improved personalized vaccine and adoptive cell transfer...

1
Improved Personalized Vaccine and Adoptive Cell Transfer Immunotherapy Design by Immunoinformatic Analysis of Cancer Neo-epitopes for Regulatory T Cell Activation Potential Lenny Moise 1,2 , Guilhem Richard 1 , Frances Terry 1 , Bill Martin 1 , Annie De Groot 1,2 1 EpiVax, Inc., Providence, RI, United States; 2 Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States Mutanome-Directed Cancer Immunotherapy For questions regarding in silico antigen screening and vaccine design, please contact: Steven Vessella at 401-272-2123, ext. 107; or at [email protected] www.epivax.com Abstract Conclusions § Sequence analysis of the two faces of T cell epitopes enables prediction of epitope phenotype. § Epitopes that share a TCR-face with numerous human sequences may activate Tregs. § Sharper definition of neo-antigens by immunoinformatic analysis may improve epitope selection for mutanome- directed cancer immunotherapy. Tumor-specific mutations discovered using whole-exomic sequencing of tumor-normal pairs can be harnessed to identify neo-epitopes capable of stimulating T cell-mediated processes leading to tumor regression. Neo- epitope prediction using computational tools rapidly identifies epitope candidates in the mutanome, but a large proportion of neo-epitopes is not immunogenic. One explanation is that some epitopes, particularly class II major histocompatibility complex (MHC) epitopes, activate regulatory T cells (Tregs) trained in the thymus on self-antigens, which reduces anti- tumor activity. To address this pitfall, we designed the JanusMatrix algorithm to identify tumor-specific sequences capable of Treg activation by parsing candidate sequences into MHC-facing and T cell receptor (TCR)-facing sequences. As epitopes with shared TCR-faces can cross- react with the same T cells, cancer epitopes that share TCR-faces with multiple human sequences may cross-react with thymic-derived Tregs and are thus counter-indicated for immunotherapy. Retrospective analysis of non-small lung cell cancer neo-epitopes showed up to ~30% of neo- epitopes in a single patient exceed the cutoff for significant TCR-face homology with human proteins. Based on prior validation of Treg activating epitope predictions in infectious disease studies, we expect application of JanusMatrix to the neo-epitope discovery pipeline will focus candidate selection on high-value sequences. Neo-epitopes with low Treg activation potential may then be used to support development of personalized therapies including vaccination and in vitro expansion of tumor infiltrating lymphocytes for adoptive cell transfer. References Identify potential neoantigens Identify potential neoantigens Create synthetic vaccine (RNA, DNA, peptide) Provide in combination with adjuvant and checkpoint blockade Induce or expand neoantigen specic T cells Provide in combination checkpoint blockade B C T cell receptor face MHC- binding face JanusMatrix separates the amino acid sequence of T cell epitopes into TCR-facing residues (epitope) and HLA binding cleft-facing residues (agretope), then compares the TCR face to other putative T cell epitopes. Cross-reactive peptides: Are predicted to bind the same MHC allele. Share same/similar T cell-facing residues. TCR cross-reactivity prediction: Given a protein or peptide, T cell epitopes are identified based on MHC contacts (P1, P4, P6, P9) using EpiMatrix. JanusMatrix searches for potentially cross-reactive TCR by screening TCR-facing residues (P2, P3, P5, P7, P8) against a preloaded, EpiMatrix- processed reference databases. JanusMatrix Discovers Potential Treg Epitopes 0 0.5 1 1.5 2 2.5 IL)10 IL)4 Average Janus Human Score A Cytokines B Avg Janus Human Score vs +/9 Cytokine Release IEDB Posi2ve IEDB Nega2ve p=0.000 p=0.008 0 5 10 15 20 25 30 IL)10 IL)4 Average Janus Microbiome Score A Cytokines B Avg Janus Microbiome Score vs +/: Cytokine Release IEDB Posi2ve IEDB Nega2ve p=0.000 p=0.004 A) Peptides documented as IL-10-positive in IEDB have significantly higher potential for Human Genome (HG, above) and Human Microbiome (HM, below) cross-reactivity as measured by Janus Matrix. B) Peptides documented as IL-4-positive in IEDB have significantly lower potential for HG and HM cross-reactivity as measured by Janus Matrix. Cancer Vaccine Epitope Selection CD4 FoxP3 9.22% 22.86% FoxP3 B CD304 Isotype controls 10 5 10 4 10 3 0 0 10 3 10 4 10 5 0 10 3 10 4 10 5 10 5 10 4 10 3 0 Predicted Treg-activating HCV sequence possesses TCR faces shared by numerous human proteins Epitope networks are shown, illustrating the abundance of TCR faces one HCV peptide shares with the human genome as determined by JanusMatrix analysis. The HCV source peptide is represented by a green diamond, its constituent 9-mer epitopes by gray squares, their cross-conserved partners in the human genome by blue triangles, and the source human proteins by light purple circles 2 0 4 6 8 10 * * ** %CD3 + CD4 + FoxP3 + cells Ab + VL + Ab - VL - Ab + VL - Medium alone HCV_G1_p7_794 HCV_G1_NS4b_1941 FoxP3 FoxP3 CD304 1.61% 69.20% CD4 B A 10 5 10 4 10 3 0 0 10 3 10 4 10 5 0 10 3 10 4 10 5 10 5 10 4 10 3 0 • HCV_G1_p7_794 expands Tregs in chronic HCV-infected individuals but not in those who clear infection or are not infected. • Its human analog expands Tregs in uninfected and chronic infected subjects, while a control peptide HCV_G1_NS4b_194, does not for any group. • CD3 + CD4 + FoxP3 + cells from chronic HCV-infected individuals, cultured in the absence of HCV_G1_p7_794 express CD304 (neuropilin), indicative of nTreg cells. • The vast majority of CD3 + CD4 + FoxP3 + cells, cultured in the presence of HCV_G1_p7_794, were CD304 - characteristic of iTreg cells. Source 9-mer epitope Source peptide Source human protein TCR-conserved human 9-mer epitope • Pentatopes were constructed from mutated sequences derived from colon carcinoma model CT26 (study performed by Kreiter et al .). • Pentatope2 showed greater antitumor activity compared to Pentatope1 in BALB/c mice inoculated with CT26 tumor cells. • EpiMatrix identified a higher number of neo-epitopes in Pentatope2 compared to Pentatope1. Sequences from Pentatope2 also have a decreased potential to cross- react with other murine proteins, as identified by JanusMatrix. Moise L, Gutierrez A, Kibria F, Martin R, Tassone R, Liu R, Terry F, Martin B, De Groot AS. iVAX: An integrated toolkit for the selection and optimization of antigens and the design of epitope-driven vaccines. Hum Vaccin Immunother. 2015;11(9):2312-21. Moise L, Gutierrez AH, Bailey-Kellogg C, Terry F, Leng Q, Abdel Hady KM, VerBerkmoes NC, Sztein MB, Losikoff PT, Martin WD, Rothman AL, De Groot AS. The two-faced T cell epitope: examining the host-microbe interface with JanusMatrix. Hum Vaccin Immunother. 2013 Jul;9(7):1577-86. Losikoff PT, Mishra S, Terry F, Gutierrez A, Ardito MT, Fast L, Nevola M, Martin WD, Bailey-Kellogg C, De Groot AS, Gregory SH. HCV epitope, homologous to multiple human protein sequences, induces a regulatory T cell response in infected patients. J Hepatol. 2015 Jan;62(1):48-55. Kreiter S, Vormehr M, van de Roemer N, Diken M, Löwer M, Diekmann J, Boegel S, Schrörs B, Vascotto F, Castle JC, Tadmor AD, Schoenberger SP, Huber C, Türeci Ö, Sahin U. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature. 2015 Apr 30;520(7549):692-6. Extended Data Figure 3b. Kreiter et al., Nature 520, 692–696. 2015 • EpiMatrix identifies neo-epitopes by analyzing the binding potential of normal and mutated peptides. • JanusMatrix further down selects candidate epitopes based on their potential to cross-react with self-proteins. Neo-epitope selection pipeline EpiMatrix analysis of cancer vaccine efficacy Tumor Biopsy Expressed Mutation Discovery Neo-Antigen Discovery I MMUNOINFORMATICS Mutanome eliminate MHC face TCR face TCR-face analysis non-self self mutation MHC binder MHC binder neo-epitope mutation non-binder neo-epitope eliminate vaccine candidate JanusMatrix analysis identical TCR-face different TCR-face Normal Tumor Tumor cell CD8 + T cell CD4 + T cell APC Immunotherapy Adapted from Schumacher and Schreiber Science 348, 69-74. 2015

Upload: lamdien

Post on 27-Jan-2019

220 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Improved Personalized Vaccine and Adoptive Cell Transfer ...epivax.com/wp-content/uploads/2017/07/Cancer_vaccine_Keystone_08... · pies that exploit either the antigen repertoire

Improved Personalized Vaccine and Adoptive Cell Transfer Immunotherapy Designby Immunoinformatic Analysis of Cancer Neo-epitopes

for Regulatory T Cell Activation Potential Lenny Moise1,2, Guilhem Richard1, Frances Terry1, Bill Martin1, Annie De Groot1,2

1 EpiVax, Inc., Providence, RI, United States; 2 Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States

Mutanome-Directed Cancer Immunotherapy

Forquestionsregardinginsilicoantigenscreeningandvaccinedesign,pleasecontact:StevenVessellaat401-272-2123,ext.107;[email protected]

Abstract

Conclusions§ Sequence analysis of the two faces of T cell epitopes enables prediction of epitope phenotype.§ Epitopes that share a TCR-face with numerous human sequences may activate Tregs.§ Sharper definition of neo-antigens by immunoinformatic analysis may improve epitope selection for mutanome-

directed cancer immunotherapy.

Tumor-specific mutations discovered using whole-exomic sequencing oftumor-normal pairs can be harnessed to identify neo-epitopes capable ofstimulating T cell-mediated processes leading to tumor regression. Neo-epitope prediction using computational tools rapidly identifies epitopecandidates in the mutanome, but a large proportion of neo-epitopes is notimmunogenic. One explanation is that some epitopes, particularly class IImajor histocompatibility complex (MHC) epitopes, activate regulatory Tcells (Tregs) trained in the thymus on self-antigens, which reduces anti-tumor activity. To address this pitfall, we designed the JanusMatrixalgorithm to identify tumor-specific sequences capable of Treg activationby parsing candidate sequences into MHC-facing and T cell receptor(TCR)-facing sequences. As epitopes with shared TCR-faces can cross-react with the same T cells, cancer epitopes that share TCR-faces withmultiple human sequences may cross-react with thymic-derived Tregs andare thus counter-indicated for immunotherapy. Retrospective analysis ofnon-small lung cell cancer neo-epitopes showed up to ~30% of neo-epitopes in a single patient exceed the cutoff for significant TCR-facehomology with human proteins. Based on prior validation of Treg activatingepitope predictions in infectious disease studies, we expect application ofJanusMatrix to the neo-epitope discovery pipeline will focus candidateselection on high-value sequences. Neo-epitopes with low Treg activationpotential may then be used to support development of personalizedtherapies including vaccination and in vitro expansion of tumor infiltratinglymphocytes for adoptive cell transfer.

References

melanocyte differentiation antigens, a prominentclass of self-antigens in melanoma, shows a rel-atively modest clinical effect that is accompa-nied by substantial on-target toxicity againsthealthy melanocytes (43). Because this toxicity isrelatively infrequent in TIL therapy, these datastrongly suggest that T cell reactivity against themelanocyte differentiation antigens is not amajor driver of the antitumor activity of thistherapy. At the same time, there is data showingthat T cell products directed against NY-eso-1,one of the nonmutant self-antigens from thefamily of cancer/germline antigens that show verylimited expression in healthy tissue, can displaysubstantial antitumor activity (44, 45). Thus, al-though the available data support the notion thatT cell recognition of neoantigens contributes sub-stantially to the effects of the currently usedimmunotherapies, it would not be justified todismiss a potential contribution of T cell re-sponses against a subset of nonmutant antigens.A direct comparison of the antitumor activity ofneoantigen-specific and self-antigen–specific Tcells obtained from individual patients would beuseful to further address this issue.

Therapeutic use of the patient-specificneoantigen repertoire

Based on the fact that, at least in tumors withhigh mutational loads, the amount of DNA dam-age is sufficient for the immune system to seeone ormultiple epitopes as foreign, it becomes ofinterest to stimulate neoantigen-specific T cellresponses in cancer patients. Such stimulationcan obviously only be of value if the strength ofthe neoantigen-specific T cell response is other-wise a limiting factor in tumor control. Humandata on this important issue are lacking. However,in mouse models, vaccination with defined neoan-tigens has been shown to result in increased tumorcontrol (10, 14, 15,20), providing sufficient rationalefor the clinical development of neoantigen-directedtherapeutics. Because the majority of possibleneoantigens are specific to the individual being

treated (Fig. 3), such therapeutic approaches willin most cases entail personalized immunothera-pies that exploit either the antigen repertoire inthe tumor cells themselves or information onthat repertoire, as obtained by tumor sequencing(Fig. 4). As a first approach, a combination ofcheckpoint-blocking antibodies with therapeuticinterventions—such as tumor radiotherapy, onco-lytic viruses, or autologous tumor cell vaccines—that can increase neoantigen exposure to the Tcell–based immune system may be synergistic(Fig. 4A). As a downside, as compared to molec-ularly defined vaccines, the neoantigens releasedby such strategies will be diluted by the largeamount of nonmutant peptides that are alsopresent. In addition, control over the maturationsignals received by antigen-presenting cells is rela-tively limited. Nevertheless, because of the relativeease of clinical development of some of these com-bination therapies, extensive testing of such ther-apies is warranted.To allow a more defined targeting of the neo-

antigen repertoire in human tumors, two alter-native approaches should be considered, in bothcases relying on sets of potential neoantigens asidentified by sequencing of tumor material (Fig. 4,B andC). First, synthetic vaccinesmay be producedthat contain or encode a set of predicted neoan-tigens. Although still a substantial departure fromthe classical pharmaceutical model, clinical devel-opment of such personalized vaccines is withinreach (46–48). Mouse model data support theclinical translation of this approach, and the twomost pressing questions appear to be (i) whetherour ability to predict the most relevant neoan-tigens is already sufficiently advanced and (ii)how such vaccines may best be administered.Second, the information obtained from tumorsequencing may be used to create neoantigen-specific T cell products in vitro. This may involveeither the expansion of neoantigen-specific T cellpopulations that can already be detected withintumor tissue or in blood or the de novo inductionof such cells.

Regardless of the strategy used to enhanceneoantigen-specific T cell reactivity, it will likelyprove important to target multiple neoantigenssimultaneously in order to prevent tumor escapeby editing of the mutated epitope concerned (1).In addition, it may be prudent to avoid thetargeting of mutations in gene products that areseen by the immune system in autoimmunedisease to avoid induction of or exacerbation ofcancer-associated autoimmune disease (49).

Concluding remarks

Based on data obtained over the past few years,it is plausible that neoantigen-specific T cellreactivity forms a major “active ingredient” ofsuccessful cancer immunotherapies. In otherwords, the genetic damage that on the one handleads to oncogenic outgrowth can also be targetedby the immune system to control malignancies.Based on this finding, it will be important toengineer therapeutic interventions by whichneoantigen-specific T cell reactivity is selectivelyenhanced. Because of the tumor-restricted expres-sion of the antigens that are being targeted, thesepersonalized cancer immunotherapies offer thepromise of high specificity and safety. Conceiv-ably, the boosting of neoantigen-specific T cellreactivity that can be achieved with such person-alized immunotherapies will further increase thespectrum of human malignancies that respond tocancer immunotherapy.

REFERENCES AND NOTES

1. R. D. Schreiber, L. J. Old, M. J. Smyth, Science 331,1565–1570 (2011).

2. F. S. Hodi et al., N. Engl. J. Med. 363, 711–723(2010).

3. C. S. Hinrichs, S. A. Rosenberg, Immunol. Rev. 257, 56–71(2014).

4. M. E. Dudley et al., J. Clin. Oncol. 31, 2152–2159(2013).

5. P. Sharma, J. P. Allison, Science 348, 56–61 (2015).6. P. C. Tumeh et al., Nature 515, 568–571 (2014).7. E. Gilboa, Immunity 11, 263–270 (1999).8. B. Heemskerk, P. Kvistborg, T. N. Schumacher, EMBO J. 32,

194–203 (2013).

SCIENCE sciencemag.org 3 APRIL 2015 • VOL 348 ISSUE 6230 73

Identify potentialneoantigens

Identify potentialneoantigens

Createsynthetic vaccine

(RNA, DNA, peptide)

Provide in combinationwith adjuvant and

checkpoint blockade

Induce or expandneoantigen

specific T cells

Provide in combinationcheckpoint blockade

Inducetumor celldestruction

Provide checkpoint blockade

IIttttudddde

A B C

Fig. 4. Strategies to target the patient-specific neoantigen repertoire. (A) Immunotherapy is given in combination with interventions such as radiotherapythat enhance exposure to autologous neoantigens. (B) Potential neoantigens are identified as in Fig. 1 steps 1 to 3, a patient-specific vaccine is produced, andthis vaccine is given together with adjuvant and Tcell checkpoint-blocking antibodies. (C) Potential neoantigens are identified as in Fig. 1 steps 1 to 3,Tcells thatare specific for these neoantigens are induced or expanded in vitro, and the resulting Tcell product is given together with Tcell checkpoint-blocking antibodies.

T cell receptor

face

MHC-binding

face

JanusMatrix separates the amino acid sequence of T cellepitopes into TCR-facing residues (epitope) and HLA bindingcleft-facing residues (agretope), then compares the TCR faceto other putative T cell epitopes.

Cross-reactive peptides:• Are predicted to bind the same MHC allele.• Share same/similar T cell-facing residues.

TCR cross-reactivity prediction:• Given a protein or peptide, T cell epitopes are

identified based on MHC contacts (P1, P4, P6, P9)using EpiMatrix.

• JanusMatrix searches for potentially cross-reactiveTCR by screening TCR-facing residues (P2, P3,P5, P7, P8) against a preloaded, EpiMatrix-processed reference databases.

JanusMatrix Discovers Potential Treg Epitopes

0"

0.5"

1"

1.5"

2"

2.5"

IL)10" IL)4"

Average'Jan

us'Human

'Score'

A'''''''''''''''''''''''''''Cytokines''''''''''''''''''''''''''''B'

Avg'Janus'Human'Score'vs'+/9'Cytokine'Release'IEDB"Posi2ve" IEDB"Nega2ve"

p=0.000 p=0.008

0"

5"

10"

15"

20"

25"

30"

IL)10" IL)4"

Average'Janus'Microbiome'Score'

A''''''''''''''''''''''''''''Cytokines'''''''''''''''''''''''''''B'

Avg'Janus'Microbiome'Score'vs'+/:'Cytokine'Release'IEDB"Posi2ve" IEDB"Nega2ve"p=0.000 p=0.004

A) Peptides documented as IL-10-positive in IEDB have significantly higher potential for Human Genome (HG, above) and Human Microbiome (HM, below) cross-reactivity as measured by Janus Matrix.

B) Peptides documented as IL-4-positive in IEDB have significantly lower potential for HG and HM cross-reactivity as measured by Janus Matrix.

Cancer Vaccine Epitope Selection 309 that elicit the activity of nTreg cells, which normally function to310 suppress autoimmune reactivity to self-antigens (proteins). In311 this regard, it is pertinent to remark that HCV_G1_p7_794 is com-312 prised of epitopes that are homologous to those found in hun-313 dreds of human proteins. This suggests that the autoimmune314 response to a large number of proteins is inhibited by a single315 or limited number of nTreg cell clones, responsive to a common316 peptide sequence, rather than a large number of clones each317 responsive to a unique sequence in a single protein only. The oli-318 goclonality of the Treg cell response to HCV_G1_p7_794 and its319 human analogue is a matter of ongoing investigation.

320The majority of individuals in our patient population were321infected with HCV genotype 1, the derivation of HCV_G1_p7_794.322The addition of HCV_G1_p7_794 to PBMC cultures derived from a323single HCV genotype 3-infected patient, however, also resulted in324a marked increase in CD3+CD4+FoxP3+CD25+ cells (Supplemen-325tary Fig. 2). It is pertinent to note, in this regard, that HCV geno-326type 3 encodes a peptide sequence: LALLVLLLPQRAYAW, which327exhibits HCV_G1_p7_794 homology.328Conceivably, other viral pathogens that cause chronic disease,329e.g., herpes simplex, Epstein-Barr, human immunodeficiency and330cytomegalovirus, also avoid or reduce Teff cell responses by331exploiting similarity to self and activating nTreg cells [29,30].332Indeed, recent analyses indicated that Epstein-Barr virus and333cytomegalovirus contained fewer T cell epitopes and exhibited334higher cross-reactivity with the human genome than did either335Ebola or Marburg virus [14]. Ebola and Marburg viruses, on the336other hand, were composed of significantly fewer peptide337sequences that were cross-reactive with human and expressed338a larger number of predicted T cell epitopes. Thus, viruses that339cause acute disease and viruses such as HCV, which adapt to340humans and cause chronic infection, may differ substantially in341terms of their Treg cell epitope content.342Although immunosuppression by Treg cells is readily343demonstrated in mice, demonstrating the suppressor activity of344human CD3+CD4+CD25+FoxP3+ Treg cells has proven problematic345[31]. Recent studies indicate that the nature of the responder T346cells (CD4+CD25! vs. CD4+CD25low) and the ratio of Treg cells to347responder cells exerts significant effects on the outcome of the348suppression assays [32,33]. Nonetheless, the HCV_G1_p7_794349responsive Treg cells suppressed the mitogenic response of cells350derived from HCV-infected patients in the experiments reported351here despite the fact that the CD3+CD4+FoxP3! responder cells far352outnumbered CD3+CD4+CD25+FoxP3+ suppressor cells among353total PBMCs by a greater than 10:1 ratio.354Notably, the addition of HCV_G1_p7_794 alone to PBMC355cultures derived from HCV-infected (Ab+VL+) patients failed to356induce cell proliferation despite a marked (3- to 4-fold) increase357in CD3+CD4+CD25+FoxP3+ cell number. This finding indicates358that HCV_G1_p7_794 induces the conversion of conventional359CD4+FoxP3! T cells to Treg cells, i.e., infectious tolerance, a sugges-360tion supported by studies demonstrating the inability of nTreg cells361to proliferate in response to their cognate antigen in vitro [34]. Fur-362thermore, in contrast to Ab+VL+ PBMCs cultured in medium alone,363only a minority of CD3+CD4+FoxP3+ cells cultured in the presence364of HCV_G1_p7_794 expressed CD304 (neuropilin), which is365expressed by a subset of FoxP3+ Treg cells in humans and associated366specifically with nTreg cells in mice [21,22]. While it has been sug-367gested alternatively that the expanded Treg cell population in368chronic, HCV-infected patients is composed of cells phenotypically

FoxP3

FoxP

3

CD

304

1.61% 69.20%

CD4

CD4 FoxP3

9.22% 22.86%

FoxP

3

B

CD

304

FoxP3 CD4 CD304

Isotype controls

A

Count

105

104

103

0

105

104

103

0

800

600

400

200

0

1000

800

600

400

200

0

1000

800

600

400

200

0

0 103 104 105 0 103 104 105

0 103 104 105 0 103 104 105

101 102 103 104 105101 102 103 104 105101 102 103 104 105

105

104

103

0

105

104

103

0

Fig. 4. Fewer HCV-G1_p7_794 responsive Treg cells express CD304 (neuropilin).PBMCs obtained from an HCV-infected patient (representative of 4 patients) wereincubated in medium alone (A) or medium that contained HCV_G1_p7_794 (B).The cells were collected on day 5, stained and analysed by flow cytometry. Panelson the right indicate the percentage of CD4+FoxP3+ cells in each population thatexpress CD304.

Table 2. JanusMatrix analysis.

Description ICS # Cross-reactive epitopes per ICS

# Human proteins containing cross-reactive epitopes

HCV_G1_p7_794a WPLLLLLLALPQRAYAQ 5.0 264Human p7_794b PLLLLLLSLPPRA 4.0 325HCV_G1_NS4b_1941c AARVTQILSSLTITQLLKRLHQWI 6.0 28

aHuman homologue; highly conserved sequence derived from hepatitis C virus, genotype 1.bAnalogue; sequence found in multiple human proteins.cValidated promiscuous, Teff cell ICS derived from hepatitis C virus, genotype 1 [17].

Research Article

Please cite this article in press as: Losikoff PT et al. HCV epitope, homologous to multiple human protein sequences, induces a regulatory T cell responsein infected patients. J Hepatol (2014), http://dx.doi.org/10.1016/j.jhep.2014.08.026

6 Journal of Hepatology 2014 vol. xxx j xxx–xxx

JHEPAT 5306 No. of Pages 9

20 October 2014

Predicted Treg-activating HCV sequence possesses TCR faces shared by numerous human proteins Epitope networks are shown, illustrating the abundance of TCR faces one HCV peptide shares with the human genome asdetermined by JanusMatrix analysis. The HCV source peptide is represented by a green diamond, its constituent 9-mer epitopes bygray squares, their cross-conserved partners in the human genome by blue triangles, and the source human proteins by light purplecircles

265 lating in the peripheral blood of chronically-infected patients,266 confirmed in the current study (Fig. 2), provided an initial indica-267 tion of the role of Treg cells in the pathogenesis of chronic hepa-268 titis C [2,3,5,11]. It remained unclear until recently, however,269 whether this increase represented the HCV epitope-specific270 response of Treg cells or the nonspecific consequence of chronic271 inflammation and liver disease [7].272 The ability of HCV-derived epitopes to stimulate Treg cell273 responses is well documented; epitopes associated with both274 structural and non-structural HCV proteins have been reported275 [20,26]. Using HLA class II-peptide tetramer complexes, other276 investigators quantified and characterized the response of Treg

277 cell specific for single HCV epitopes [20,26]. The study described278 herein is the first, however, to identify a promiscuous HCV pep-279 tide sequence (HCV_G1_p7_794) that exhibits extensive human280 homology and the ability to elicit a Treg cell response in a wide,281 HLA-diverse human population, such as the infected patient pop-282 ulation shown in Table 1. Unlike the Treg cell epitopes described283 previously, HCV_G1_p7_794 is an ICS composed of multiple284 HLA binding motifs, able to bind a number of DRB1 alleles. Unfor-285 tunately, this precludes construction of an HLA-peptide tetramer286 complex and quantitation of the HCV_G1_p7_794 responsive287 cells. In accord with the literature, HCV_G1_p7_794 added to288 PBMCs cultures derived from HCV-infected patients, but not289 non-infected individuals or patients who cleared infection,290 induced a marked increase in CD3+CD4+FoxP3+ cells [20,26]. In291 addition to expressing CD25, the vast majority of Treg cells

292expressed CD39, a marker that distinguishes FoxP3+ Treg cells293from activated Teff cells that transiently express FoxP3 [6,19].294In contrast to HCV_G1_p7_794, the human peptide analogue295(p7_794) elicited a significant increase in CD3+CD4+FoxP3+ cells296in PBMC cultures derived from non-infected individuals with no297evidence of prior HCV exposure, as well as HCV-infected patients.298This finding is congruent with the suggestion that viral epitopes299with human homology influence the pathogenesis of chronic HCV300by activating pre-existing, cross-reactive nTreg cells [11]. Indeed,301an extensive homology between the HCV polyprotein and pro-302teins that comprise the human proteome is well documented303[27,28]. JanusMatrix, a bioinformatics algorithm that interrogates304potential T cell epitopes from both their HLA-binding and TcR-305facing aspects, confirmed the existence of significant homology306between HCV_G1_p7_794 and proteins that compose the human307proteome. The results of this analysis demonstrate the potential308efficacy of JanusMatrix in identifying pathogen-encoded epitopes

100

80

60

40

20

0

10

5

0 103 104 105

Comp-FITC-A::-CFSE

Patient ID#229 #261 #265

% o

f M

ax

A

proliferation

MediumAnti-CD3HCV_G1_p7_794Anti-CD3 + HCV_G1_p7_794

MediumHCV_G1_p7_794Anti-CD3 Anti-DC3 + HCV_G1_p7_794IL-2IL-2 + HCV_G1_p7_794

B

3H

-Th

ym

idin

e in

co

rpo

ratio

n (

cp

m x

10

3)

Fig. 3. HCV_G1_p7_794 suppresses the proliferation of PBMCs derived fromHCV-infected patients. CSFE-labelled, Ab+VL+ PBMCs were cultured withmedium alone, or medium containing anti-CD3, HCV_G1_p7_794, or anti-CD3and HCV_G1_p7_794. Cells were collected after 5 days; proliferation wasestimated by flow cytometry as a loss in fluorescence intensity. Data wereobtained in a single experiment representative of two HCV-infected patients (A).PBMCs obtained from the three patients (#229, #261, #265) were cultured 5 daysin the presence of anti-CD3 or 20 ng/ml IL-2 with or without HCV_G1_p7_794;cell proliferation was estimated by [3H]-thymidine incorporation during the last18 h of incubation (B).

2

4

6

8

10

A

B

2

0

0

4

6

8

10

*

*

**

*

**

%C

D3

+C

D4

+F

oxP

3+ c

ells

%C

D3

+C

D4

+F

oxP

3+ c

ells

Ab+VL+ Ab-VL- Ab+VL-

Ab+VL+ Ab-VL-

Medium aloneHCV_G1_p7_794HCV_G1_NS4b_1941

Medium alonep7_794 analog

Fig. 2. HCV_G1_p7_794 fails to elicit an increase in CD3+CD4+FoxP3+ cellsamong PBMCs derived from non-infected individuals. (A) PBMCs obtainedfrom infected patients (Ab+VL+, n = 4), patients who cleared infection (Ab+VL!,n = 6) and non-infected controls (Ab!VL!, n = 4) were cultured with mediumalone, HCV_G1_p7_794 or HCV_G1_NS4b_1941. Cells were collected after 5 daysand were analysed by flow cytometry. ⁄Significantly different: ⁄p = 0.014;⁄⁄p <0.001. (B) PBMCs obtained from Ab+VL+ patients (n = 4) and Ab!VL! controls(n = 4) were cultured in the presence or absence of the human p7_794 analogue.Significantly more CD3+CD4+FoxP3+ cells were recovered from PBMCs culturedwith the analogue than from medium alone: ⁄p = 0.001; ⁄⁄p = 0.048.

JOURNAL OF HEPATOLOGY

Please cite this article in press as: Losikoff PT et al. HCV epitope, homologous to multiple human protein sequences, induces a regulatory T cell responsein infected patients. J Hepatol (2014), http://dx.doi.org/10.1016/j.jhep.2014.08.026

Journal of Hepatology 2014 vol. xxx j xxx–xxx 5

JHEPAT 5306 No. of Pages 9

20 October 2014

309 that elicit the activity of nTreg cells, which normally function to310 suppress autoimmune reactivity to self-antigens (proteins). In311 this regard, it is pertinent to remark that HCV_G1_p7_794 is com-312 prised of epitopes that are homologous to those found in hun-313 dreds of human proteins. This suggests that the autoimmune314 response to a large number of proteins is inhibited by a single315 or limited number of nTreg cell clones, responsive to a common316 peptide sequence, rather than a large number of clones each317 responsive to a unique sequence in a single protein only. The oli-318 goclonality of the Treg cell response to HCV_G1_p7_794 and its319 human analogue is a matter of ongoing investigation.

320The majority of individuals in our patient population were321infected with HCV genotype 1, the derivation of HCV_G1_p7_794.322The addition of HCV_G1_p7_794 to PBMC cultures derived from a323single HCV genotype 3-infected patient, however, also resulted in324a marked increase in CD3+CD4+FoxP3+CD25+ cells (Supplemen-325tary Fig. 2). It is pertinent to note, in this regard, that HCV geno-326type 3 encodes a peptide sequence: LALLVLLLPQRAYAW, which327exhibits HCV_G1_p7_794 homology.328Conceivably, other viral pathogens that cause chronic disease,329e.g., herpes simplex, Epstein-Barr, human immunodeficiency and330cytomegalovirus, also avoid or reduce Teff cell responses by331exploiting similarity to self and activating nTreg cells [29,30].332Indeed, recent analyses indicated that Epstein-Barr virus and333cytomegalovirus contained fewer T cell epitopes and exhibited334higher cross-reactivity with the human genome than did either335Ebola or Marburg virus [14]. Ebola and Marburg viruses, on the336other hand, were composed of significantly fewer peptide337sequences that were cross-reactive with human and expressed338a larger number of predicted T cell epitopes. Thus, viruses that339cause acute disease and viruses such as HCV, which adapt to340humans and cause chronic infection, may differ substantially in341terms of their Treg cell epitope content.342Although immunosuppression by Treg cells is readily343demonstrated in mice, demonstrating the suppressor activity of344human CD3+CD4+CD25+FoxP3+ Treg cells has proven problematic345[31]. Recent studies indicate that the nature of the responder T346cells (CD4+CD25! vs. CD4+CD25low) and the ratio of Treg cells to347responder cells exerts significant effects on the outcome of the348suppression assays [32,33]. Nonetheless, the HCV_G1_p7_794349responsive Treg cells suppressed the mitogenic response of cells350derived from HCV-infected patients in the experiments reported351here despite the fact that the CD3+CD4+FoxP3! responder cells far352outnumbered CD3+CD4+CD25+FoxP3+ suppressor cells among353total PBMCs by a greater than 10:1 ratio.354Notably, the addition of HCV_G1_p7_794 alone to PBMC355cultures derived from HCV-infected (Ab+VL+) patients failed to356induce cell proliferation despite a marked (3- to 4-fold) increase357in CD3+CD4+CD25+FoxP3+ cell number. This finding indicates358that HCV_G1_p7_794 induces the conversion of conventional359CD4+FoxP3! T cells to Treg cells, i.e., infectious tolerance, a sugges-360tion supported by studies demonstrating the inability of nTreg cells361to proliferate in response to their cognate antigen in vitro [34]. Fur-362thermore, in contrast to Ab+VL+ PBMCs cultured in medium alone,363only a minority of CD3+CD4+FoxP3+ cells cultured in the presence364of HCV_G1_p7_794 expressed CD304 (neuropilin), which is365expressed by a subset of FoxP3+ Treg cells in humans and associated366specifically with nTreg cells in mice [21,22]. While it has been sug-367gested alternatively that the expanded Treg cell population in368chronic, HCV-infected patients is composed of cells phenotypically

FoxP3

FoxP

3

CD

304

1.61% 69.20%

CD4

CD4 FoxP3

9.22% 22.86%

FoxP

3

B

CD

304

FoxP3 CD4 CD304

Isotype controls

A

Count

105

104

103

0

105

104

103

0

800

600

400

200

0

1000

800

600

400

200

0

1000

800

600

400

200

0

0 103 104 105 0 103 104 105

0 103 104 105 0 103 104 105

101 102 103 104 105101 102 103 104 105101 102 103 104 105

105

104

103

0

105

104

103

0

Fig. 4. Fewer HCV-G1_p7_794 responsive Treg cells express CD304 (neuropilin).PBMCs obtained from an HCV-infected patient (representative of 4 patients) wereincubated in medium alone (A) or medium that contained HCV_G1_p7_794 (B).The cells were collected on day 5, stained and analysed by flow cytometry. Panelson the right indicate the percentage of CD4+FoxP3+ cells in each population thatexpress CD304.

Table 2. JanusMatrix analysis.

Description ICS # Cross-reactive epitopes per ICS

# Human proteins containing cross-reactive epitopes

HCV_G1_p7_794a WPLLLLLLALPQRAYAQ 5.0 264Human p7_794b PLLLLLLSLPPRA 4.0 325HCV_G1_NS4b_1941c AARVTQILSSLTITQLLKRLHQWI 6.0 28

aHuman homologue; highly conserved sequence derived from hepatitis C virus, genotype 1.bAnalogue; sequence found in multiple human proteins.cValidated promiscuous, Teff cell ICS derived from hepatitis C virus, genotype 1 [17].

Research Article

Please cite this article in press as: Losikoff PT et al. HCV epitope, homologous to multiple human protein sequences, induces a regulatory T cell responsein infected patients. J Hepatol (2014), http://dx.doi.org/10.1016/j.jhep.2014.08.026

6 Journal of Hepatology 2014 vol. xxx j xxx–xxx

JHEPAT 5306 No. of Pages 9

20 October 2014

• HCV_G1_p7_794 expands Tregs in chronic HCV-infected individuals but not in those who clear infection or are not infected.

• Its human analog expands Tregs in uninfected and chronic infected subjects, while a control peptide HCV_G1_NS4b_194, does not for any group.

• CD3+CD4+FoxP3+ cells from chronic HCV-infected individuals, cultured in the absence of HCV_G1_p7_794 express CD304 (neuropilin), indicative of nTreg cells.

• The vast majority of CD3+CD4+FoxP3+ cells, cultured in the presence of HCV_G1_p7_794, were CD304-

characteristic of iTreg cells.

Source 9-mer epitope

Source peptide

Source human protein

TCR-conserved human 9-mer epitope

• Pentatopes were constructed from mutated sequences derived from colon carcinoma model CT26 (study performed by Kreiter et al.).

• Pentatope2 showed greater antitumor activity compared to Pentatope1 in BALB/c mice inoculated with CT26 tumor cells.

• EpiMatrix identified a higher number of neo-epitopes in Pentatope2 compared to Pentatope1. Sequences from Pentatope2 also have a decreased potential to cross-react with other murine proteins, as identified by JanusMatrix.

• Moise L, Gutierrez A, Kibria F, Martin R, Tassone R, Liu R, Terry F, Martin B, De Groot AS. iVAX: An integrated toolkit for the selection and optimization of antigens and the design of epitope-driven vaccines. Hum Vaccin Immunother. 2015;11(9):2312-21.

• Moise L, Gutierrez AH, Bailey-Kellogg C, Terry F, Leng Q, Abdel Hady KM, VerBerkmoes NC, Sztein MB, Losikoff PT, Martin WD, Rothman AL, De Groot AS. The two-faced T cell epitope: examining the host-microbe interface with JanusMatrix. Hum Vaccin Immunother. 2013 Jul;9(7):1577-86.

• Losikoff PT, Mishra S, Terry F, Gutierrez A, Ardito MT, Fast L, Nevola M, Martin WD, Bailey-Kellogg C, De Groot AS, Gregory SH. HCV epitope, homologous to multiple human protein sequences, induces a regulatory T cell response in infected patients. J Hepatol. 2015 Jan;62(1):48-55.

• Kreiter S, Vormehr M, van de Roemer N, Diken M, Löwer M, Diekmann J, Boegel S, Schrörs B, Vascotto F, Castle JC, Tadmor AD, Schoenberger SP, Huber C, Türeci Ö, Sahin U. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature. 2015 Apr 30;520(7549):692-6.

Extended Data Figure 3b.Kreiter et al., Nature 520, 692–696. 2015

• EpiMatrix identifies neo-epitopes by analyzing the binding potential of normal and mutated peptides.

• JanusMatrix further down selects candidate epitopes based on their potential to cross-react with self-proteins.

Neo-epitope selection pipeline EpiMatrix analysis of cancer vaccine efficacy

Tumor Biopsy Expressed Mutation Discovery Neo-Antigen Discovery

IMMUNOINFORMATICS

Mutanome

eliminate

MHC face

TCR face

TCR-face analysis

non-selfself

mutation

MHC binder

MHC binder

neo-epitope

mutation

non-binder

neo-epitope

eliminate vaccine candidate

JanusMatrix analysis

identical TCR-facedifferent TCR-face

Normal

Tumor

Tumorcell

CD8+

T cell

CD4+

T cell

APC

Immunotherapy

Adapted from Schumacher and Schreiber Science 348, 69-74. 2015