'Final common pathway' of human cancer immunotherapy: targeting random somatic mutations

Eric Tran, Paul F Robbins, Steven A Rosenberg, Eric Tran, Paul F Robbins, Steven A Rosenberg

Abstract

Effective clinical cancer immunotherapies, such as administration of the cytokine IL-2, adoptive cell transfer (ACT) and the recent success of blockade of the checkpoint modulators CTLA-4 and PD-1, have been developed without clear identification of the immunogenic targets expressed by human cancers in vivo. Immunotherapy of patients with cancer through the use of ACT with autologous lymphocytes has provided an opportunity to directly investigate the antigen recognition of lymphocytes that mediate cancer regression in humans. High-throughput immunological testing of such lymphocytes in combination with improvements in deep sequencing of the autologous cancer have provided new insight into the molecular characterization and incidence of anti-tumor lymphocytes present in patients with cancer. Here we highlight evidence suggesting that T cells that target tumor neoantigens arising from cancer mutations are the main mediators of many effective cancer immunotherapies in humans.

Conflict of interest statement

COMPETING FINANCIAL INTERESTS

The authors declare no competing financial interests.

Figures

Figure 1
Figure 1
Identification of neoantigen-reactive T cells from patients with cancer. Next-generation sequencing (whole exome and whole transcriptome) is performed on tumor and matched normal cells to identify non-synonymous somatic mutations expressed by the cancer (left). Next, two approaches that do not rely on predictions of HLA–peptide binding can be used to investigate the reactivity of T cells to neoantigens encoded by the identified mutations. In the first approach (middle), minigenes encoding the mutation flanked by nucleotides encoding 12 amino acids from the wild-type gene can be synthesized in tandem to create TMG constructs, which are then cloned into an appropriate expression vector. Linking multiple minigenes in tandem allows a relatively large number of mutations to be evaluated at once. Plasmids encoding TMGs or TMG RNAs transcribed in vitro are then introduced into the appropriate antigen-presenting cells (APCs), such as autologous dendritic cells or B cells, through techniques such as electroporation or lipid-based transfection, to allow processing and presentation of the neoantigens in the context of the patient’s own HLA class I and II molecules. T cells derived from tumor (TILs) or from the blood (right) are then co-cultured with the antigen-presenting cells expressing the TMGs, and T cell reactivity is evaluated by immunological methods such as cytokine ELISPOT or ELISA or the analysis of T cell–activation molecules such as CD137 (4–1BB) or CD134 (OX40) by flow cytometry (far right). The second approach (bottom) is identical to the first approach, except that instead of genetic constructs encoding the mutations, long peptides containing the mutant amino acid flanked by 12 amino acids from the wild-type protein are synthesized and then pulsed onto antigen-presenting cells, which process and present the mutant peptides to T cells. Similar to the minigenes and TMG concept, in this approach, a variable number of individual long peptides can be combined to generate peptide pools, which increases the throughput of neoantigen screening.

References

    1. Robbins PF et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin. Cancer Res 21, 1019–1027 (2015).
    1. Kerkar SP et al. MAGE-A is more highly expressed than NY-ESO-1 in a systematic immunohistochemical analysis of 3668 cases. J. Immunother 39, 181–187 (2016).
    1. Morgan RA et al. Cancer regression and neurological toxicity following anti- MAGE-A3 TCR gene therapy. J. Immunother 36, 133–151 (2013).
    1. Linette GP et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood 122, 863–871 (2013).
    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 114, 535–546 (2009).
    1. Kochenderfer JN et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 116, 4099–4102 (2010).
    1. Brentjens RJ et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118, 4817–4828 (2011).
    1. Porter DL, Levine BL, Kalos M, Bagg A & June CH Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med 365, 725–733 (2011).
    1. Lee DW et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385, 517–528 (2015).
    1. Kochenderfer JN et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J. Clin. Oncol 33, 540–549 (2015).
    1. Turtle CJ et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci. Transl. Med 8, 355ra116 (2016).
    1. Parkhurst MR et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol. Ther 19, 620–626 (2011).
    1. Rosenberg SA, Yang JC & Restifo NP Cancer immunotherapy: moving beyond current vaccines. Nat. Med 10, 909–915 (2004).
    1. Schiller JT & Lowy DR Understanding and learning from the success of prophylactic human papillomavirus vaccines. Nat. Rev. Microbiol 10, 681–692 (2012).
    1. de Vos van Steenwijk PJ et al. The long-term immune response after HPV16 peptide vaccination in women with low-grade pre-malignant disorders of the uterine cervix: a placebo-controlled phase II study. Cancer Immunol. Immunother 63, 147–160 (2014).
    1. Stevanović S et al. Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells. J. Clin. Oncol 33, 1543–1550 (2015).
    1. De Plaen E et al. Immunogenic (tum-) variants of mouse tumor P815: cloning of the gene of tum- antigen P91A and identification of the tum- mutation. Proc. Natl. Acad. Sci. USA 85, 2274–2278 (1988).
    1. Sibille C et al. Structure of the gene of tum- transplantation antigen P198: a point mutation generates a new antigenic peptide. J. Exp. Med 172, 35–45 (1990).
    1. Van Pel A et al. Tum- antigens, TSTA, and T cell immune surveillance. Ann. NY Acad. Sci 636, 43–51 (1991).
    1. Monach PA, Meredith SC, Siegel CT & Schreiber H A unique tumor antigen produced by a single amino acid substitution. Immunity 2, 45–59 (1995).
    1. Dubey P et al. The immunodominant antigen of an ultraviolet-induced regressor tumor is generated by a somatic point mutation in the DEAD box helicase p68. J. Exp. Med 185, 695–705 (1997).
    1. Beck-Engeser GB et al. Point mutation in essential genes with loss or mutation of the second allele: relevance to the retention of tumor-specific antigens. J. Exp. Med 194, 285–300 (2001).
    1. Matsushita H et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 482, 400–404 (2012).
    1. Castle JC et al. Exploiting the mutanome for tumor vaccination. Cancer Res 72, 1081–1091 (2012).
    1. Kreiter S et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 520, 692–696 (2015).
    1. Gubin MM et al. Checkpoint blockade cancer immunotherapy targets tumourspecific mutant antigens. Nature 515, 577–581 (2014).
    1. Yadav M et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 515, 572–576 (2014).
    1. Motzer RJ et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J. Clin. Oncol 33, 1430–1437 (2015).
    1. Motzer RJ et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N. Engl. J. Med 373, 1803–1813 (2015).
    1. Nghiem PT et al. PD-1 blockade with Pembrolizumab in advanced Merkel-cell carcinoma. N. Engl. J. Med 374, 2542–2552 (2016).
    1. Topalian SL et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med 366, 2443–2454 (2012).
    1. van Rooij N et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J. Clin. Oncol 31, e439–e442 (2013).
    1. Snyder A et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med 371, 2189–2199 (2014).
    1. Van Allen EM et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350, 207–211 (2015).
    1. Hugo W et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 165, 35–44 (2016).
    1. Rizvi NA et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol 16, 257–265 (2015).
    1. Rizvi NA et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).
    1. Powles T et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature 515, 558–562 (2014).
    1. Le DT et al. PD-1 Blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med 372, 2509–2520 (2015).
    1. McGranahan N et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016).
    1. Rosenberg SA et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res 17, 4550–4557 (2011).
    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 34, 2389–2397 (2016).
    1. Robbins PF et al. A mutated β-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J. Exp. Med 183, 1185–1192 (1996).
    1. Huang J et al. T cells associated with tumor regression recognize frameshifted products of the CDKN2A tumor suppressor gene locus and a mutated HLA class I gene product. J. Immunol 172, 6057–6064 (2004).
    1. Zhou J, Dudley ME, Rosenberg SA & Robbins PF Persistence of multiple tumor-specific T-cell clones is associated with complete tumor regression in a melanoma patient receiving adoptive cell transfer therapy. J. Immunother 28, 53–62 (2005).
    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 190, 6034–6042 (2013).
    1. Robbins PF et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat. Med 19, 747–752 (2013).
    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 4, 669–678 (2016).
    1. Linnemann C et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat. Med 21, 81–85 (2015).
    1. Rodenko B et al. Generation of peptide-MHC class I complexes through UV-mediated ligand exchange. Nat. Protoc 1, 1120–1132 (2006).
    1. Cohen CJ et al. Isolation of neoantigen-specific T cells from tumor and peripheral lymphocytes. J. Clin. Invest 125, 3981–3991 (2015).
    1. Kalaora S et al. Use of HLA peptidomics and whole exome sequencing to identify human immunogenic neo-antigens. Oncotarget 7, 5110–5117 (2016).
    1. Lu YC et al. Efficient identification of mutated cancer antigens recognized by T cells associated with durable tumor regressions. Clin. Cancer Res 20, 3401–3410 (2014).
    1. Gros A et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med 22, 433–438 (2016).
    1. Parkhurst MR et al. Isolation of T cell receptors reactive with tumor neoantigens from tumor infiltrating lymphocytes based on CD137 expression. Clin. Cancer Res (8 November 2016).
    1. Lawrence MS et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).
    1. Hogan KT et al. The peptide recognized by HLA-A68.2-restricted, squamous cell carcinoma of the lung-specific cytotoxic T lymphocytes is derived from a mutated elongation factor 2 gene. Cancer Res 58, 5144–5150 (1998).
    1. Karanikas V et al. High frequency of cytolytic T lymphocytes directed against a tumor-specific mutated antigen detectable with HLA tetramers in the blood of a lung carcinoma patient with long survival. Cancer Res 61, 3718–3724 (2001).
    1. Echchakir H et al. A point mutation in the α-actinin-4 gene generates an antigenic peptide recognized by autologous cytolytic T lymphocytes on a human lung carcinoma. Cancer Res 61, 4078–4083 (2001).
    1. Takenoyama M et al. A point mutation in the NFYC gene generates an antigenic peptide recognized by autologous cytolytic T lymphocytes on a human squamous cell lung carcinoma. Int. J. Cancer 118, 1992–1997 (2006).
    1. Guéguen M et al. An antigen recognized by autologous CTLs on a human bladder carcinoma. J. Immunol 160, 6188–6194 (1998).
    1. Mandruzzato S, Brasseur F, Andry G, Boon T & van der Bruggen P A CASP-8 mutation recognized by cytolytic T lymphocytes on a human head and neck carcinoma. J. Exp. Med 186, 785–793 (1997).
    1. Ito D et al. Immunological characterization of missense mutations occurring within cytotoxic T cell-defined p53 epitopes in HLA-A*0201+ squamous cell carcinomas of the head and neck. Int. J. Cancer 120, 2618–2624 (2007).
    1. Wick DA et al. Surveillance of the tumor mutanome by T cells during progression from primary to recurrent ovarian cancer. Clin. Cancer Res 20, 1125–1134 (2014).
    1. Gjertsen MK, Bjorheim J, Saeterdal I, Myklebust J & Gaudernack G Cytotoxic CD4+ and CD8+ T lymphocytes, generated by mutant p21-ras (12Val) peptide vaccination of a patient, recognize 12Val-dependent nested epitopes present within the vaccine peptide and kill autologous tumour cells carrying this mutation. Int. J. Cancer 72, 784–790 (1997).
    1. Kloor M & Doeberitz MK The immune biology of microsatellite-unstable cancer. Trends in Cancer 2, 121–133 (2016).
    1. Tran E et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 350, 1387–1390 (2015).
    1. Tran E et al. T-cell transfer therapy targeting mutated KRAS in cancer. N. Engl. J. Med 375, 2255–2262 (2016).
    1. Tran E et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 344, 641–645 (2014).
    1. Gattinoni L, Klebanoff CA & Restifo NP Paths to stemness: building the ultimate antitumour T cell. Nat. Rev. Cancer 12, 671–684 (2012).
    1. Crompton JG et al. Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics. Cancer Res 75, 296–305 (2015).
    1. Macintyre AN et al. Protein kinase B controls transcriptional programs that direct cytotoxic T cell fate but is dispensable for T cell metabolism. Immunity 34, 224–236 (2011).
    1. Li Y, Bleakley M & Yee C IL-21 influences the frequency, phenotype, and affinity of the antigen-specific CD8 T cell response. J. Immunol 175, 2261–2269 (2005).
    1. Vizcardo R et al. Regeneration of human tumor antigen-specific T cells from iPSCs derived from mature CD8(+) T cells. Cell Stem Cell 12, 31–36 (2013).
    1. Beane JD et al. Clinical scale zinc finger nuclease-mediated gene editing of PD-1 in Tumor infiltrating lymphocytes for the treatment of metastatic melanoma. Mol. Ther 23, 1380–1390 (2015).
    1. Carreno BM et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science 348, 803–808 (2015).
    1. Coulie PG et al. A mutated intron sequence codes for an antigenic peptide recognized by cytolytic T lymphocytes on a human melanoma. Proc. Natl. Acad. Sci. USA 92, 7976–7980 (1995).

Source: PubMed

3
Abonner