1. Academic Validation
  2. Inhibiting myeloid-derived suppressor cell trafficking enhances T cell immunotherapy

Inhibiting myeloid-derived suppressor cell trafficking enhances T cell immunotherapy

  • JCI Insight. 2019 Apr 4;4(7):e126853. doi: 10.1172/jci.insight.126853.
Lillian Sun 1 Paul E Clavijo 1 Yvette Robbins 1 Priya Patel 1 Jay Friedman 1 Sarah Greene 1 Rita Das 2 Chris Silvin 2 Carter Van Waes 2 Lucas A Horn 3 Jeffrey Schlom 3 Claudia Palena 3 Dean Maeda 4 John Zebala 4 Clint T Allen 1 5
Affiliations

Affiliations

  • 1 Translation Tumor Immunology Program and.
  • 2 Tumor Biology Section, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland, USA.
  • 3 Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
  • 4 Syntrix Pharmaceuticals, Auburn, Washington, USA.
  • 5 Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
Abstract

Recruitment of myeloid-derived suppressor cells (MDSCs) into tumors induces local immunosuppression in carcinomas. Here, we assessed whether SX-682, an orally bioavailable small-molecule inhibitor of CXCR1 and CXCR2, could block tumor MDSC recruitment and enhance T cell activation and antitumor immunity following multiple forms of immunotherapy. CXCR2+ neutrophilic MDSCs (PMN-MDSCs) were the most abundant myeloid cell subset within oral and lung syngeneic carcinomas. PMN-MDSCs demonstrated greater suppression of tumor-infiltrating lymphocyte killing of targets compared with macrophages. SX-682 significantly inhibited trafficking of PMN-MDSCs without altering CXCR2 ligand expression. Trafficking of CXCR1+ macrophages was unaltered, possibly due to coexpression of CSF1R. Reduced PMN-MDSC tumor infiltration correlated with enhanced accumulation of endogenous or adoptively transferred T cells. Accordingly, tumor growth inhibition or the rate of established tumor rejection following programed death-axis (PD-axis) immune checkpoint blockade or adoptive cell transfer of engineered T cells was enhanced in combination with SX-682. Despite CXCR1/2 expression on tumor cells, SX-682 appeared to have little direct antitumor effect on these carcinoma models. These data suggest that tumor-infiltrating CXCR2+ PMN-MDSCs may prevent optimal responses following both PD-axis immune checkpoint blockade and adoptive T cell transfer therapy. Abrogation of PMN-MDSC trafficking with SX-682 enhances T cell-based immunotherapeutic efficacy and may be of benefit to patients with MDSC-infiltrated cancers.

Keywords

Immunotherapy; Oncology.

Figures
Products