Abstract

Abstract Sex differences in glioblastoma (GBM) incidence and outcome are well recognized; males show higher incidence and worse prognosis than females. Emerging evidence suggests that these extend to genetic/epigenetic and cellular differences, including immune responses. However, the mechanisms driving immunological sex differences are not fully understood. Using orthotopic GBM animal models, we recapitulated these sex differences in immune-competent B6 mice but not in immune-deficient mouse strains (NSG and RAG1KO). By depleting CD8+ T cells, we further confirm that CD8+ T cells play a critical role in driving sex differences in survival. Flow cytometry analysis revealed that male CD8+ T cells from tumor expressed higher levels of exhaustion markers such as PD-1, CTLA-4 and Tox, with higher frequency of progenitor exhausted T cell subset whereas female tumor contained more effector-like CD8+ T cells expressing elevated cytokine production. Anti-PD-1 antibody treatment exclusively extended the survival of male mice via inducing decreased exhausted CD8+ T cell subsets and increased effector function and proliferation. Survival analysis on bone marrow chimera model and adoptive transfer model indicated that T cell-mediated tumor control was predominantly regulated in a cell-intrinsic manner, as the sex of donor cells was a critical contributing factor. Finally, we confirmed these observations in GBM patient tumor samples with male tumors having a higher frequency of progenitor exhausted T cells with increased TOX expression. Taken together, these findings demonstrate sex-specific pre-determined behavior of T cells is critical in inducing sex differences in GBM progression and immunotherapy responses. Supported by grants from NIH (R35 NS127083, P01 CA245705).

Full Text
Published version (Free)

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