Using computational tools, mutational signature information, as well as whole genome and RNA sequencing of bulk, high grade ovarian carcinoma pre- and post-chemotherapy tumor samples, O’Donnell et al. demonstrated that relapsed tumors have an increase in neoantigen load compared with primary tumors, but most of the increase is due to pre-existing mutational processes and not chemotherapy-related mutagenesis. Pre-chemotherapy neoantigen load and CD8+ T cell infiltration independently predicted survival outcomes.

BACKGROUND: Patients with highly mutated tumors, such as melanoma or smoking-related lung cancer, have higher rates of response to immune checkpoint blockade therapy, perhaps due to increased neoantigen expression. Many chemotherapies including platinum compounds are known to be mutagenic, but the impact of standard treatment protocols on mutational burden and resulting neoantigen expression in most human cancers is unknown. METHODS: We sought to quantify the effect of chemotherapy treatment on computationally predicted neoantigen expression for high grade serous ovarian carcinoma patients enrolled in the Australian Ovarian Cancer Study. In this series, 35 of 114 samples were collected after exposure to chemotherapy; 14 are matched with an untreated sample from the same patient. Our approach integrates whole genome and RNA sequencing of bulk tumor samples with class I MHC binding prediction and mutational signatures extracted from studies of chemotherapy-exposed Caenorhabditis elegans and Gallus gallus cells. We additionally investigated the relationship between neoantigens, tumor infiltrating immune cells estimated from RNA-seq with CIBERSORT, and patient survival. RESULTS: Greater neoantigen burden and CD8+ T cell infiltration in primary, pre-treatment samples were independently associated with improved survival. Relapse samples collected after chemotherapy harbored a median of 78% more expressed neoantigens than untreated primary samples, a figure that combines the effects of chemotherapy and other processes operative during relapse. The contribution from chemotherapy-associated signatures was small, accounting for a mean of 5% (range 0-16) of the expressed neoantigen burden in relapse samples. In both treated and untreated samples, most neoantigens were attributed to COSMIC Signature (3), associated with BRCA disruption, Signature (1), associated with a slow mutagenic process active in healthy tissue, and Signature (8), of unknown etiology. CONCLUSION: Relapsed ovarian cancers harbor more predicted neoantigens than primary tumors, but the increase is due to pre-existing mutational processes, not mutagenesis from chemotherapy.

Author Info: (1) Icahn School of Medicine at Mount Sinai, New York, NY, USA. (2) Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia. (3) Icahn School of Medicine at Mount

Author Info: (1) Icahn School of Medicine at Mount Sinai, New York, NY, USA. (2) Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia. (3) Icahn School of Medicine at Mount Sinai, New York, NY, USA. (4) Icahn School of Medicine at Mount Sinai, New York, NY, USA. (5) Icahn School of Medicine at Mount Sinai, New York, NY, USA. (6) Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia. (7) Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA. Adaptive Biotechnologies, Seattle, WA, USA. (8) Icahn School of Medicine at Mount Sinai, New York, NY, USA. correspondence@hammerlab.org. Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA. correspondence@hammerlab.org.