[HTML][HTML] Inhibiting myeloid-derived suppressor cell trafficking enhances T cell immunotherapy

L Sun, PE Clavijo, Y Robbins, P Patel, J Friedman… - JCI insight, 2019 - ncbi.nlm.nih.gov
L Sun, PE Clavijo, Y Robbins, P Patel, J Friedman, S Greene, R Das, C Silvin, C Van Waes…
JCI insight, 2019ncbi.nlm.nih.gov
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 …
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.
ncbi.nlm.nih.gov