Glioblastoma eradication following immune checkpoint blockade in an orthotopic, immunocompetent model

DA Reardon, PC Gokhale, SR Klein, KL Ligon… - Cancer immunology …, 2016 - AACR
DA Reardon, PC Gokhale, SR Klein, KL Ligon, SJ Rodig, SH Ramkissoon, KL Jones…
Cancer immunology research, 2016AACR
Inhibition of immune checkpoints, including cytotoxic T-lymphocyte antigen-4 (CTLA-4),
programmed death-1 (PD-1), and its ligand PD-L1, has demonstrated exciting and durable
remissions across a spectrum of malignancies. Combinatorial regimens blocking
complementary immune checkpoints further enhance the therapeutic benefit. The activity of
these agents for patients with glioblastoma, a generally lethal primary brain tumor
associated with significant systemic and microenvironmental immunosuppression, is not …
Abstract
Inhibition of immune checkpoints, including cytotoxic T-lymphocyte antigen-4 (CTLA-4), programmed death-1 (PD-1), and its ligand PD-L1, has demonstrated exciting and durable remissions across a spectrum of malignancies. Combinatorial regimens blocking complementary immune checkpoints further enhance the therapeutic benefit. The activity of these agents for patients with glioblastoma, a generally lethal primary brain tumor associated with significant systemic and microenvironmental immunosuppression, is not known. We therefore systematically evaluated the antitumor efficacy of murine antibodies targeting a broad panel of immune checkpoint molecules, including CTLA-4, PD-1, PD-L1, and PD-L2 when administered as single-agent therapy and in combinatorial regimens against an orthotopic, immunocompetent murine glioblastoma model. In these experiments, we observed long-term tumor-free survival following single-agent anti–PD-1, anti–PD-L1, or anti–CTLA-4 therapy in 50%, 20%, and 15% of treated animals, respectively. Combination therapy of anti–CTLA-4 plus anti–PD-1 cured 75% of the animals, even against advanced, later-stage tumors. In long-term survivors, tumor growth was not seen upon intracranial tumor rechallenge, suggesting that tumor-specific immune memory responses were generated. Inhibitory immune checkpoint blockade quantitatively increased activated CD8+ and natural killer cells and decreased suppressive immune cells in the tumor microenvironment and draining cervical lymph nodes. Our results support prioritizing the clinical evaluation of PD-1, PD-L1, and CTLA-4 single-agent targeted therapy as well as combination therapy of CTLA-4 plus PD-1 blockade for patients with glioblastoma. Cancer Immunol Res; 4(2); 124–35. ©2015 AACR.
AACR