Tumor-infiltrating BRAFV600E-specific CD4+ T cells correlated with complete clinical response in melanoma

T cells specific for neoantigens encoded by mutated genes in cancers are increasingly recognized as mediators of tumor destruction after immune checkpoint inhibitor therapy or adoptive cell transfer. Unfortunately, most neoantigens result from random mutations and are patient specific, and some canc...

Full description

Saved in:
Bibliographic Details
Published inThe Journal of clinical investigation Vol. 128; no. 4; pp. 1563 - 1568
Main Authors Veatch, Joshua R, Lee, Sylvia M, Fitzgibbon, Matthew, Chow, I-Ting, Jesernig, Brenda, Schmitt, Tom, Kong, Ying Ying, Kargl, Julia, Houghton, A McGarry, Thompson, John A, McIntosh, Martin, Kwok, William W, Riddell, Stanley R
Format Journal Article
LanguageEnglish
Published United States American Society for Clinical Investigation 02.04.2018
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:T cells specific for neoantigens encoded by mutated genes in cancers are increasingly recognized as mediators of tumor destruction after immune checkpoint inhibitor therapy or adoptive cell transfer. Unfortunately, most neoantigens result from random mutations and are patient specific, and some cancers contain few mutations to serve as potential antigens. We describe a patient with stage IV acral melanoma who achieved a complete response following adoptive transfer of tumor-infiltrating lymphocytes (TILs). Tumor exome sequencing surprisingly revealed fewer than 30 nonsynonymous somatic mutations, including oncogenic BRAFV600E. Analysis of the specificity of TILs identified rare CD4+ T cells specific for BRAFV600E and diverse CD8+ T cells reactive to nonmutated self-antigens. These specificities increased in blood after TIL transfer and persisted long-term, suggesting they contributed to the effective antitumor immune response. Gene transfer of the BRAFV600E-specific T cell receptor (TCR) conferred recognition of class II MHC-positive cells expressing the BRAF mutation. Therapy with TCR-engineered BRAFV600E-specific CD4+ T cells may have direct antitumor effects and augment CD8+ T cell responses to self- and/or mutated tumor antigens in patients with BRAF-mutated cancers.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:0021-9738
1558-8238
DOI:10.1172/JCI98689