immunotherapy with birch and Fagales
pollen extracts adsorbed onto aluminum hydroxideInt Arch Allergy Immunol1512010172719672093
8. FockeM.SwobodaI.MarthK.ValentaR.Developments in allergen-specific immunotherapy: from
allergen extracts to allergy vaccines bypassing allergen-specific immunoglobulin E and T cell
reactivityClin Exp Allergy40201038539720210812
9. MullerU.AkdisC.A.FrickerM.AkdisM.BleskenT.BettensF.BlaserK.Successful immunotherapy with
T-cell epitope peptides of bee venom phospholipase A2 induces specific T-cell anergy in patients
allergic to bee venomJ Allergy Clin Immunol10119987477549648701
10.
FellrathJ.M.KettnerA.DufourN.FrigerioC.SchneebergerD.LeimgruberA.CorradinG.SpertiniF.Aller
gen-specific T-cell tolerance induction with allergen-derived long synthetic peptides: results of a
phase I trialJ Allergy Clin Immunol111200385486112704369
11. CampbellJ.D.BucklandK.F.McMillanS.J.KearleyJ.OldfieldW.L.SternL.J.GronlundH.van
HageM.ReynoldsC.J.BoytonR.J.Peptide immunotherapy in allergic asthma generates IL-10dependent immunological tolerance associated with linked epitope suppressionJ Exp
Med20620091535154719528258This paper demonstrates induction of IL-10-mediated bystander
suppression following the administration of a peptide cocktail in allergic asthma.
12.
EbnerC.SchenkS.NajafianN.SiemannU.SteinerR.FischerG.W.HoffmannK.SzepfalusiZ.ScheinerO.
KraftD.Nonallergic individuals recognize the same T cell epitopes of Bet v 1, the major birch pollen
allergen, as atopic patientsJ Immunol1541995193219407530747
13.
AkdisM.VerhagenJ.TaylorA.KaramlooF.KaragiannidisC.CrameriR.ThunbergS.DenizG.ValentaR.
FiebigH.Immune responses in healthy and allergic individuals are characterized by a fine balance
between allergen-specific T regulatory 1 and T helper 2 cellsJ Exp Med19920041567157515173208
14. MeilerF.ZumkehrJ.KlunkerS.RuckertB.AkdisC.A.AkdisM.In vivo switch to IL-10-secreting T
regulatory cells in high dose allergen exposureJ Exp Med20520082887289819001136This study
clearly demonstrates natural control of the allergic response to a seasonal allergen through
differentiation of effector cells to an IL-10-secreting regulatory population.
15. OldfieldW.L.LarcheM.KayA.B.Effect of T-cell peptides derived from Fel d 1 on allergic reactions
and cytokine production in patients sensitive to cats: a randomised controlled
trialLancet3602002475312114041
16.
YamanakaK.YutaA.KakedaM.SasakiR.KitagawaH.GabazzaE.C.OkuboK.KurokawaI.MizutaniH.I
nduction of IL-10-producing regulatory T cells with TCR diversity by epitope-specific
immunotherapy in pollinosisJ Allergy Clin Immunol1242009842845e84719703707
17.
HoriguchiS.OkamotoY.YonekuraS.OkawaT.YamamotoH.KuniiN.SakuraiD.FujimuraT.Nakazawa
K.YasuedaH.A randomized controlled trial of sublingual immunotherapy for Japanese cedar
pollinosisInt Arch Allergy Immunol1462008768418087165
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 6
Sponsored Document
Sponsored Document
Sponsored Document
18. VanderlugtC.L.MillerS.D.Epitope spreading in immune-mediated diseases: implications for
immunotherapyNat Rev Immunol22002859511910899
19. MillerA.LiderO.WeinerH.L.Antigen-driven bystander suppression after oral administration of
antigensJ Exp Med17419917917981717632
20. ChenY.KuchrooV.K.InobeJ.HaflerD.A.WeinerH.L.Regulatory T cell clones induced by oral
tolerance: suppression of autoimmune encephalomyelitisScience2651994123712407520605
21. CampbellB.VogelP.J.FisherE.LorenzR.Myelin basic protein administration in multiple sclerosisArch
Neurol29197310154123119
22. GonsetteR.E.DelmotteP.DemontyL.Failure of basic protein therapy for multiple sclerosisJ
Neurol2161977273172790
23. RomineJ.S.SalkJ.A study of myelin basic protein as therapeutic probe in patients with multiple
sclerosisHallpikeJ.AdamsC.W.M.TourtellotteW.W.Multiple Sclerosis1983Cambridge University
Press621630
24. FariaA.M.WeinerH.L.Oral tolerance: mechanisms and therapeutic applicationsAdv
Immunol73199915326410399007
25.
RuizP.J.GarrenH.RuizI.U.HirschbergD.L.NguyenL.V.KarpujM.V.CooperM.T.MitchellD.J.Fathma
nC.G.SteinmanL.Suppressive immunization with DNA encoding a self-peptide prevents
autoimmune disease: modulation of T cell costimulationJ Immunol16219993336334110092787
26. WeaverD.J.Jr.LiuB.TischR.Plasmid DNAs encoding insulin and glutamic acid decarboxylase 65 have
distinct effects on the progression of autoimmune diabetes in nonobese diabetic miceJ
Immunol167200158659211418698
27. KargesW.PechholdK.Al
DahoukS.RieggerI.RiefM.WissmannA.SchirmbeckR.BarthC.BoehmB.O.Induction of autoimmune
diabetes through insulin (but not GAD65) DNA vaccination in nonobese diabetic and in RIP-B7.1
miceDiabetes5120023237324412401715
28. HoP.P.FontouraP.RuizP.J.SteinmanL.GarrenH.An immunomodulatory GpG oligonucleotide for the
treatment of autoimmunity via the innate and adaptive immune systemsJ
Immunol17120034920492614568974
29. BarOrA.VollmerT.AntelJ.ArnoldD.L.BodnerC.A.CampagnoloD.GianettoniJ.JaliliF.KachuckN.Lapier
reY.Induction of antigen-specific tolerance in multiple sclerosis after immunization with DNA
encoding myelin basic protein in a randomized, placebo-controlled phase 1/2 trialArch
Neurol6420071407141517698695This report provides evidence that antigen-encoding DNA can
modulate MS.
30.
GarrenH.RuizP.J.WatkinsT.A.FontouraP.NguyenL.T.EstlineE.R.HirschbergD.L.SteinmanL.Comb
ination of gene delivery and DNA vaccination to protect from and reverse Th1 autoimmune disease
via deviation to the Th2 pathwayImmunity152001152211485734
31. TischR.WangB.WeaverD.J.LiuB.BuiT.ArthosJ.SerrezeD.V.Antigen-specific mediated suppression
of beta cell autoimmunity by plasmid DNA vaccinationJ Immunol16620012122213211160264
32. AnnoniA.BrownB.D.CantoreA.SergiL.S.NaldiniL.RoncaroloM.G.In vivo delivery of a microRNAregulated transgene induces antigen-specific regulatory T cells and promotes immunologic
toleranceBlood11420095152516119794140The first description of microRNA application designed
to prevent conventional antigen presentation.
33.
SilverP.B.AgarwalR.K.SuS.B.SuffiaI.GrajewskiR.S.LugerD.ChanC.C.MahdiR.M.NickersonJ.M.C
aspiR.R.Hydrodynamic vaccination with DNA encoding an immunologically privileged retinal
antigen protects from autoimmunity through induction of regulatory T cellsJ
Immunol17920075146515817911600See annotation to Ref. [34•].
34.
LuthS.HuberS.SchrammC.BuchT.ZanderS.StadelmannC.BruckW.WraithD.C.HerkelJ.LohseA.W.
Ectopic expression of neural autoantigen in mouse liver suppresses experimental autoimmune
neuroinflammation by inducing antigen-specific TregsJ Clin Invest11820083403341018802476This
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 7
paper along with Ref. [33•] demonstrates how expression of antigen in the liver can promote selection
of inducible FoxP3+ regulatory T cells.
35.
Sponsored Document
Sponsored Document
Sponsored Document
LudvigssonJ.FaresjoM.HjorthM.AxelssonS.CheramyM.PihlM.VaaralaO.ForsanderG.IvarssonS.Jo
hanssonC.GAD treatment and insulin secretion in recent-onset type 1 diabetesN Engl J
Med35920081909192018843118Evidence that a single antigen can be used to suppress autoimmune
diabetes in human.
36. MillerS.D.TurleyD.M.PodojilJ.R.Antigen-specific tolerance strategies for the prevention and
treatment of autoimmune diseaseNat Rev Immunol7200766567717690713
37. SmithP.A.Morris-DownesM.HeijmansN.PryceG.ArterE.O’NeillJ.K.‘t
HartB.BakerD.AmorS.Epitope spread is not critical for the relapse and progression of MOG 8-21
induced EAE in Biozzi ABH miceJ Neuroimmunol1642005768415927270
38. LutterottiA.SospedraM.MartinR.Antigen-specific therapies in MS — current concepts and novel
approachesJ Neurol Sci2742008182218599087
39. AndertonS.M.VinerN.J.MatharuP.LowreyP.A.WraithD.C.Influence of a dominant cryptic epitope on
autoimmune T cell toleranceNat Immunol3200217518111812995
40. MonneauxF.HoebekeJ.SordetC.NonnC.BriandJ.P.MaillereB.SibilliaJ.MullerS.Selective modulation
of CD4+ T cells from lupus patients by a promiscuous, protective peptide analogJ
Immunol17520055839584716237076
41. TischR.WangB.SerrezeD.V.Induction of glutamic acid decarboxylase 65-specific Th2 cells and
suppression of autoimmune diabetes at late stages of disease is epitope dependentJ
Immunol16319991178118710415012
42. LiL.YiZ.WangB.TischR.Suppression of ongoing T cell-mediated autoimmunity by peptide-MHC
class II dimer vaccinationJ Immunol18320094809481619752238
43.
SinhaS.SubramanianS.MillerL.ProctorT.M.RobertsC.BurrowsG.G.VandenbarkA.A.OffnerH.Cyto
kine switch and bystander suppression of autoimmune responses to multiple antigens in experimental
autoimmune encephalomyelitis by a single recombinant T-cell receptor ligandJ
Neurosci2920093816382319321778
44. JankovicD.KuglerD.G.SherA.IL-10 production by CD4+ effector T cells: a mechanism for selfregulationMucosal Immunol3201023924620200511This review describes the emergence of IL-10secreting regulatory populations from effector T cells in infectious diseases.
45. SaraivaM.O’GarraA.The regulation of IL-10 production by immune cellsNat Rev
Immunol10201017018120154735
46.
BielekovaB.GoodwinB.RichertN.CorteseI.KondoT.AfsharG.GranB.EatonJ.AntelJ.FrankJ.A.Encep
halitogenic potential of the myelin basic protein peptide (amino acids 83–99) in multiple sclerosis:
results of a phase II clinical trial with an altered peptide ligandNat Med620001167117511017150
47. KapposL.ComiG.PanitchH.OgerJ.AntelJ.ConlonP.SteinmanL.Induction of a non-encephalitogenic
type 2 T helper-cell autoimmune response in multiple sclerosis after administration of an altered
peptide ligand in a placebo-controlled, randomized phase II trial. The Altered Peptide Ligand in
Relapsing MS Study GroupNat Med620001176118211017151
48. GabrysovaL.WraithD.C.Antigenic strength controls the generation of antigen-specific IL-10secreting T regulatory cellsEur J Immunol4020091386139520162554See annotation to Ref. [49••].
49. SaraivaM.ChristensenJ.R.VeldhoenM.MurphyT.L.MurphyK.M.O’GarraA.Interleukin-10
production by Th1 cells requires interleukin-12-induced STAT4 transcription factor and ERK MAP
kinase activation by high antigen doseImmunity31200920921919646904This paper along with Ref.
[47] describes the role of antigenic strength in promoting IL-10 secretion by Th1 cells.
50. ChangH.D.HelbigC.TykocinskiL.KreherS.KoeckJ.NiesnerU.RadbruchA.Expression of IL-10 in Th
memory lymphocytes is conditional on IL-12 or IL-4, unless the IL-10 gene is imprinted by
GATA-3Eur J Immunol37200780781717304625
51. ImS.H.HueberA.MonticelliS.KangK.H.RaoA.Chromatin-level regulation of the IL10 gene in T cellsJ
Biol Chem2792004468184682515319439
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 8
52.
Sponsored Document
KoffemanE.C.GenoveseM.AmoxD.KeoghE.SantanaE.MattesonE.L.KavanaughA.MolitorJ.A.Schi
ffM.H.PoseverJ.O.Epitope-specific immunotherapy of rheumatoid arthritis: clinical responsiveness
occurs with immune deviation and relies on the expression of a cluster of molecules associated with
T cell tolerance in a double-blind, placebo-controlled, pilot phase II trialArthritis
Rheum6020093207321619877047
53.
Sponsored Document
MullerS.MonneauxF.SchallN.RashkovR.K.OparanovB.A.WieselP.GeigerJ.M.ZimmerR.Spliceoso
mal peptide P140 for immunotherapy of systemic lupus erythematosus: results of an early phase II
clinical trialArthritis Rheum5820083873388319035498
54. ThrowerS.L.JamesL.HallW.GreenK.M.ArifS.AllenJ.S.Van-KrinksC.LozanoskaOchserB.MarquesiniL.BrownS.Proinsulin peptide immunotherapy in type 1 diabetes: report of a
first-in-man Phase I safety studyClin Exp Immunol155200915616519040615
55. FischerB.EliasD.BretzelR.G.LinnT.Immunomodulation with heat shock protein DiaPep277 to
preserve beta cell function in type 1 diabetes — an updateExpert Opin Biol
Ther10201026527220034363
56. HuurmanV.A.van der
MeideP.E.DuinkerkenG.WillemenS.CohenI.R.EliasD.RoepB.O.Immunological efficacy of heat
shock protein 60 peptide DiaPep277 therapy in clinical type I diabetesClin Exp
Immunol152200848849718422727
57. WarrenK.G.CatzI.FerencziL.Z.KrantzM.J.Intravenous synthetic peptide MBP8298 delayed disease
progression in an HLA Class II-defined cohort of patients with progressive multiple sclerosis: results
of a 24-month double-blind placebo-controlled clinical trial and 5 years of follow-up treatmentEur
J Neurol13200688789516879301
Acknowledgments
The authors wish to thank Bronwen R. Burton for useful discussions during the writing of this review. CASP receives
a fellowship from the MS Society of GB & NI; JV and DCW are supported by a Wellcome Trust Programme Grant.
Sponsored Document
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 9
Sponsored Document
Figure 1.
Self-regulatory properties of effector Th populations through chronic/repetitive stimulation.
Differentiation of naïve CD4+ T cells into effector Th1, Th2 and Th17 populations is well
established. Recent evidence from viral-infection and helminth-infection models, as well as
numerous allergen-specific and autoantigen-specific peptide therapy trials, suggests that
upregulation of IL-10 occurs during chronic or repetitive stimulation and can serve as a selflimiting mechanism [43,44•]. Shown are the potential cytokines/transcription factors/signalling
molecules that mediate the switch from effector T (T eff) to IL-10-secreting regulatory
populations. Note that Th2 cells produce IL-10 upon initial differentiation, and while evidence
does suggest that IL-10 helps host survival during helminth infection [44•], whether this is a
general feature of well-differentiated Th2 cells remains unclear.
Sponsored Document
Sponsored Document
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 10
Sponsored Document
Figure 2.
Dosing strategy for antigen-SIT. Repeated administration of high-dose/affinity antigen has the
potential to induce the highest level of anergy and tolerance, but the first few treatments may
induce acute and sometimes severe side effects. Low-dose antigen does not carry a risk of side
effects, nor does it induce robust tolerance. The low dose does, however, allow for the gradual
increase of the dose without the adverse effects normally associated with the higher dose.
Sponsored Document
Sponsored Document
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 11
Table 1
Studies demonstrating the importance of IL-10 induction for successful allergen-SIT.
Sponsored Document
Allergen
Treatment
Patients
Outcome
Ref.
Peanut
Titrated oral administration of peanut protein
up to 1800 mg (total 36 months)
29
Reduction in IgE; increased IgG4; increase in
IL-10, IL-5, IFN-γ and TNF-α. Increase in
antigen-specific FoxP3+ cells until 12 months
[4]
Birch pollen
Incremental weekly doses of s.c. standard
quality birch pollen allergen up to 100,000
units, followed by monthly maintenance dose
13
Increase in antigen-specific IL-10-secreting
cells; increase in allergen-specific IgG
antibodies
[5]
Fel d 1 (cat)
Asthma patients received intradermal (i.d.)
fel d 1 peptides in increments up to a 90 μg
total over two weeks
16
Reduced late reaction to cat dander; increased
IL-10, with linked epitope suppression
[11••,15]
Japanese cedar pollen
Sublingual application of a pool of 7 Cry j 1
and 2 derived peptides
75
Increase in IL-10-secreting regulatory cells;
reduction in allergy symptoms to cedar pollen
and other allergens
[16,17]
Sponsored Document
Sponsored Document
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.
Sabatos-Peyton et al.
Page 12
Table 2
Studies demonstrating the efficacy of self-antigenic peptide SIT in autoimmune diseases.
Sponsored Document
AID
Treatment
Patients
Outcome
Ref.
Rheumatoid Arthritis
DNAJP1 peptide
peroral, 25 mg/day
over six months
160
Immune deviation from TNF-α to IL-10; combination of
peptide and hydroxychloroquine most effective
[52]
Systemic Lupus Erythematosus
Three s.c. doses of
spliceosomal
peptide P140 at twoweek intervals
20
Anti-dsDNA antibody levels reduced in 200 μg group
[53]
T1D
Three i.d. doses of
proinsulin peptide at
monthly intervals
48
Increase of IL-10-secreting T cells in patients receiving
10 μg dose
[54]
T1D
DiaPep 277, hsp60
peptide, range of
doses around 1 mg
>300
IL-10 production in response to therapy associated with
preservation of C-peptide
[55,56]
Primary Progressive MS/Secondary
Progressive MS
MBP8298, 500 mg
i.v. every six months
32
Reduction in CSF anti-MBP. Delay to progression in
DR2/DR4 subgroup
[57]
Sponsored Document
Sponsored Document
Published as: Curr Opin Immunol. 2010 October ; 22(5): 609–615.