CD8+ T Cells Specific for Tumor Antigens Can Be Rendered Dysfunctional by the Tumor Microenvironment through Upregulation of the Inhibitory Receptors BTLA …

J Fourcade, Z Sun, O Pagliano, P Guillaume… - Cancer research, 2012 - AACR
J Fourcade, Z Sun, O Pagliano, P Guillaume, IF Luescher, C Sander, JM Kirkwood, D Olive
Cancer research, 2012AACR
Cytotoxic T cells that are present in tumors and capable of recognizing tumor epitopes are
nevertheless generally impotent in eliciting tumor rejection. Thus, identifying the immune
escape mechanisms responsible for inducing tumor-specific CD8+ T-cell dysfunction may
reveal effective strategies for immune therapy. The inhibitory receptors PD-1 and Tim-3 are
known to negatively regulate CD8+ T-cell responses directed against the well-characterized
tumor antigen NY-ESO-1. Here, we report that the upregulation of the inhibitory molecule …
Abstract
Cytotoxic T cells that are present in tumors and capable of recognizing tumor epitopes are nevertheless generally impotent in eliciting tumor rejection. Thus, identifying the immune escape mechanisms responsible for inducing tumor-specific CD8+ T-cell dysfunction may reveal effective strategies for immune therapy. The inhibitory receptors PD-1 and Tim-3 are known to negatively regulate CD8+ T-cell responses directed against the well-characterized tumor antigen NY-ESO-1. Here, we report that the upregulation of the inhibitory molecule BTLA also plays a critical role in restricting NY-ESO-1–specific CD8+ T-cell expansion and function in melanoma. BTLA-expressing PD-1+Tim-3 CD8+ T cells represented the largest subset of NY-ESO-1–specific CD8+ T cells in patients with melanoma. These cells were partially dysfunctional, producing less IFN-γ than BTLA T cells but more IFN-γ, TNF, and interleukin-2 than the highly dysfunctional subset expressing all three receptors. Expression of BTLA did not increase with higher T-cell dysfunction or upon cognate antigen stimulation, as it does with PD-1, suggesting that BTLA upregulation occurs independently of functional exhaustion driven by high antigen load. Added with PD-1 and Tim-3 blockades, BTLA blockade enhanced the expansion, proliferation, and cytokine production of NY-ESO-1–specific CD8+ T cells. Collectively, our findings indicate that targeting BTLA along with the PD-1 and Tim-3 pathways is critical to reverse an important mechanism of immune escape in patients with advanced melanoma. Cancer Res; 72(4); 887–96. ©2011 AACR.
AACR