Abstract

Abstract Recent literature demonstrates that tumor hijacks physiological mechanisms of C-type lectin receptors (CLRs) that normally restrain immune cell-mediated tissue damage to suppress myeloid cell activation and promotes immune escape. We previously demonstrated that the orphan CLR CLEC-1 expressed by myeloid cells, is enhanced by the immunosuppressive cytokine TGFβ and tempers downstream CD4+ Th1 and Th17 responses following sterile inflammation. Interestingly, we observed a high CLEC-1 expression by human CD11b+ myeloid cells from ovarian ascites and from in-vitro generated pro-tumoral M2 macrophages. Moreover, open-source data show that Clec1a is highly expressed by XCR1-expressing mouse dendritic cells (DC). As these cDC1 are specialized in cross-presentation of dead cell-associated antigens, we evaluated whether CLEC-1 could be a receptor of damaged cells and regulate anti-tumor immunity. Interestingly, we observed that both human and mouse CLEC-1 bind specifically to secondary necrotic cells. No binding was observed with viable, primary necrotic or with early apoptotic cells suggesting that the ligand(s) of CLEC-1 correspond(s) to Damage-Associated Molecular Pattern(s) induced by the cell death process. Then, to test if CLEC-1 in DCs influences cross-presentation of dead cell-associated antigens, we immunized WT and Clec1a KO mice with OVA-loaded dead cells and co-injected OVA-specific TCR transgenic CD8+ OT-I T cells. Impressively, we observed an increased proliferation of CD8+ T cells in the absence of CLEC-1, demonstrating that CLEC-1 in DCs tempers cross-presentation of dead cell-associated antigens. Interestingly, we observed in a model of subcutaneous MC38 colon carcinoma that the combination of the absence of CLEC-1 with cytotoxic and immunogenic chemotherapy (cyclophosphamide), reduces significantly the tumor growth and cures most of the mice. Similarly, in orthotopic models of mesothelioma (AK7) or hepatocarcinoma (Hepa1.6), we observed a prolongation of survival of Clec1a KO mice. Mechanistically, this was associated with an increase of effector CD69+ and of central memory CD44hi CD62Lhi CCR7hi CD4+ and CD8+ T cells in both liver and spleen. Furthermore, we observed a decrease in myeloid-derived suppressor cells. Besides, macrophages display a more mature phenotype relative to MHC class II expression. In addition, transcriptomics analysis performed in tumor-bearing liver of Clec1a KO mice revealed a profound decrease in CSF1 expression suggesting a defect in immunosuppressive myeloid cell recruitment. Moreover, we observed an increase in CCL17 and CCL7 expression, both known to activate CD8+ T cells through DCs and to enhance anti-tumor immunity. Taken together, these results suggest that CLEC-1 in DCs by acting as sensor of cell damage tempers immune response and represents a novel pharmacological target for cancer immunotherapy. Citation Format: Marion Drouin, Javier Saenz, Bérangère Evrard, Vanessa Gauttier, Géraldine Teppaz, Maria-Dolores Lopez-Robles, Cédric Louvet, Nicolas Poirier, Elise Chiffoleau. CLEC-1 suppress dendritic cell antigen presentation and is a novel myeloid immune checkpoint target for cancer immunotherapy [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3423.

Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call