Flores-Toro et al. studied the role of CCR2 in PD-1-resistant, syngeneic glioma models. Genetic or pharmacologic approaches to CCR2 inhibition significantly improved survival in combination with anti-PD-1 compared to either monotherapy. The combination resulted in elevated TILs with increased IFNγ expression and decreased PD-1+/Tim3+ CD4+ and CD8+ T cells. CCR2 deficiency correlated with decreased CD45hi/CD11b+/Ly6Chi MDSCs in tumors and concomitant increases in bone marrow (BM), suggesting CCR2 blockade reduced egress of BM MDSCs to gliomas, thus attenuating immunosuppression.

Contributed by Katherine Turner

Immunotherapy directed at the PD-L1/PD-1 axis has produced treatment advances in various human cancers. Unfortunately, progress has not extended to glioblastoma (GBM), with phase III clinical trials assessing anti-PD-1 monotherapy failing to show efficacy in newly diagnosed and recurrent tumors. Myeloid-derived suppressor cells (MDSCs), a subset of immunosuppressive myeloid derived cells, are known to infiltrate the tumor microenvironment of GBM. Growing evidence suggests the CCL2-CCR2 axis is important for this process. This study evaluated the combination of PD-1 blockade and CCR2 inhibition in anti-PD-1-resistant gliomas. CCR2 deficiency unmasked an anti-PD-1 survival benefit in KR158 glioma-bearing mice. CD11b(+)/Ly6C(hi)/PD-L1(+) MDSCs within established gliomas decreased with a concomitant increase in overall CCR2(+) cells and MDSCs within bone marrow of CCR2-deficient mice. The CCR2 antagonist CCX872 increased median survival as a monotherapy in KR158 glioma-bearing animals and further increased median and overall survival when combined with anti-PD-1. Additionally, combination of CCX872 and anti-PD-1 prolonged median survival time in 005 GSC GBM-bearing mice. In both models, CCX872 decreased tumor associated MDSCs and increased these cells within the bone marrow. Examination of tumor-infiltrating lymphocytes revealed an elevated population, increased IFNgamma expression, indicating enhanced cytolytic activity, as well as decreased expression of exhaustion markers in CD4(+) and CD8(+) T cells following combination treatment. These data establish that combining CCR2 and PD-1 blockade extends survival in clinically relevant murine glioma models and provides the basis on which to advance this combinatorial treatment toward early-phase human trials.

Author Info: (1) Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610. (2) Department of Pharmacology & Therapeutics, College of Medicine

Author Info: (1) Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610. (2) Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610. (3) Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610. (4) Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610. (5) ChemoCentryx, Mountain View, CA 94043. (6) ChemoCentryx, Mountain View, CA 94043. (7) ChemoCentryx, Mountain View, CA 94043. (8) ChemoCentryx, Mountain View, CA 94043. (9) Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114. (10) Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; jain@steele.mgh.harvard.edu jharriso@ufl.edu. (11) Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL 32610. Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32610. (12) Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610; jain@steele.mgh.harvard.edu jharriso@ufl.edu. Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL 32610.