Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digests

Background Cellular immunotherapies using autologous tumor-infiltrating lymphocytes (TIL) can induce durable regression of epithelial cancers in selected patients with treatment-refractory metastatic disease. As the genetic engineering of T cells with tumor-reactive T-cell receptors (TCRs) comes to the forefront of clinical investigation, the rapid, scalable, and cost-effective detection of patient-specific neoantigen-reactive TIL remains a top priority. Methods We analyzed the single-cell transcriptomic states of 31 neoantigen-specific T-cell clonotypes to identify cell surface dysfunction markers that best identified the metastatic transcriptional states enriched with antitumor TIL. We developed an efficient method to capture neoantigen-reactive TCRs directly from resected human tumors based on cell surface co-expression of CD39, programmed cell death protein-1, and TIGIT dysfunction markers (CD8+ TILTP). Results TILTP TCR isolation achieved a high degree of correlation with single-cell transcriptomic signatures that identify neoantigen-reactive TCRs, making it a cost-effective strategy using widely available resources. Reconstruction of additional TILTP TCRs from tumors identified known and novel antitumor TCRs, showing that at least 39.5% of TILTP TCRs are neoantigen-reactive or tumor-reactive. Despite their substantial enrichment for neoantigen-reactive TCR clonotypes, clonal dynamics of 24 unique antitumor TILTP clonotypes from four patients indicated that most in vitro expanded TILTP populations failed to demonstrate neoantigen reactivity, either by loss of neoantigen-reactive clones during TIL expansion, or through functional impairment during cognate neoantigen recognition. Conclusions While direct usage of in vitro-expanded CD8+ TILTP as a source for cellular therapy might be precluded by profound TIL dysfunction, isolating TILTP represents a streamlined effective approach to rapidly identify neoantigen-reactive TCRs to design engineered cellular immunotherapies against cancer.

[1]  Jonathan M. Hernandez,et al.  Adoptive Cellular Therapy with Autologous Tumor-Infiltrating Lymphocytes and T-cell Receptor–Engineered T Cells Targeting Common p53 Neoantigens in Human Solid Tumors , 2022, Cancer immunology research.

[2]  B. Fox,et al.  Neoantigen T-Cell Receptor Gene Therapy in Pancreatic Cancer. , 2022, The New England journal of medicine.

[3]  Charles H. Yoon,et al.  Landscape of helper and regulatory antitumour CD4+ T cells in melanoma , 2022, Nature.

[4]  J. Gartner,et al.  A phenotypic signature that identifies neoantigen-reactive T cells in fresh human lung cancers. , 2022, Cancer cell.

[5]  Brady Bernard,et al.  Transcriptomic profiles of neoantigen-reactive T cells in human gastrointestinal cancers. , 2022, Cancer cell.

[6]  J. Gartner,et al.  Neoantigen Identification and Response to Adoptive Cell Transfer in anti PD-1 Naïve and Experienced Patients with Metastatic Melanoma. , 2022, Clinical cancer research : an official journal of the American Association for Cancer Research.

[7]  J. Gartner,et al.  Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers , 2022, Science.

[8]  Jonathan M. Hernandez,et al.  Breast Cancers Are Immunogenic: Immunologic Analyses and a Phase II Pilot Clinical Trial Using Mutation-Reactive Autologous Lymphocytes , 2022, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[9]  David R. Jones,et al.  Author Correction: Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers , 2021, Nature.

[10]  E. Wherry,et al.  Author Correction: Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation , 2021, Nature immunology.

[11]  Todd M. Allen,et al.  Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans , 2021, Nature Immunology.

[12]  Shuqiang Li,et al.  Phenotype, specificity and avidity of antitumour CD8+ T cells in melanoma , 2021, Nature.

[13]  Howard Y. Chang,et al.  Transient “rest” restores functionality in exhausted CAR-T cells via epigenetic remodeling , 2021, Science.

[14]  G. Altan-Bonnet,et al.  Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer , 2020, Science.

[15]  Sri Krishna,et al.  Rapid Identification and Evaluation of Neoantigen-reactive T-Cell Receptors From Single Cells , 2020, Journal of immunotherapy.

[16]  S. H. van der Burg,et al.  CD39 Identifies the CD4+ Tumor-Specific T-cell Population in Human Cancer , 2020, Cancer Immunology Research.

[17]  R. Offringa,et al.  The Outcome of Ex Vivo TIL Expansion Is Highly Influenced by Spatial Heterogeneity of the Tumor T-Cell Repertoire and Differences in Intrinsic In Vitro Growth Capacity between T-Cell Clones , 2020, Clinical Cancer Research.

[18]  J. Gartner,et al.  Recognition of human gastrointestinal cancer neoantigens by circulating PD-1+ lymphocytes. , 2019, The Journal of clinical investigation.

[19]  W. Telford,et al.  The transcription factor c-Myb regulates CD8+ T cell stemness and antitumor immunity , 2018, Nature Immunology.

[20]  Douglas B. Johnson,et al.  Biological Consequences of MHC-II Expression by Tumor Cells in Cancer , 2018, Clinical Cancer Research.

[21]  J. Gartner,et al.  Enhanced detection of neoantigen-reactive T cells targeting unique and shared oncogenes for personalized cancer immunotherapy. , 2018, JCI insight.

[22]  B. Fox,et al.  Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors , 2018, Nature Communications.

[23]  J. Gartner,et al.  Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer , 2018, Nature Medicine.

[24]  M. Fehlings,et al.  Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates , 2018, Nature.

[25]  N. Restifo,et al.  Epigenetic control of CD8+ T cell differentiation , 2018, Nature Reviews Immunology.

[26]  E. Jaffee,et al.  Tumor Mutational Burden and Response Rate to PD-1 Inhibition. , 2017, The New England journal of medicine.

[27]  Purvesh Khatri,et al.  Antigen Identification for Orphan T Cell Receptors Expressed on Tumor-Infiltrating Lymphocytes , 2017, Cell.

[28]  S. Hervás-Stubbs,et al.  Expansion of Tumor-Infiltrating CD8+ T cells Expressing PD-1 Improves the Efficacy of Adoptive T-cell Therapy. , 2017, Cancer research.

[29]  S. Rosenberg,et al.  'Final common pathway' of human cancer immunotherapy: targeting random somatic mutations , 2017, Nature Immunology.

[30]  J. Gartner,et al.  T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. , 2016, The New England journal of medicine.

[31]  S. Chanock,et al.  Burden of Nonsynonymous Mutations among TCGA Cancers and Candidate Immune Checkpoint Inhibitor Responses. , 2016, Cancer research.

[32]  Jared Gartner,et al.  Tumor- and Neoantigen-Reactive T-cell Receptors Can Be Identified Based on Their Frequency in Fresh Tumor , 2016, Cancer Immunology Research.

[33]  J. Gartner,et al.  Durable Complete Response from Metastatic Melanoma after Transfer of Autologous T Cells Recognizing 10 Mutated Tumor Antigens , 2016, Cancer Immunology Research.

[34]  M. Donia,et al.  Long-Lasting Complete Responses in Patients with Metastatic Melanoma after Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes and an Attenuated IL2 Regimen , 2016, Clinical Cancer Research.

[35]  S. Rosenberg,et al.  Cancer Immunotherapy Based on Mutation-Specific CD4+ T Cells in a Patient with Epithelial Cancer , 2014, Science.

[36]  S. Rosenberg,et al.  Myeloid Cells Obtained from the Blood but Not from the Tumor Can Suppress T-cell Proliferation in Patients with Melanoma , 2012, Clinical Cancer Research.

[37]  Marianna Sabatino,et al.  Simplified Method of the Growth of Human Tumor Infiltrating Lymphocytes in Gas-permeable Flasks to Numbers Needed for Patient Treatment , 2012, Journal of immunotherapy.

[38]  M. Raffeld,et al.  Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[39]  S. Rosenberg,et al.  Clinical-scale Lentiviral Vector Transduction of PBL for TCR Gene Therapy and Potential for Expression in Less-differentiated Cells , 2008, Journal of immunotherapy.

[40]  S. Rosenberg,et al.  Cancer Regression in Patients After Transfer of Genetically Engineered Lymphocytes , 2006, Science.

[41]  S. Rosenberg,et al.  Persistence of Multiple Tumor-Specific T-Cell Clones Is Associated with Complete Tumor Regression in a Melanoma Patient Receiving Adoptive Cell Transfer Therapy , 2005, Journal of immunotherapy.

[42]  J. Gartner,et al.  Unique neoantigens arise from somatic mutations in patients with gastrointestinal cancers. , 2019, Cancer discovery.