Blockade of programmed death 1 augments the ability of human T cells engineered to target NY-ESO-1 to control tumor growth after adoptive transfer

EK Moon, R Ranganathan, E Eruslanov, S Kim… - Clinical Cancer …, 2016 - AACR
EK Moon, R Ranganathan, E Eruslanov, S Kim, K Newick, S O'Brien, A Lo, X Liu, Y Zhao
Clinical Cancer Research, 2016AACR
Abstract Purpose: Tumor-infiltrating lymphocytes (TILs) become hypofunctional, although the
mechanisms are not clear. Our goal was to generate a model of human tumor-induced TIL
hypofunction to study mechanisms and to test anti-human therapeutics. Experimental
Design: We transduced human T cells with a published, optimized T-cell receptor (TCR) that
is directed to a peptide within the cancer testis antigen, NY-ESO-1. After demonstrating
antigen-specific in vitro activity, these cells were used to target a human lung cancer line that …
Abstract
Purpose: Tumor-infiltrating lymphocytes (TILs) become hypofunctional, although the mechanisms are not clear. Our goal was to generate a model of human tumor-induced TIL hypofunction to study mechanisms and to test anti-human therapeutics.
Experimental Design: We transduced human T cells with a published, optimized T-cell receptor (TCR) that is directed to a peptide within the cancer testis antigen, NY-ESO-1. After demonstrating antigen-specific in vitro activity, these cells were used to target a human lung cancer line that expressed NY-ESO-1 in the appropriate HLA context growing in immunodeficient mice. The ability of anti-PD1 antibody to augment efficacy was tested.
Results: Injection of transgenic T cells had some antitumor activity, but did not eliminate the tumors. The injected T cells became profoundly hypofunctional accompanied by upregulation of PD1, Tim3, and Lag3 with coexpression of multiple inhibitory receptors in a high percentage of cells. This model allowed us to test reagents targeted specifically to human T cells. We found that injections of an anti-PD1 antibody in combination with T cells led to decreased TIL hypofunction and augmented the efficacy of the adoptively transferred T cells.
Conclusions: This model offers a platform for preclinical testing of adjuvant immunotherapeutics targeted to human T cells prior to transition to the bedside. Because the model employs engineering of human T cells with a TCR clone instead of a CAR, it allows for study of the biology of tumor-reactive TILs that signal through an endogenous TCR. The lessons learned from TCR-engineered TILs can thus be applied to tumor-reactive TILs. Clin Cancer Res; 22(2); 436–47. ©2015 AACR.
See related commentary by Yang, p. 275
AACR