KOR

e-Article

IRIS: Discovery of cancer immunotherapy targets arising from pre-mRNA alternative splicing
Document Type
article
Source
Proceedings of the National Academy of Sciences of the United States of America. 120(21)
Subject
Biomedical and Clinical Sciences
Oncology and Carcinogenesis
Immunology
Cancer Genomics
Biotechnology
Immunotherapy
Immunization
Cancer
Vaccine Related
Human Genome
Genetics
5.1 Pharmaceuticals
Development of treatments and therapeutic interventions
Good Health and Well Being
Male
Humans
RNA Precursors
Alternative Splicing
Leukocytes
Mononuclear
Receptors
Antigen
T-Cell
Epitopes
T-Lymphocyte
Antigens
Neoplasm
Peptides
Neoplasms
RNA splicing
immunotherapy
T cell receptors
Language
Abstract
Alternative splicing (AS) is prevalent in cancer, generating an extensive but largely unexplored repertoire of novel immunotherapy targets. We describe Isoform peptides from RNA splicing for Immunotherapy target Screening (IRIS), a computational platform capable of discovering AS-derived tumor antigens (TAs) for T cell receptor (TCR) and chimeric antigen receptor T cell (CAR-T) therapies. IRIS leverages large-scale tumor and normal transcriptome data and incorporates multiple screening approaches to discover AS-derived TAs with tumor-associated or tumor-specific expression. In a proof-of-concept analysis integrating transcriptomics and immunopeptidomics data, we showed that hundreds of IRIS-predicted TCR targets are presented by human leukocyte antigen (HLA) molecules. We applied IRIS to RNA-seq data of neuroendocrine prostate cancer (NEPC). From 2,939 NEPC-associated AS events, IRIS predicted 1,651 epitopes from 808 events as potential TCR targets for two common HLA types (A*02:01 and A*03:01). A more stringent screening test prioritized 48 epitopes from 20 events with "neoantigen-like" NEPC-specific expression. Predicted epitopes are often encoded by microexons of ≤30 nucleotides. To validate the immunogenicity and T cell recognition of IRIS-predicted TCR epitopes, we performed in vitro T cell priming in combination with single-cell TCR sequencing. Seven TCRs transduced into human peripheral blood mononuclear cells (PBMCs) showed high activity against individual IRIS-predicted epitopes, providing strong evidence of isolated TCRs reactive to AS-derived peptides. One selected TCR showed efficient cytotoxicity against target cells expressing the target peptide. Our study illustrates the contribution of AS to the TA repertoire of cancer cells and demonstrates the utility of IRIS for discovering AS-derived TAs and expanding cancer immunotherapies.