T-cell exhaustion signatures vary with tumor type and are severe in glioblastoma

K Woroniecka, P Chongsathidkiet, K Rhodin… - Clinical Cancer …, 2018 - AACR
K Woroniecka, P Chongsathidkiet, K Rhodin, H Kemeny, C Dechant, SH Farber…
Clinical Cancer Research, 2018AACR
Purpose: T-cell dysfunction is a hallmark of glioblastoma (GBM). Although anergy and
tolerance have been well characterized, T-cell exhaustion remains relatively unexplored.
Exhaustion, characterized in part by the upregulation of multiple immune checkpoints, is a
known contributor to failures amid immune checkpoint blockade, a strategy that has lacked
success thus far in GBM. This study is among the first to examine, and credential as bona
fide, exhaustion among T cells infiltrating human and murine GBM. Experimental Design …
Abstract
Purpose: T-cell dysfunction is a hallmark of glioblastoma (GBM). Although anergy and tolerance have been well characterized, T-cell exhaustion remains relatively unexplored. Exhaustion, characterized in part by the upregulation of multiple immune checkpoints, is a known contributor to failures amid immune checkpoint blockade, a strategy that has lacked success thus far in GBM. This study is among the first to examine, and credential as bona fide, exhaustion among T cells infiltrating human and murine GBM.
Experimental Design: Tumor-infiltrating and peripheral blood lymphocytes (TILs and PBLs) were isolated from patients with GBM. Levels of exhaustion-associated inhibitory receptors and poststimulation levels of the cytokines IFNγ, TNFα, and IL2 were assessed by flow cytometry. T-cell receptor Vβ chain expansion was also assessed in TILs and PBLs. Similar analysis was extended to TILs isolated from intracranial and subcutaneous immunocompetent murine models of glioma, breast, lung, and melanoma cancers.
Results: Our data reveal that GBM elicits a particularly severe T-cell exhaustion signature among infiltrating T cells characterized by: (1) prominent upregulation of multiple immune checkpoints; (2) stereotyped T-cell transcriptional programs matching classical virus-induced exhaustion; and (3) notable T-cell hyporesponsiveness in tumor-specific T cells. Exhaustion signatures differ predictably with tumor identity, but remain stable across manipulated tumor locations.
Conclusions: Distinct cancers possess similarly distinct mechanisms for exhausting T cells. The poor TIL function and severe exhaustion observed in GBM highlight the need to better understand this tumor-imposed mode of T-cell dysfunction in order to formulate effective immunotherapeutic strategies targeting GBM. Clin Cancer Res; 24(17); 4175–86. ©2018 AACR.
See related commentary by Jackson and Lim, p. 4059
AACR