Low Mutation Burden in Ovarian Cancer May Limit the Utility of Neoantigen-Targeted Vaccines

Due to advances in sequencing technology, somatically mutated cancer antigens, or neoantigens, are now readily identifiable and have become compelling targets for immunotherapy. In particular, neoantigen-targeted vaccines have shown promise in several pre-clinical and clinical studies. However, to date, neoantigen-targeted vaccine studies have involved tumors with exceptionally high mutation burdens. It remains unclear whether neoantigen-targeted vaccines will be broadly applicable to cancers with intermediate to low mutation burdens, such as ovarian cancer. To address this, we assessed whether a derivative of the murine ovarian tumor model ID8 could be targeted with neoantigen vaccines. We performed whole exome and transcriptome sequencing on ID8-G7 cells. We identified 92 somatic mutations, 39 of which were transcribed, missense mutations. For the 17 top predicted MHC class I binding mutations, we immunized mice subcutaneously with synthetic long peptide vaccines encoding the relevant mutation. Seven of 17 vaccines induced robust mutation-specific CD4 and/or CD8 T cell responses. However, none of the vaccines prolonged survival of tumor-bearing mice in either the prophylactic or therapeutic setting. Moreover, none of the neoantigen-specific T cell lines recognized ID8-G7 tumor cells in vitro, indicating that the corresponding mutations did not give rise to bonafide MHC-presented epitopes. Additionally, bioinformatic analysis of The Cancer Genome Atlas data revealed that only 12% (26/220) of HGSC cases had a ≥90% likelihood of harboring at least one authentic, naturally processed and presented neoantigen versus 51% (80/158) of lung cancers. Our findings highlight the limitations of applying neoantigen-targeted vaccines to tumor types with intermediate/low mutation burdens.

[1]  S. Begum,et al.  Sequence Alignment , 2018, Beginners Guide to Bioinformatics for High Throughput Sequencing.

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

[3]  Bhartendu Nath Mishra,et al.  Major histocompatibility complex linked databases and prediction tools for designing vaccines. , 2016, Human immunology.

[4]  Lauren L. Ritterhouse,et al.  Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer , 2016, Oncotarget.

[5]  Yong-Chen Lu,et al.  Cancer immunotherapy targeting neoantigens. , 2016, Seminars in immunology.

[6]  J. Gartner,et al.  Immunogenicity of somatic mutations in human gastrointestinal cancers , 2015, Science.

[7]  Lauren L. Ritterhouse,et al.  Association of Polymerase e-Mutated and Microsatellite-Instable Endometrial Cancers With Neoantigen Load, Number of Tumor-Infiltrating Lymphocytes, and Expression of PD-1 and PD-L1. , 2015, JAMA oncology.

[8]  R. Holt,et al.  Targeting the undruggable: immunotherapy meets personalized oncology in the genomic era. , 2015, Annals of oncology : official journal of the European Society for Medical Oncology.

[9]  O. Kohlbacher,et al.  Immunoinformatics and epitope prediction in the age of genomic medicine , 2015, Genome Medicine.

[10]  V. Beral,et al.  Rethinking ovarian cancer II: reducing mortality from high-grade serous ovarian cancer , 2015, Nature Reviews Cancer.

[11]  S. Gabriel,et al.  Genomic correlates of response to CTLA-4 blockade in metastatic melanoma , 2015, Science.

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

[13]  M. Marra,et al.  Next-Generation Sequencing Approaches in Cancer: Where Have They Brought Us and Where Will They Take Us? , 2015, Cancers.

[14]  Razelle Kurzrock,et al.  Breast Cancer Experience of the Molecular Tumor Board at the University of California, San Diego Moores Cancer Center. , 2015, Journal of oncology practice.

[15]  Joshy George,et al.  Whole–genome characterization of chemoresistant ovarian cancer , 2015, Nature.

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

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

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

[19]  T. Blankenstein,et al.  Identification of human T-cell receptors with optimal affinity to cancer antigens using antigen-negative humanized mice , 2015, Nature Biotechnology.

[20]  N. Hacohen,et al.  Molecular and Genetic Properties of Tumors Associated with Local Immune Cytolytic Activity , 2015, Cell.

[21]  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.

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

[23]  Z. Modrušan,et al.  Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing , 2014, Nature.

[24]  Maxim N. Artyomov,et al.  Checkpoint Blockade Cancer Immunotherapy Targets Tumour-Specific Mutant Antigens , 2014, Nature.

[25]  J. Sidney,et al.  Genomic and bioinformatic profiling of mutational neoepitopes reveals new rules to predict anticancer immunogenicity , 2014, The Journal of experimental medicine.

[26]  J. Tanyi,et al.  Immunotherapy for ovarian cancer: recent advances and perspectives , 2014, Current opinion in oncology.

[27]  K. Cibulskis,et al.  Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia. , 2014, Blood.

[28]  S. Lippman,et al.  Molecular tumor board: the University of California-San Diego Moores Cancer Center experience. , 2014, The oncologist.

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

[30]  Scott D. Brown,et al.  Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival , 2014, Genome research.

[31]  S. Rosenberg,et al.  Efficient Identification of Mutated Cancer Antigens Recognized by T Cells Associated with Durable Tumor Regressions , 2014, Clinical Cancer Research.

[32]  T. Schumacher,et al.  High sensitivity of cancer exome-based CD8 T cell neo-antigen identification , 2014, Oncoimmunology.

[33]  N. Hacohen,et al.  HLA-Binding Properties of Tumor Neoepitopes in Humans , 2014, Cancer Immunology Research.

[34]  S. Gabriel,et al.  Discovery and saturation analysis of cancer genes across 21 tumor types , 2014, Nature.

[35]  S. Steinberg,et al.  The immunological and clinical effects of mutated ras peptide vaccine in combination with IL-2, GM-CSF, or both in patients with solid tumors , 2014, Journal of Translational Medicine.

[36]  Bjoern Peters,et al.  HLA Class I Alleles Are Associated with Peptide-Binding Repertoires of Different Size, Affinity, and Immunogenicity , 2013, The Journal of Immunology.

[37]  R. Holt,et al.  Surveillance of the Tumor Mutanome by T Cells during Progression from Primary to Recurrent Ovarian Cancer , 2013, Clinical Cancer Research.

[38]  M. Stratton,et al.  Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[39]  B. Clarke,et al.  Identifying Lynch Syndrome in Patients With Ovarian Carcinoma: The Significance of Tumor Subtype , 2013, Advances in anatomic pathology.

[40]  Benjamin J. Raphael,et al.  Mutational landscape and significance across 12 major cancer types , 2013, Nature.

[41]  Chris Sander,et al.  Emerging landscape of oncogenic signatures across human cancers , 2013, Nature Genetics.

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

[43]  G. Freeman,et al.  Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. , 2013, Cancer research.

[44]  Jimmy Lin,et al.  Mining Exomic Sequencing Data to Identify Mutated Antigens Recognized by Adoptively Transferred Tumor-reactive T cells , 2013, Nature Medicine.

[45]  T. Blankenstein,et al.  It's the peptide-MHC affinity, stupid. , 2013, Cancer cell.

[46]  John Sidney,et al.  Relapse or eradication of cancer is predicted by peptide-major histocompatibility complex affinity. , 2013, Cancer cell.

[47]  C. Peterson,et al.  Characterization and evaluation of pre-clinical suitability of a syngeneic orthotopic mouse ovarian cancer model. , 2013, Anticancer research.

[48]  K. Kinzler,et al.  Cancer Genome Landscapes , 2013, Science.

[49]  Pia Kvistborg,et al.  The cancer antigenome , 2012, The EMBO journal.

[50]  Steven A. Roberts,et al.  Mutational heterogeneity in cancer and the search for new cancer-associated genes , 2013 .

[51]  C. Morrison,et al.  Efficacy of vaccination with recombinant vaccinia and fowlpox vectors expressing NY-ESO-1 antigen in ovarian cancer and melanoma patients , 2012, Proceedings of the National Academy of Sciences.

[52]  J. Castle,et al.  Exploiting the mutanome for tumor vaccination. , 2012, Cancer research.

[53]  E. Mardis,et al.  Cancer Exome Analysis Reveals a T Cell Dependent Mechanism of Cancer Immunoediting , 2012, Nature.

[54]  S. Steinberg,et al.  A gynecologic oncology group phase II trial of two p53 peptide vaccine approaches: subcutaneous injection and intravenous pulsed dendritic cells in high recurrence risk ovarian cancer patients , 2012, Cancer Immunology, Immunotherapy.

[55]  E. Goode,et al.  Tumor-Infiltrating Programmed Death Receptor-1+ Dendritic Cells Mediate Immune Suppression in Ovarian Cancer , 2011, The Journal of Immunology.

[56]  B. Nelson,et al.  Profound CD8+ T cell immunity elicited by sequential daily immunization with exogenous antigen plus the TLR3 agonist poly(I:C). , 2011, Vaccine.

[57]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer , 2011, Nature Biotechnology.

[58]  H. Hakonarson,et al.  ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data , 2010, Nucleic acids research.

[59]  J. Sakamoto,et al.  Wilms' tumor 1 (WT1) peptide immunotherapy for gynecological malignancy. , 2009, Anticancer research.

[60]  Sylvia Janetzki,et al.  "MIATA"-minimal information about T cell assays. , 2009, Immunity.

[61]  D. Scheinberg,et al.  Synthetic tumor‐specific breakpoint peptide vaccine in patients with chronic myeloid leukemia and minimal residual disease , 2009, Cancer.

[62]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

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

[64]  N. Shastri,et al.  SPAS-1 (stimulator of prostatic adenocarcinoma-specific T cells)/SH3GLB2: A prostate tumor antigen identified by CTLA-4 blockade , 2008, Proceedings of the National Academy of Sciences.

[65]  Uthaman Gowthaman,et al.  In silico tools for predicting peptides binding to HLA-class II molecules: more confusion than conclusion. , 2008, Journal of proteome research.

[66]  O. Lund,et al.  NetMHCpan, a method for MHC class I binding prediction beyond humans , 2008, Immunogenetics.

[67]  P. Watson,et al.  CD8+ T Cells Induce Complete Regression of Advanced Ovarian Cancers by an Interleukin (IL)-2/IL-15–Dependent Mechanism , 2007, Clinical Cancer Research.

[68]  S. Ogawa,et al.  Alternative splicing due to an intronic SNP in HMSD generates a novel minor histocompatibility antigen. , 2007, Blood.

[69]  P. Watson,et al.  Spontaneous mammary tumors differ widely in their inherent sensitivity to adoptively transferred T cells. , 2007, Cancer research.

[70]  Bjoern Peters,et al.  A Quantitative Analysis of the Variables Affecting the Repertoire of T Cell Specificities Recognized after Vaccinia Virus Infection1 , 2007, The Journal of Immunology.

[71]  Gang Wang,et al.  A Phase I Study on Adoptive Immunotherapy Using Gene-Modified T Cells for Ovarian Cancer , 2006, Clinical Cancer Research.

[72]  Mitchell Ho,et al.  Identification of Novel Human CTL Epitopes and Their Agonist Epitopes of Mesothelin , 2005, Clinical Cancer Research.

[73]  J. Berzofsky,et al.  Immunization with mutant p53- and K-ras-derived peptides in cancer patients: immune response and clinical outcome. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[74]  R. Schreiber,et al.  The three Es of cancer immunoediting. , 2004, Annual review of immunology.

[75]  George Coukos,et al.  Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. , 2003, The New England journal of medicine.

[76]  A. Tomassetti,et al.  A step further in understanding the biology of the folate receptor in ovarian carcinoma. , 2003, Gynecologic oncology.

[77]  J L Pace,et al.  Development of a syngeneic mouse model for events related to ovarian cancer. , 2000, Carcinogenesis.

[78]  C. Farina,et al.  Translation of a Retained Intron in Tyrosinase-related Protein (TRP) 2 mRNA Generates a New Cytotoxic T Lymphocyte (CTL)-defined and Shared Human Melanoma Antigen Not Expressed in Normal Cells of the Melanocytic Lineage , 1998, The Journal of experimental medicine.

[79]  F. Lemonnier,et al.  Cytotoxic T cell response against the chimeric p210 BCR-ABL protein in patients with chronic myelogenous leukemia. , 1998, The Journal of clinical investigation.

[80]  Kristin A. Hogquist,et al.  T cell receptor antagonist peptides induce positive selection , 1994, Cell.