Toward Personalized Lymphoma Immunotherapy: Identification of Common Driver Mutations Recognized by Patient CD8+ T Cells

JS Nielsen, CG Sedgwick, A Shahid, Z Zong… - Clinical Cancer …, 2016 - AACR
JS Nielsen, CG Sedgwick, A Shahid, Z Zong, ZL Brumme, S Yu, L Liu, DR Kroeger
Clinical Cancer Research, 2016AACR
Purpose: A fundamental challenge in the era of next-generation sequencing (NGS) is to
design effective treatments tailored to the mutational profiles of tumors. Many newly
discovered cancer mutations are difficult to target pharmacologically; however, T-cell–based
therapies may provide a valuable alternative owing to the exquisite sensitivity and specificity
of antigen recognition. To explore this concept, we assessed the immunogenicity of a panel
of genes that are common sites of driver mutations in follicular lymphoma, an …
Abstract
Purpose: A fundamental challenge in the era of next-generation sequencing (NGS) is to design effective treatments tailored to the mutational profiles of tumors. Many newly discovered cancer mutations are difficult to target pharmacologically; however, T-cell–based therapies may provide a valuable alternative owing to the exquisite sensitivity and specificity of antigen recognition. To explore this concept, we assessed the immunogenicity of a panel of genes that are common sites of driver mutations in follicular lymphoma, an immunologically sensitive yet currently incurable disease.
Experimental Design: Exon capture and NGS were used to interrogate tumor samples from 53 patients with follicular lymphoma for mutations in 10 frequently mutated genes. For 13 patients, predicted mutant peptides and proteins were evaluated for recognition by autologous peripheral blood T cells after in vitro priming.
Results: Mutations were identified in 1–5 genes in 81% (43/53) of tumor samples. Autologous, mutation-specific CD8+ T cells were identified in 23% (3/13) of evaluated cases. T-cell responses were directed toward putative driver mutations in CREBBP and MEF2B. Responding T cells showed exquisite specificity for mutant versus wild-type proteins and recognized lymphoma cells expressing the appropriate mutations. Responding T cells appeared to be from the naïve repertoire, as they were found at low frequencies and only at single time points in each patient.
Conclusions: Patients with follicular lymphoma harbor rare yet functionally competent CD8+ T cells specific for recurrent mutations. Our results support the concept of using NGS to design individualized immunotherapies targeting common driver mutations in follicular lymphoma and other malignancies. Clin Cancer Res; 22(9); 2226–36. ©2015 AACR.
AACR