Abstract

Abstract Sex differences in cancer susceptibility and mortality are well documented. In general, men have a higher cancer incidence and poorer outcomes when compared to women. Of immunologic relevance, and in contrast to cancer, women account for almost 80% of the autoimmune cases reported. These findings suggest male sex is a ‘risk factor’ for cancer but protective against autoimmunity and also highlight the potential involvement of sex hormones in regulating the immune response. Several recent publications have put a spotlight on androgen-mediated immune regulation and its impact on anti-tumor immunity. Guan and colleagues demonstrated that blockade of sex hormones in prostate tumor-bearing mice resulted in improved adaptive immune responses via direct modulation of CD8+ T cell-intrinsic androgen receptor signaling. Androgen-deprivation led to increased interferon gamma (IFNγ) expression by T cells and improved overall anti-tumor immunity. Additionally, others have shown using non-reproductive cancer mouse models (melanoma, bladder, colon and liver) that androgen receptor signaling promotes T cell exhaustion, and inhibits the activity and stemness of anti-tumor CD8+ T cells. Importantly, there is a clear role for androgen-mediated immune regulation in non-reproductive human cancer given a recent publication showing improved progression-free survival and overall survival in female versus male melanoma patients who were treated with neoadjuvant BRAF/MEK-targeted therapy. Follow-up preclinical mouse models showed significantly higher expression of the androgen receptor in tumors of both male and female BRAF/MEK-treated mice versus control and pharmacological inhibition of androgen receptor signaling improved responses to BRAF/MEK-targeted therapy in both male and female mice. Taken together, these publications stress the importance of understanding androgen-mediated immune control in both male and female cancer patients and the broad applicability of this signaling pathway in tissues including, but also beyond, the reproductive tract. There is mounting evidence to support a role androgen signaling in T cell biology. However, professional antigen presenting cells such as dendritic cells (DCs) are an essential part of generating robust anti-tumor T cell immunity were not addressed in these studies. Notably, DCs have been reported to be sensitive to androgens. Castration studies in mice found that removal of testosterone increased antigen presentation and expression of co-stimulatory molecules on DCs. In addition, hypogonadal men are at elevated risk of developing autoimmunity. Isolation of DCs from these patients showed a significant increase in DC maturation. HIV infected DCs derived from males produced significantly less type I interferon compared to female patients, resulting in weaker CD8 T cell responses against the virus. These data establish that androgens can shape DC function, yet they fall short in identifying the mechanisms of how androgens regulate DCs and the impact this may have on anti-tumor immunity. In our initial interrogation of a single-cell RNA sequencing dataset of human prostate cancer patients, we found that human tumors with low androgen receptor activity had increased IFNγ pathway activity and that IFNγ activity was positively associated with an increase in antigen presentation. Importantly, IFNγ is a cytokine that strongly induces antigen processing and presentation by antigen presenting cells and androgen deprivation therapy increases T cell derived IFNγ in tumor bearing mice suggesting that androgen receptor activity may also regulate antigen presentation. Furthermore, our analysis showed that elevated androgen receptor activity correlated with lower antigen presentation, conceivably through suppression of DC function. We hypothesized that inhibition of androgen receptor-mediated suppression of DC function would improve antigen presentation and anti-tumor T cell responses. Here we demonstrate using a B16 melanoma model that male and female DCs, both bone marrow-derived and those sorted from murine steady-state skin and melanoma tumors, express androgen receptor. Interestingly, there is a striking upregulation of androgen receptor expression specifically on type 1 conventional DCs (cDC1s) following uptake of tumor-derived antigen in vivo. cDC1s are a subset of DC that is particularly specialized in cross-presentation of exogenous antigens on MHCI. In this way, cDC1s are dominant drivers of CD8+ T cell responses and, via this function, have been shown to be essential for anti-tumor immunity. Further by inhibiting androgen receptor signaling in vitro using the clinically relevant inhibitor, enzalutamide, we find that bone marrow-derived DCs have increased phagocytic activity as shown by fluorescent bead uptake. Next, we injected male mice with B16 melanoma tumors and, after allowing the tumors to establish, we treated them with degarelix to reduce testosterone levels and enzalutamide to further inhibit androgen receptor signaling. On day 15 we sacrificed the mice, measured the tumors and interrogated DC function by flow cytometry. Strikingly, we found that inhibition of androgen receptor signaling in vivo significantly increased tumor-derived antigen uptake by cDC1s and, importantly, this correlated with improved tumor control as seen by significantly smaller tumors upon androgen receptor inhibition. These data provide insight into the regulation of DCs within the tumor microenvironment and how suppressing inhibitory hormone molecules may lead to better antigen presentation and more effective downstream T cell responses. Cancer immunotherapy is designed to generate an autoimmune-like response against an individual’s cancer. Given the striking sexual dimorphism in both autoimmunity and cancer, there is a clear and pressing need to investigate the impact of sex hormones on the effectiveness and function of cancer immunotherapy. And while myeloid cell targeted therapies and DC-based anti-tumor vaccines hold great promise for boosting anti-tumor T cells responses, current clinical use and efficacy has been limited pointing to an urgent need for improved vaccine and therapeutic design. This will rely on a better understanding of DC activation in this context. Our data indicates that there is a role for androgen receptor signaling in DC function and highlights the potential impact of DC-intrinsic androgen receptor inhibition on optimal DC vaccine design and the generation of more productive DC targeted immunotherapies. Citation Format: Kasidy Brown, Fanny Polesso, Victoria Schuster, Amy Moran, Megan Ruhland. Dendritic cell intrinsic androgen receptor signaling reduces dendritic cell function and anti-tumor immunity. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr NG01.

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