Sensitive and frequent identification of high avidity neo-epitope specific CD8 + T cells in immunotherapy-naive ovarian cancer

Sara Bobisse, Raphael Genolet, Annalisa Roberti, Janos L Tanyi, Julien Racle, Brian J Stevenson, Christian Iseli, Alexandra Michel, Marie-Aude Le Bitoux, Philippe Guillaume, Julien Schmidt, Valentina Bianchi, Denarda Dangaj, Craig Fenwick, Laurent Derré, Ioannis Xenarios, Olivier Michielin, Pedro Romero, Dimitri S Monos, Vincent Zoete, David Gfeller, Lana E Kandalaft, George Coukos, Alexandre Harari, Sara Bobisse, Raphael Genolet, Annalisa Roberti, Janos L Tanyi, Julien Racle, Brian J Stevenson, Christian Iseli, Alexandra Michel, Marie-Aude Le Bitoux, Philippe Guillaume, Julien Schmidt, Valentina Bianchi, Denarda Dangaj, Craig Fenwick, Laurent Derré, Ioannis Xenarios, Olivier Michielin, Pedro Romero, Dimitri S Monos, Vincent Zoete, David Gfeller, Lana E Kandalaft, George Coukos, Alexandre Harari

Abstract

Immunotherapy directed against private tumor neo-antigens derived from non-synonymous somatic mutations is a promising strategy of personalized cancer immunotherapy. However, feasibility in low mutational load tumor types remains unknown. Comprehensive and deep analysis of circulating and tumor-infiltrating lymphocytes (TILs) for neo-epitope specific CD8+ T cells has allowed prompt identification of oligoclonal and polyfunctional such cells from most immunotherapy-naive patients with advanced epithelial ovarian cancer studied. Neo-epitope recognition is discordant between circulating T cells and TILs, and is more likely to be found among TILs, which display higher functional avidity and unique TCRs with higher predicted affinity than their blood counterparts. Our results imply that identification of neo-epitope specific CD8+ T cells is achievable even in tumors with relatively low number of somatic mutations, and neo-epitope validation in TILs extends opportunities for mutanome-based personalized immunotherapies to such tumors.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Identification of neo-epitope specific PBLs. a, b Representative example of peripheral blood CD8+ T lymphocytes (PBLs) response against the HHATL75F neo-epitope in patient CTE-0013. a T-cell reactivity measured by IFNγ ELISpot against the neo-epitope (mut) from HHATL75F gene but not against the wild-type (wt) peptide (PHA: phytohemagglutinin; No Ag: no peptide). Shown are the average of triplicate + SD. b Representative example of validation of neo-epitope specific CD8+ T cells using peptide-MHC multimers. c T-cell reactivity measured by IFNγ ELISpot against the neo-epitope from KIR2DS4I7S gene but not against the wild-type peptide in patient CTE-0012 (PHA: phytohemagglutinin; No Ag: no peptide). Shown are the average of triplicate + SD. d Representative example of neo-epitope validation by polychromatic intracellular cytokine staining (ICS) in patient CTE-0012 without (No Ag) or after stimulation with a cognate neo-epitope (KIR2DS4I7S). All other PBL neo-epitopes are described in Supplementary Fig. 1 and Table 2. e Proportion of EOC patients with documented peripheral blood CD8+ T-cell (PBL) response against neo-epitopes. f Frequency and g number (mean ± SEM) of neo-epitope specific CD8+ PBLs per patient, detected after one round of in vitro stimulation (Supplementary Table 2). h Cumulative analysis showing the cytokine profiles of neo-epitope specific CD8+ PBLs. The SPICE analysis represents the functional composition of cytokine-producing neo-epitope specific CD8+ T cells (mean ± SEM). i Heatmap showing expression of all the genes from Reactome’s antigen processing and cross-presentation pathway, . The number of neo-epitopes recognized by PBL of each patient is shown on top of the heatmap. Genes from this pathway are ordered based on the differential expression fold change between patients with or without PBL neo-epitope recognition. j Gene set enrichment analysis (GSEA) curves for various Reactome pathways significantly different between patients with or without PBL neo-epitope recognition. Vertical bars indicate the ranked positions of each gene from the respective gene set. Statistical significance of these enrichments is reported by the FDR of the GSEA. Corresponding heatmaps for IFNγ signaling, PD-1 signaling and collagen formation are reported in Supplementary Fig. 2
Fig. 2
Fig. 2
Identification of neo-epitope specific TILs. ac Representative example of neo-epitope (ZCCHC11P1265H)-specific TILs in patient CTE-0015. a IFNγ ELISpot showing the average of triplicate + SD (PHA: phytohemagglutinin; No Ag: no peptide) and peptide-MHC multimer staining. All other neo-epitopes are described in Supplementary Fig. 3 and Table 2. b Intracellular cytokine staining showing the frequency of viable IFNγ, TNFα, and IL-2 cytokine-producing TILs. c Proportion of patients with neo-epitope reactive TILs. d Cumulative analysis (SPICE) showing the functional composition (cytokine profiles) of neo-epitope specific TILs (mean ± SD). e Repertoire of neo-epitopes in PBLs and TILs in 19 patients. Each colored square represents one neo-epitope for which T-cell reactivity was validated either exclusively in PBLs (blue), exclusively in TILs (red), in both compartments (orange) or in none of them (white). For 5 out of 19 patients no tumor samples were available for TIL generation (black squares). f Neo-epitope-specific TILs from patient CTE-0011 were FACS sorted with HLA-A1101-SEPT9R289H multimer. Sorted cells (left) underwent TCR sequencing, as well as autologous bulk PBLs, bulk tumor and bulk TIL cultures. The Manhattan plot reports the V/J recombination of the T-cell receptor β (TCRβ) of SEPT9R289H-specific T cells: V and J segments are represented according to chromosomal location on the x and y-axis, respectively. On the right, the distribution of the TCR frequencies is shown for PBL, tumor and TILs. Dominant SEPT9R289H-specific TCRβ are identified by red bars (and arrows) in tumor and TILS, whereas no SEPT9R289H-specific TCRs was identified in PBLs. gTRIM26G497W-specific circulating T cells from patient CTE-0010 were isolated based on CD137 upregulation and immediately processed for TCR-sequencing analysis (center). TCR repertoire analysis was performed in parallel on matched bulk PBLs, bulk tumor, and bulk TIL cultures (right). The most frequent shared neo-epitope specific TCR was ranked according to its relative frequency in PBL and tumor (red bars and arrows). The same TCR was undetectable in IL-2-expanded TIL cultures. Elements of box plots represent median (line), 25% and 75% confidence limit (box limits) and 10% and 90% confidence limit (whiskers)
Fig. 3
Fig. 3
TIL cultures can be enriched for neo-epitope specific T cells. a Schematic representation of the alternative procedures for TIL expansion. Single cell tumor suspensions were plated in the presence of high-dose IL-2 alone (conventional TILs) or alternatively, in the presence of high-dose IL-2 supplemented with pools of predicted mutated peptides (primed TILs). b Representative examples of conventional and primed TILs, interrogated for the presence of neo-epitope specific TILs by multimer staining. c Cumulative analysis of the frequencies of neo-epitope specific CD8+ T lymphocytes detected in conventional (x-axis) and primed (y-axis) TIL cultures. d Proportions of patients with documented neo-epitope specific T-cell responses in the different compartments and within the distinct TIL culture conditions. e Landscape of neo-epitope specific T-cell responses in circulating, conventional TILs and primed TILs in nine patients for which primed TILs were available. T-cell responses against mutated epitopes identified exclusively in PBLs (blue), in TILs (red), in both the compartments (orange) or in none of them (white) are indicated
Fig. 4
Fig. 4
Functional analyses of neo-epitope specific PBLs and TILs. a Higher functional avidity of HHATL75F-specific TIL and PBL clones. Shown are the relative frequencies of IFNγ-producing CD8+ T cells (mean ± SD). b Higher functional avidity (mean ± SEM) of HHATL75F-specific TILs compared with PBL clones based on TNFα and IL-2 release (Supplementary Fig. 5 shows raw data). c Higher cytolytic capacity of HHATL75F-specific TILs compared with PBLs. Data show the percentage of lysis of T2 cells pulsed with different concentrations of the cognate neo-epitope. d Mass cytometry analysis showing the relative phosphorylation of ERK1/2 and S6 proteins involved in TCR-signaling pathways following CD3 stimulation of PBL and TIL clones. Graphs represent mean ± SEM. Supplementary Fig. 6 shows raw data. e Mass cytometry analysis showing the relative expression of different markers on neo-epitope specific PBLs and TILs. Naive and memory bulk peripheral blood CD8+ T cells are shown for comparison. f Analysis of the repertoire of the T-cell receptor β (TCRβ) V-J segment recombination of HHATL75F-specific CD8+ T cells isolated from either PBLs (top) or TILs (bottom) after FACS sorting using multimers. V and J segments are represented according to chromosomal location on the x and y-axis, respectively. Arrows identify dominant TCRβ sequences that were associated to TCRα sequences shown in Supplementary Fig. 8. g Calculated TCR/pMHC complexes for PBL and TIL-related TCRs: HHATL75F-PBL-hTRAV05 + hTRAJ34/hTRBV11-2 + hTRBJ02-7 on the top, and HHATL75F-TIL-hTRAV38-2 + hTRAJ33/hTRBV12-3 + hTRBJ02-3 at the bottom. The peptide is shown in ball and stick, colored according to the atom types. MHC ribbon is colored in brown, with residues displayed in thick lines and colored according to the atom types, with carbon colored in brown. TCRα ribbon is colored in light blue, with residues displayed in thick lines and colored according to the atom types, with carbon colored in light blue. TCRβ ribbon is colored in pink, with residues displayed in thick lines and colored according to the atom types, with carbon colored in pink. Hydrogen bonds and ionic interactions are shown as thin blue lines. Dotted blue lines indicate hydrogen bonds accessible through thermal fluctuations. Residues are labeled in brown, black, blue, and pink, for MHC, peptide, TCRα, and TCRβ, respectively
Fig. 5
Fig. 5
Functional analyses of neo-epitope specific PBLs and TILs. a Representative analyses of the functional avidity of neo-epitope specific PBLs and TILs as assessed by IFNγ ELISpot. HS6ST1S405I-specific PBLs from patient CTE-0005 and ZCCHC11P1265H-specific TILs from patient CTE-0015 were stimulated with serial dilutions of cognate neo-epitopes or native (wt) peptides. Data show mean ± SEM of technical replicates. b Cumulative analysis (individual values and median) of the functional avidity of neo-epitope specific CD8+ PBLs and TILs (raw data are shown in Supplementary Fig. 10). c Analysis of ODZ3A2490V-specific (top, high avidity) and SEPT9R289H-specific (bottom, low-avidity) TCRs (red bars and arrows) among total TCRs from the autologous tumor samples of patients CTE-0015 and CTE-0011, respectively. Elements of box plots represent median (line), 25% and 75% confidence limit (box limits) and 10% and 90% confidence limit (whiskers)

References

    1. Goff SL, et al. Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J. Clin. Oncol. 2016;34:2389–2397. doi: 10.1200/JCO.2016.66.7220.
    1. Rosenberg SA, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res. 2011;17:4550–4557. doi: 10.1158/1078-0432.CCR-11-0116.
    1. Barrett DM, Grupp SA, June CH. Chimeric antigen receptor- and tcr-modified t cells enter main street and wall street. J. Immunol. 2015;195:755–761. doi: 10.4049/jimmunol.1500751.
    1. Johnson LA, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114:535–546. doi: 10.1182/blood-2009-03-211714.
    1. Linette GP, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013;122:863–871. doi: 10.1182/blood-2013-03-490565.
    1. Maude SL, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Eng. J. Med. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222.
    1. Linnemann C, et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4 + T cells in human melanoma. Nat. Med. 2015;21:81–85. doi: 10.1038/nm.3773.
    1. Rajasagi M, et al. Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia. Blood. 2014;124:453–462. doi: 10.1182/blood-2014-04-567933.
    1. Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128. doi: 10.1126/science.aaa1348.
    1. Robbins PF, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat. Med. 2013;19:747–752. doi: 10.1038/nm.3161.
    1. Snyder A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Eng. J. Med. 2014;371:2189–2199. doi: 10.1056/NEJMoa1406498.
    1. Tran E, et al. Cancer immunotherapy based on mutation-specific CD4 + T cells in a patient with epithelial cancer. Science. 2014;344:641–645. doi: 10.1126/science.1251102.
    1. van Rooij N, et al. Tumor exome analysis reveals neo-antigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J. Clin. Oncol. 2013;31:e439–e442. doi: 10.1200/JCO.2012.47.7521.
    1. Van Allen EM, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350:207–211. doi: 10.1126/science.aad0095.
    1. Tran E, Robbins PF, Rosenberg SA. ‘Final common pathway’ of human cancer immunotherapy: targeting random somatic mutations. Nat. Immunol. 2017;18:255–262. doi: 10.1038/ni.3682.
    1. Robbins PF, et al. A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J. Exp. Med. 1996;183:1185–1192. doi: 10.1084/jem.183.3.1185.
    1. Lu YC, et al. Mutated PPP1R3B is recognized by T cells used to treat a melanoma patient who experienced a durable complete tumor regression. J. Immunol. 2013;190:6034–6042. doi: 10.4049/jimmunol.1202830.
    1. Prickett TD, et al. Durable complete response from metastatic melanoma after transfer of autologous T cells recognizing 10 mutated tumor antigens. Cancer Immunol. Res. 2016;4:669–678. doi: 10.1158/2326-6066.CIR-15-0215.
    1. Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421. doi: 10.1038/nature12477.
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74. doi: 10.1126/science.aaa4971.
    1. Wick DA, et al. Surveillance of the tumor mutanome by T cells during progression from primary to recurrent ovarian cancer. Clin. Cancer Res. 2014;20:1125–1134. doi: 10.1158/1078-0432.CCR-13-2147.
    1. Zhang L, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Eng. J. Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177.
    1. Schlienger K, et al. TRANCE- and CD40 ligand-matured dendritic cells reveal MHC class-I-restricted T cells specific for autologous tumor in late-stage ovarian cancer patients. Clin. Cancer Res. 2003;9:1517–1527.
    1. Santoiemma PP, et al. Systematic evaluation of multiple immune markers reveals prognostic factors in ovarian cancer. Gynecol. Oncol. 2016;143:120–127. doi: 10.1016/j.ygyno.2016.07.105.
    1. Rodriguez-Garcia A, Minutolo NG, Robinson JM, Powell DJ. T-cell target antigens across major gynecologic cancers. Gynecol. Oncol. 2017;145:426–435. doi: 10.1016/j.ygyno.2017.03.510.
    1. Deniger, D. C. P., et al. Mutated tumor neoantigens are recognized by tumor infiltrating lymphocytes from metastatic ovarian cancer in in ASGCT2016, (Washington DC, USA, 2016).
    1. Iseli C, Ambrosini G, Bucher P, Jongeneel CV. Indexing strategies for rapid searches of short words in genome sequences. PLoS. ONE. 2007;2:e579. doi: 10.1371/journal.pone.0000579.
    1. Lundegaard C, et al. NetMHC-3.0: accurate web accessible predictions of human, mouse and monkey MHC class I affinities for peptides of length 8-11. Nucleic Acids Res. 2008;36:W509–W512. doi: 10.1093/nar/gkn202.
    1. Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61. doi: 10.1016/j.cell.2014.12.033.
    1. Brown SD, et al. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res. 2014;24:743–750. doi: 10.1101/gr.165985.113.
    1. Strickland KC, et al. Association and prognostic significance of BRCA1/2-mutation status with neo-antigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget. 2016;7:13587–13598. doi: 10.18632/oncotarget.7277.
    1. Leimgruber A, et al. TCRep 3D: an automated in silico approach to study the structural properties of TCR repertoires. PloS ONE. 2011;6:e26301. doi: 10.1371/journal.pone.0026301.
    1. Harari A, et al. An HIV-1 clade C DNA prime, NYVAC boost vaccine regimen induces reliable, polyfunctional, and long-lasting T cell responses. J. Exp. Med. 2008;205:63–77. doi: 10.1084/jem.20071331.
    1. Harari A, et al. Dominant TNF-alpha + Mycobacterium tuberculosis-specific CD4 + T cell responses discriminate between latent infection and active disease. Nat. Med. 2011;17:372–376. doi: 10.1038/nm.2299.
    1. Schmidt J, et al. Reversible major histocompatibility complex I-peptide multimers containing Ni(2 + )-nitrilotriacetic acid peptides and histidine tags improve analysis and sorting of CD8( + ) T cells. J. Biol. Chem. 2011;286:41723–41735. doi: 10.1074/jbc.M111.283127.
    1. Vigano S, et al. Rapid perturbation in viremia levels drives increases in functional avidity of HIV-specific CD8 T cells. PLoS Pathog. 2013;9:e1003423. doi: 10.1371/journal.ppat.1003423.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8.
    1. Mootha VK, et al. PGC-1 alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 2003;34:267–273. doi: 10.1038/ng1180.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Giudicelli V, Chaume D, Lefranc MP. IMGT/GENE-DB: a comprehensive database for human and mouse immunoglobulin and T cell receptor genes. Nucleic Acids Res. 2005;33:D256–D261. doi: 10.1093/nar/gki010.
    1. Eswar N, Eramian D, Webb B, Shen MY, Sali A. Protein structure modeling with MODELLER. Methods Mol. Biol. 2008;426:145–159. doi: 10.1007/978-1-60327-058-8_8.
    1. Eswar N, et al. Comparative protein structure modeling using Modeller. Curr. Protoc. Bioinformatics. 2006;Chapter 5:6.
    1. Rose PW, et al. The RCSB Protein Data Bank: redesigned web site and web services. Nucleic Acids Res. 2011;39:D392–D401. doi: 10.1093/nar/gkq1021.
    1. Edgar RC. MUSCLE: a multiple sequence alignment method with reduced time and space complexity. BMC Bioinformatics. 2004;5:113. doi: 10.1186/1471-2105-5-113.
    1. Edgar RC. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 2004;32:1792–1797. doi: 10.1093/nar/gkh340.
    1. Zoete V, Irving M, Ferber M, Cuendet MA, Michielin O. Structure-based, rational design of T cell receptors. Front. Immunol. 2013;4:268.
    1. Zoete V, Michielin O. Comparison between computational alanine scanning and per-residue binding free energy decomposition for protein-protein association using MM-GBSA: application to the TCR-p-MHC complex. Proteins. 2007;67:1026–1047. doi: 10.1002/prot.21395.
    1. Irving M, et al. Interplay between T cell receptor binding kinetics and the level of cognate peptide presented by major histocompatibility complexes governs CD8 + T cell responsiveness. J. Biol. Chem. 2012;287:23068–23078. doi: 10.1074/jbc.M112.357673.
    1. MacKerell AD, Jr, Banavali N, Foloppe N. Development and current status of the CHARMM force field for nucleic acids. Biopolymers. 2000;56:257–265. doi: 10.1002/1097-0282(2000)56:4<257::AID-BIP10029>;2-W.
    1. Brooks BR, et al. CHARMM: the biomolecular simulation program. J. Comput. Chem. 2009;30:1545–1614. doi: 10.1002/jcc.21287.
    1. Lee MS, Feig M, Salsbury FR, Jr., Brooks CL. 3rd. New analytic approximation to the standard molecular volume definition and its application to generalized Born calculations. J. Comput. Chem. 2003;24:1348–1356. doi: 10.1002/jcc.10272.
    1. Pettersen EF, et al. UCSF Chimera--a visualization system for exploratory research and analysis. J. Comput. Chem. 2004;25:1605–1612. doi: 10.1002/jcc.20084.
    1. Fabregat A, et al. The reactome pathway knowledgebase. Nucleic Acids Res. 2016;44:D481–D487. doi: 10.1093/nar/gkv1351.
    1. Milacic M, et al. Annotating cancer variants and anti-cancer therapeutics in reactome. Cancers. 2012;4:1180–1211. doi: 10.3390/cancers4041180.

Source: PubMed

3
S'abonner