Sex-specific T cell exhaustion drives differential immune responses in glioblastoma

Sex differences in glioblastoma (GBM) incidence and outcome are well recognized, and 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 GBM models, we demonstrate that T cells play a critical role in driving GBM sex differences. Male mice exhibited accelerated tumor growth, with decreased T cell infiltration and increased T cell exhaustion. Furthermore, a higher frequency of progenitor exhausted T cells was found in males, with improved responsiveness to anti-PD1 treatment. Bone marrow chimera and adoptive transfer models indicated that T cell-mediated tumor control was predominantly regulated in a cell-intrinsic manner, which was further corroborated by in vitro exhaustion assays. Moreover, increased T cell exhaustion was observed in male GBM patients. These findings demonstrate sex-specific pre-determined behavior of T cells is critical in inducing sex differences in GBM progression and immunotherapy response. Statement of significance Immunotherapies in GBM patients have been unsuccessful due to a variety of factors including the highly immunosuppressive tumor microenvironment in GBM. This study demonstrates that sex-specific T cell behaviors are predominantly intrinsically regulated, further suggesting sex-specific approaches can be leveraged to potentially improve therapeutic efficacy of immunotherapy in GBM.

[1]  R. Cui,et al.  Brain–immune interaction mechanisms: Implications for cognitive dysfunction in psychiatric disorders , 2022, Cell proliferation.

[2]  G. Giaccone,et al.  Sex and cancer immunotherapy: Current understanding and challenges. , 2022, Cancer cell.

[3]  Lingling Wu,et al.  Androgen receptor-mediated CD8+ T cell stemness programs drive sex differences in antitumor immunity. , 2022, Immunity.

[4]  J. C. Eickhoff,et al.  Sexual dimorphism of the immune system predicts clinical outcomes in glioblastoma immunotherapy: A systematic review and meta-analysis , 2022, Neuro-oncology advances.

[5]  Ansuman T. Satpathy,et al.  Epigenetic regulation of T cell exhaustion , 2022, Nature Immunology.

[6]  A. Arnold,et al.  Sex differences in NK cells mediated by the X-linked epigenetic regulator UTX , 2022, bioRxiv.

[7]  Lei Zhou,et al.  Androgen conspires with the CD8+ T cell exhaustion program and contributes to sex bias in cancer , 2022, Science Immunology.

[8]  Susan E. Murray,et al.  Androgen receptor activity in T cells limits checkpoint blockade efficacy , 2022, Nature.

[9]  T. Cloughesy,et al.  Nivolumab plus radiotherapy with or without temozolomide in newly diagnosed glioblastoma: Results from exploratory phase I cohorts of CheckMate 143 , 2022, Neuro-oncology advances.

[10]  Zachary L. Skidmore,et al.  Characterization of the Genomic and Immunologic Diversity of Malignant Brain Tumors through Multisector Analysis , 2021, Cancer discovery.

[11]  G. Giaccone,et al.  Sex-based dimorphism of anticancer immune response and molecular mechanisms of immune evasion , 2021, Clinical Cancer Research.

[12]  S. Klein,et al.  Sex disparities matter in cancer development and therapy , 2021, Nature Reviews Cancer.

[13]  A. Arnold,et al.  X chromosome escapee genes are involved in ischemic sexual dimorphism through epigenetic modification of inflammatory signals , 2021, Journal of neuroinflammation.

[14]  Aaron J. Johnson,et al.  Brain cancer induces systemic immunosuppression through release of non-steroid soluble mediators. , 2020, Brain : a journal of neurology.

[15]  S. Erkeland,et al.  Rapid in vitro generation of bona fide exhausted CD8+ T cells is accompanied by Tcf7 promotor methylation , 2020, PLoS pathogens.

[16]  F. Hodi,et al.  Abstract A83: Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade , 2020, Poster Presentations - Proffered Abstracts.

[17]  I. Meester,et al.  SeXY chromosomes and the immune system: reflections after a comparative study , 2020, Biology of Sex Differences.

[18]  S. Freedland,et al.  Association of Patient Sex With Efficacy of Immune Checkpoint Inhibitors and Overall Survival in Advanced Cancers: A Systematic Review and Meta-analysis , 2019, JAMA oncology.

[19]  F. Hodi,et al.  Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade , 2019, Nature Immunology.

[20]  Daniel E. Speiser,et al.  Intratumoral Tcf1+PD‐1+CD8+ T Cells with Stem‐like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy , 2019, Immunity.

[21]  P. Wen,et al.  Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma , 2018, Nature Medicine.

[22]  B. Nahed,et al.  Sequestration of T-cells in bone marrow in the setting of glioblastoma and other intracranial tumors , 2018, Nature Medicine.

[23]  C. Bettegowda,et al.  Current state of immunotherapy for glioblastoma , 2018, Nature Reviews Clinical Oncology.

[24]  S. Chanock,et al.  Sex-specific glioma genome-wide association study identifies new risk locus at 3p21.31 in females, and finds sex-differences in risk at 8q24.21 , 2018, Scientific Reports.

[25]  Jill S Barnholtz-Sloan,et al.  Females have the survival advantage in glioblastoma. , 2018, Neuro-oncology.

[26]  D. Bigner,et al.  T-Cell Exhaustion Signatures Vary with Tumor Type and Are Severe in Glioblastoma , 2018, Clinical Cancer Research.

[27]  Jingqin Luo,et al.  Sexual dimorphism in glioma glycolysis underlies sex differences in survival. , 2017, JCI insight.

[28]  A. Daud,et al.  Partially exhausted tumor-infiltrating lymphocytes predict response to combination immunotherapy. , 2017, JCI insight.

[29]  Ava C. Carter,et al.  Free-living human cells reconfigure their chromosomes in the evolution back to uni-cellularity , 2017, bioRxiv.

[30]  D. Quail,et al.  The Microenvironmental Landscape of Brain Tumors. , 2017, Cancer cell.

[31]  S. Klein,et al.  Sex differences in immune responses , 2016, Nature Reviews Immunology.

[32]  C. Libert,et al.  The X chromosome in immune functions: when a chromosome makes the difference , 2010, Nature Reviews Immunology.

[33]  Xiaowei Wang,et al.  PrimerBank: a resource of human and mouse PCR primer pairs for gene expression detection and quantification , 2009, Nucleic Acids Res..