Tumor-Infiltrating T Cells From Clear Cell Renal Cell Carcinoma Patients Recognize Neoepitopes Derived From Point and Frameshift Mutations

Mutation-derived neoantigens are important targets for T cell-mediated reactivity toward tumors and, due to their unique tumor expression, an attractive target for immunotherapy. Neoepitope-specific T cells have been detected across a number of solid cancers with high mutational burden tumors, but neoepitopes have been mostly selected from single nucleotide variations (SNVs), and little focus has been given to neoepitopes derived from in-frame and frameshift indels, which might be equally important and potentially highly immunogenic. Clear cell renal cell carcinomas (ccRCCs) are medium-range mutational burden tumors with a high pan-cancer proportion of frameshift mutations. In this study, the mutational landscape of tumors from six RCC patients was analyzed by whole-exome sequencing (WES) of DNA from tumor fragments (TFs), autologous tumor cell lines (TCLs), and tumor-infiltrating lymphocytes (TILs, germline reference). Neopeptides were predicted using MuPeXI, and patient-specific peptide–MHC (pMHC) libraries were created for all neopeptides with a rank score < 2 for binding to the patient's HLAs. T cell recognition toward neoepitopes in TILs was evaluated using the high-throughput technology of DNA barcode-labeled pMHC multimers. The patient-specific libraries consisted of, on average, 258 putative neopeptides (range, 103–397, n = 6). In four patients, WES was performed on two different sources (TF and TCL), whereas in two patients, WES was performed only on TF. Most of the peptides were predicted from both sources. However, a fraction was predicted from one source only. Among the total predicted neopeptides, 16% were derived from frameshift indels. T cell recognition of 52 neoepitopes was detected across all patients (range, 4–18, n = 6) and spanning two to five HLA restrictions per patient. On average, 21% of the recognized neoepitopes were derived from frameshift indels (range, 0–43%, n = 6). Thus, frameshift indels are equally represented in the pool of immunogenic neoepitopes as SNV-derived neoepitopes. This suggests the importance of a broad neopeptide prediction strategy covering multiple sources of tumor material, and including different genetic alterations. This study, for the first time, describes the T cell recognition of frameshift-derived neoepitopes in RCC and determines their immunogenic profile.

[1]  Cheryl F. Lichti,et al.  MHC-II neoantigens shape tumor immunity and response to immunotherapy , 2019, Nature.

[2]  R. Motzer,et al.  Avelumab plus Axitinib versus Sunitinib for Advanced Renal‐Cell Carcinoma , 2019, The New England journal of medicine.

[3]  J. Borst,et al.  CD4+ T cell help in cancer immunology and immunotherapy , 2018, Nature Reviews Immunology.

[4]  B. Seliger,et al.  T-cell Responses in the Microenvironment of Primary Renal Cell Carcinoma—Implications for Adoptive Cell Therapy , 2018, Cancer Immunology Research.

[5]  S. Hadrup,et al.  Novel tools to assist neoepitope targeting in personalized cancer immunotherapy. , 2017, Annals of oncology : official journal of the European Society for Medical Oncology.

[6]  M. Ringnér,et al.  Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma , 2017, Nature Communications.

[7]  Z. Szallasi,et al.  An Analysis of Natural T Cell Responses to Predicted Tumor Neoepitopes , 2017, Front. Immunol..

[8]  M. Nielsen,et al.  NetMHCpan-4.0: Improved Peptide–MHC Class I Interaction Predictions Integrating Eluted Ligand and Peptide Binding Affinity Data , 2017, The Journal of Immunology.

[9]  Nicolai J. Birkbak,et al.  Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. , 2017, The Lancet. Oncology.

[10]  J. Utikal,et al.  Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer , 2017, Nature.

[11]  Charles H. Yoon,et al.  An immunogenic personal neoantigen vaccine for patients with melanoma , 2017, Nature.

[12]  Zoltan Szallasi,et al.  MuPeXI: prediction of neo-epitopes from tumor sequencing data , 2017, Cancer Immunology, Immunotherapy.

[13]  E. Jaffee,et al.  Targeting neoantigens to augment antitumour immunity , 2017, Nature Reviews Cancer.

[14]  N. McGranahan,et al.  Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future , 2017, Cell.

[15]  Andrew P. Weng,et al.  Defining the clonality of peripheral T cell lymphomas using RNA-seq , 2016, Bioinform..

[16]  Ludmila V. Danilova,et al.  Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures , 2016, Genome Biology.

[17]  Ludmila V. Danilova,et al.  Erratum to: Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures , 2016, Genome Biology.

[18]  Suzanne F. Jones,et al.  Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma , 2016, Nature Communications.

[19]  Z. Szallasi,et al.  Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes , 2016, Nature Biotechnology.

[20]  Ton N. Schumacher,et al.  Targeting of cancer neoantigens with donor-derived T cell receptor repertoires , 2016, Science.

[21]  J. Galon,et al.  Frameshift mutations, neoantigens and tumor-specific CD8+ T cells in microsatellite unstable colorectal cancers , 2016, Oncoimmunology.

[22]  Lior Pachter,et al.  Near-optimal probabilistic RNA-seq quantification , 2016, Nature Biotechnology.

[23]  Nicolai J. Birkbak,et al.  Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade , 2016, Science.

[24]  H. Kume,et al.  Neoantigen Load, Antigen Presentation Machinery, and Immune Signatures Determine Prognosis in Clear Cell Renal Cell Carcinoma , 2016, Cancer Immunology Research.

[25]  J. Gartner,et al.  Isolation of neoantigen-specific T cells from tumor and peripheral lymphocytes. , 2015, The Journal of clinical investigation.

[26]  Mikhail Shugay,et al.  MiXCR: software for comprehensive adaptive immunity profiling , 2015, Nature Methods.

[27]  J. Castle,et al.  Mutant MHC class II epitopes drive therapeutic immune responses to cancer , 2015, Nature.

[28]  T. Schumacher,et al.  Neoantigens in cancer immunotherapy , 2015, Science.

[29]  Martin L. Miller,et al.  Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer , 2015, Science.

[30]  Michael R Stratton,et al.  High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma , 2014, Nature Medicine.

[31]  J. Wolchok,et al.  Genetic basis for clinical response to CTLA-4 blockade in melanoma. , 2014, The New England journal of medicine.

[32]  Benjamin Schubert,et al.  OptiType: precision HLA typing from next-generation sequencing data , 2014, Bioinform..

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

[34]  Mauricio O. Carneiro,et al.  From FastQ Data to High‐Confidence Variant Calls: The Genome Analysis Toolkit Best Practices Pipeline , 2013, Current protocols in bioinformatics.

[35]  David T. W. Jones,et al.  Signatures of mutational processes in human cancer , 2013, Nature.

[36]  A. Sivachenko,et al.  Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples , 2013, Nature Biotechnology.

[37]  Knut Reinert,et al.  RazerS 3: Faster, fully sensitive read mapping , 2012, Bioinform..

[38]  Hau-San Wong,et al.  Towards a Mathematical Foundation of Immunology and Amino Acid Chains , 2012, ArXiv.

[39]  Ton N Schumacher,et al.  Parallel detection of antigen-specific T cell responses by combinatorial encoding of MHC multimers , 2012, Nature Protocols.

[40]  P. A. Futreal,et al.  Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. , 2012, The New England journal of medicine.

[41]  M. Donia,et al.  Characterization and Comparison of ‘Standard’ and ‘Young’ Tumour‐Infiltrating Lymphocytes for Adoptive Cell Therapy at a Danish Translational Research Institution , 2012, Scandinavian journal of immunology.

[42]  Marcel Martin Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .

[43]  P. Royston,et al.  Interferon alfa-2a versus combination therapy with interferon alfa-2a, interleukin-2, and fluorouracil in patients with untreated metastatic renal cell carcinoma (MRC RE04/EORTC GU 30012): an open-label randomised trial , 2010 .

[44]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

[45]  T. Schumacher,et al.  Conditional MHC class I ligands and peptide exchange technology for the human MHC gene products HLA-A1, -A3, -A11, and -B7 , 2008, Proceedings of the National Academy of Sciences.

[46]  O. Lund,et al.  NetMHCpan, a Method for Quantitative Predictions of Peptide Binding to Any HLA-A and -B Locus Protein of Known Sequence , 2007, PloS one.

[47]  J. Yang,et al.  Immunotherapy for renal cell cancer. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[48]  T. Schumacher,et al.  Generation of peptide–MHC class I complexes through UV-mediated ligand exchange , 2006, Nature Protocols.

[49]  T. Schumacher,et al.  Design and use of conditional MHC class I ligands , 2006, Nature Medicine.

[50]  S. Rosenberg,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 , 2004, The Journal of Immunology.

[51]  M. Shlomchik,et al.  A Shannon entropy analysis of immunoglobulin and T cell receptor. , 1997, Molecular immunology.

[52]  C. E. SHANNON,et al.  A mathematical theory of communication , 1948, MOCO.

[53]  Ton N Schumacher,et al.  Cancer Neoantigens. , 2019, Annual review of immunology.

[54]  D. Egan,et al.  High-throughput T-cell epitope discovery through MHC peptide exchange. , 2009, Methods in molecular biology.

[55]  D. Holdstock Past, present--and future? , 2005, Medicine, conflict, and survival.

[56]  K. Shadan,et al.  Available online: , 2012 .