Tumor Frameshift Mutation Proportion Predicts Response to Immunotherapy in Mismatch Repair‐Deficient Prostate Cancer

Abstract Background Genomic biomarkers that predict response to anti‐PD1 therapy in prostate cancer are needed. Frameshift mutations are predicted to generate more neoantigens than missense mutations; therefore, we hypothesized that the number or proportion of tumor frameshift mutations would correlate with response to anti‐PD1 therapy in prostate cancer. Methods To enrich for response to anti‐PD1 therapy, we assembled a multicenter cohort of 65 men with mismatch repair‐deficient (dMMR) prostate cancer. Patient characteristics and outcomes were determined by retrospective chart review. Clinical somatic DNA sequencing was used to determine tumor mutational burden (TMB), frameshift mutation burden, and frameshift mutation proportion (FSP), which were correlated to outcomes on anti‐PD1 treatment. We subsequently used data from a clinical trial of pembrolizumab in patients with nonprostatic dMMR cancers of various histologies as a biomarker validation cohort. Results Nineteen of 65 patients with dMMR metastatic castration‐resistant prostate cancer were treated with anti‐PD1 therapy. The PSA50 response rate was 65%, and the median progression‐free survival (PFS) was 24 (95% confidence interval 16–54) weeks. Tumor FSP, more than overall TMB, correlated most strongly with prolonged PFS and overall survival (OS) on anti‐PD1 treatment and with density of CD8+ tumor‐infiltrating lymphocytes. High FSP similarly identified patients with longer PFS as well as OS on anti‐PD1 therapy in a validation cohort. Conclusion Tumor FSP correlated with prolonged efficacy of anti‐PD1 treatment among patients with dMMR cancers and may represent a new biomarker of immune checkpoint inhibitor sensitivity. Implications for Practice Given the modest efficacy of immune checkpoint inhibition (ICI) in unselected patients with advanced prostate cancer, biomarkers of ICI sensitivity are needed. To facilitate biomarker discovery, a cohort of patients with DNA mismatch repair‐deficient (dMMR) prostate cancer was assembled, as these patients are enriched for responses to ICI. A high response rate to anti‐PD1 therapy in these patients was observed; however, these responses were not durable in most patients. Notably, tumor frameshift mutation proportion (FSP) was identified as a novel biomarker that was associated with prolonged response to anti‐PD1 therapy in this cohort. This finding was validated in a separate cohort of patients with nonprostatic dMMR cancers of various primary histologies. This works suggests that FSP predicts response to anti‐PD1 therapy in dMMR cancers, which should be validated prospectively in larger independent cohorts.

[1]  N. Agarwal,et al.  Clinical activity of pembrolizumab in metastatic prostate cancer with microsatellite instability high (MSI-H) detected by circulating tumor DNA , 2020, Journal for ImmunoTherapy of Cancer.

[2]  P. Nelson,et al.  Mismatch repair deficiency in metastatic prostate cancer: Response to PD-1 blockade and standard therapies , 2020, PloS one.

[3]  A. Partin,et al.  T-Cell Infiltration and Adaptive Treg Resistance in Response to Androgen Deprivation With or Without Vaccination in Localized Prostate Cancer , 2020, Clinical Cancer Research.

[4]  C. Drake,et al.  Pembrolizumab for Treatment-Refractory Metastatic Castration-Resistant Prostate Cancer: Multicohort, Open-Label Phase II KEYNOTE-199 Study. , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[5]  A. Wyatt,et al.  Identification of Hypermutation and Defective Mismatch Repair in ctDNA from Metastatic Prostate Cancer , 2019, Clinical Cancer Research.

[6]  Ahmet Zehir,et al.  Genetic diversity of tumors with mismatch repair deficiency influences anti–PD-1 immunotherapy response , 2019, Science.

[7]  Kaanan P. Shah,et al.  Clinical validation of the tempus xT next-generation targeted oncology sequencing assay , 2019, Oncotarget.

[8]  E. V. Van Allen,et al.  Genomic correlates of response to immune checkpoint blockade , 2019, Nature Medicine.

[9]  E. Antonarakis,et al.  Clinical Features and Therapeutic Outcomes in Men with Advanced Prostate Cancer and DNA Mismatch Repair Gene Mutations. , 2019, European urology.

[10]  Q. Dai,et al.  Genome-Wide CRISPR Screening Identifies JAK1 Deficiency as a Mechanism of T-Cell Resistance , 2019, Front. Immunol..

[11]  N. Chaput,et al.  Predictive biomarkers of response for immune checkpoint inhibitors in non-small-cell lung cancer. , 2019, European journal of cancer.

[12]  P. Kantoff,et al.  Analysis of the Prevalence of Microsatellite Instability in Prostate Cancer and Response to Immune Checkpoint Blockade , 2019, JAMA oncology.

[13]  M. Rubin,et al.  Immunogenomic analyses associate immunological alterations with mismatch repair defects in prostate cancer , 2018, The Journal of clinical investigation.

[14]  V. Velculescu,et al.  Ipilimumab plus nivolumab and DNA-repair defects in AR-V7-expressing metastatic prostate cancer , 2018, Oncotarget.

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

[16]  J. Eshleman,et al.  MSH2 Loss in Primary Prostate Cancer , 2017, Clinical Cancer Research.

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

[18]  Ludmila V. Danilova,et al.  Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade , 2017, Science.

[19]  Levi Garraway,et al.  Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden , 2017, Genome Medicine.

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

[21]  R. DePinho,et al.  Effective Combinatorial Immunotherapy for Castration Resistant Prostate Cancer , 2017, Nature.

[22]  F. Saad,et al.  Randomized, Double-Blind, Phase III Trial of Ipilimumab Versus Placebo in Asymptomatic or Minimally Symptomatic Patients With Metastatic Chemotherapy-Naive Castration-Resistant Prostate Cancer. , 2017, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[23]  R. Bourgon,et al.  Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial , 2016, The Lancet.

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

[25]  M. Scott Lucia,et al.  Paucity of PD-L1 Expression in Prostate Cancer: Innate and Adaptive Immune Resistance , 2015, Prostate Cancer and Prostatic Disease.

[26]  David C. Smith,et al.  Integrative Clinical Genomics of Advanced Prostate Cancer , 2015, Cell.

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

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

[29]  Ming Yu,et al.  Complex MSH2 and MSH6 mutations in hypermutated microsatellite unstable advanced prostate cancer , 2014, Nature Communications.

[30]  N. Agarwal,et al.  Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. , 2014, The Lancet. Oncology.

[31]  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.

[32]  K. Kinzler,et al.  Epitope landscape in breast and colorectal cancer. , 2008, Cancer research.

[33]  R. Paules,et al.  DNA protein kinase-dependent G2 checkpoint revealed following knockdown of ataxia-telangiectasia mutated in human mammary epithelial cells. , 2008, Cancer research.

[34]  Guo-Min Li,et al.  Mechanisms and functions of DNA mismatch repair , 2008, Cell Research.

[35]  Yan P. Yuan,et al.  Frameshift peptide‐derived T‐cell epitopes: A source of novel tumor‐specific antigens , 2001, International journal of cancer.