CCR2 inhibition reduces tumor myeloid cells and unmasks a checkpoint inhibitor effect to slow progression of resistant murine gliomas

Proc Natl Acad Sci U S A. 2020 Jan 14;117(2):1129-1138. doi: 10.1073/pnas.1910856117. Epub 2019 Dec 26.

Abstract

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+/Ly6Chi/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 IFNγ 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.

Keywords: glioblastoma; immunotherapy; myeloid-derived suppressor cells; programmed death-1; tumor microenvironment.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • B7-H1 Antigen / antagonists & inhibitors*
  • CD4-Positive T-Lymphocytes
  • CD8-Positive T-Lymphocytes
  • CX3C Chemokine Receptor 1 / genetics
  • CX3C Chemokine Receptor 1 / metabolism
  • Chemokine CCL2
  • Disease Models, Animal
  • Gene Knock-In Techniques
  • Glioblastoma / drug therapy
  • Glioblastoma / pathology
  • Glioma / drug therapy*
  • Glioma / pathology
  • Humans
  • Immunotherapy
  • Lymphocytes, Tumor-Infiltrating / drug effects
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Myeloid Cells / drug effects
  • Myeloid Cells / metabolism*
  • Myeloid-Derived Suppressor Cells / metabolism
  • Programmed Cell Death 1 Receptor
  • Receptors, CCR2 / drug effects*
  • Receptors, CCR2 / genetics
  • Receptors, CCR2 / metabolism*
  • Survival Analysis
  • Tumor Microenvironment / drug effects

Substances

  • B7-H1 Antigen
  • CX3C Chemokine Receptor 1
  • Ccl2 protein, mouse
  • Ccr2 protein, mouse
  • Cd274 protein, mouse
  • Chemokine CCL2
  • Cx3cr1 protein, mouse
  • Pdcd1 protein, mouse
  • Programmed Cell Death 1 Receptor
  • Receptors, CCR2