Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Androgen receptor activity in T cells limits checkpoint blockade efficacy

Abstract

Immune checkpoint blockade has revolutionized the field of oncology, inducing durable anti-tumour immunity in solid tumours. In patients with advanced prostate cancer, immunotherapy treatments have largely failed1,2,3,4,5. Androgen deprivation therapy is classically administered in these patients to inhibit tumour cell growth, and we postulated that this therapy also affects tumour-associated T cells. Here we demonstrate that androgen receptor (AR) blockade sensitizes tumour-bearing hosts to effective checkpoint blockade by directly enhancing CD8 T cell function. Inhibition of AR activity in CD8 T cells prevented T cell exhaustion and improved responsiveness to PD-1 targeted therapy via increased IFNγ expression. AR bound directly to Ifng and eviction of AR with a small molecule significantly increased cytokine production in CD8 T cells. Together, our findings establish that T cell intrinsic AR activity represses IFNγ expression and represents a novel mechanism of immunotherapy resistance.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The immune landscape of tumours from patients with mCRPC prior to checkpoint therapy.
Fig. 2: CD8 T cell signature associated with response implicates a functional role for AR.
Fig. 3: Dual inhibition of AR and PD-1/PD-L1 improves T cell function and overall survival in mouse tumour models.
Fig. 4: Suppressing AR function in T cells promotes IFNG activity.

Similar content being viewed by others

Data availability

The sequence data generated in this study will be deposited in the Gene Expression Omnibus (GEO). Additional datasets generated during the current study for Clinical Trial NCT02312557 are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

The code for reproducibility of data is publicly available or will be available upon request.

References

  1. Beer, T. M. et al. Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J. Clin. Oncol. 35, 40–47 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Kwon, E. D. et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 15, 700–712 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Fong, P. C. et al. Pembrolizumab plus enzalutamide in abiraterone-pretreated patients with metastatic castrate resistant prostate cancer: cohort C of the phase 1b/2 KEYNOTE-365 study. J. Clin. Oncol. 37, suppl:abstr 5010 (2019)

  4. Sharma, P. et al. Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the CheckMate 650 trial. Cancer Cell. 38, 489–499 (2020).

  5. Antonarakis, E. S. et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. J. Clin. Oncol. 38, 395–405 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Conforti, F. et al. Cancer immunotherapy efficacy and patients' sex: a systematic review and meta-analysis. Lancet Oncol. 19, 737–746 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Small, E. J. et al. A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer. Clin. Cancer Res. 13, 1810–1815 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Kissick, H. T. et al. Androgens alter T-cell immunity by inhibiting T-helper 1 differentiation. Proc. Natl Acad. Sci. USA 111, 9887–9892 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Benten, W. P. et al. Functional testosterone receptors in plasma membranes of T cells. FASEB J. 13, 123–133 (1999).

    Article  CAS  PubMed  Google Scholar 

  10. Liva, S. M. & Voskuhl, R. R. Testosterone acts directly on CD4+ T lymphocytes to increase IL-10 production. J. Immunol. 167, 2060–2067 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Walecki, M. et al. Androgen receptor modulates Foxp3 expression in CD4+CD25+Foxp3+ regulatory T-cells. Mol. Biol. Cell 26, 2845–2857 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ayers, M. et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Prat, A. et al. Immune-related gene expression profiling after PD-1 blockade in non-small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma. Cancer Res. 77, 3540–3550 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 171, 934–949.e916 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Graff, J. N. et al. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget 7, 52810–52817 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Graff, J. N. et al. A phase II single-arm study of pembrolizumab with enzalutamide in men with metastatic castration-resistant prostate cancer progressing on enzalutamide alone. J. Immunother. Cancer. 8, e000642 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chen, P. L. et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 6, 827–837 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Wherry, E. J. et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Hwang, S. S. et al. mRNA destabilization by BTG1 and BTG2 maintains T cell quiescence. Science 367, 1255–1260 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  21. Lefebvre, C. et al. A human B-cell interactome identifies MYB and FOXM1 as master regulators of proliferation in germinal centers. Mol. Syst. Biol. 6, 377 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Alvarez, M. J. et al. Functional characterization of somatic mutations in cancer using network-based inference of protein activity. Nat. Genet. 48, 838–847 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chen, J. et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 567, 530–534 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lu, X. et al. Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature 543, 728–732 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Northrop, J. K., Thomas, R. M., Wells, A. D. & Shen, H. Epigenetic remodeling of the IL-2 and IFN-γ loci in memory CD8 T cells is influenced by CD4 T cells. J. Immunol. 177, 1062–1069 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Zediak, V. P., Johnnidis, J. B., Wherry, E. J. & Berger, S. L. Cutting edge: persistently open chromatin at effector gene loci in resting memory CD8+ T cells independent of transcriptional status. J. Immunol. 186, 2705–2709 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Kersh, E. N. et al. Rapid demethylation of the IFN-γ gene occurs in memory but not naive CD8 T cells. J. Immunol. 176, 4083–4093 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Pauken, K. E. et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 354, 1160–1165 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Fornes, O. et al. JASPAR 2020: update of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 48, D87–D92 (2020).

    Article  CAS  PubMed  Google Scholar 

  30. Nussing, S. et al. Efficient CRISPR/Cas9 gene editing in uncultured naive mouse T cells for in vivo studies. J. Immunol. 204, 2308–2315 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Wherry, E. J., Blattman, J. N., Murali-Krishna, K., van der Most, R. & Ahmed, R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J. Virol. 77, 4911–4927 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ahmed, R., Salmi, A., Butler, L. D., Chiller, J. M. & Oldstone, M. B. Selection of genetic variants of lymphocytic choriomeningitis virus in spleens of persistently infected mice. Role in suppression of cytotoxic T lymphocyte response and viral persistence. J. Exp. Med. 160, 521–540 (1984).

    Article  CAS  PubMed  Google Scholar 

  33. Quigley, D. A. et al. Genomic hallmarks and structural variation in metastatic prostate cancer. Cell 174, 758–769.e759 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Van Allen, E. M. et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350, 207–211 (2015).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  35. Hugo, W. et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 168, 542 (2016).

    Article  CAS  Google Scholar 

  36. Bebo, B. F., Schuster, J. C., Vandenbark, A. A. & Offner, H. Androgens alter the cytokine profile and reduce encephalitogenicity of myelin-reactive T cells. J. Immunol. 162, 35–40 (1999).

    CAS  PubMed  Google Scholar 

  37. Gubbels Bupp, M. R., Potluri, T., Fink, A. L. & Klein, S. L. The confluence of sex hormones and aging on immunity. Front. Immunol. 9, 1269 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Lin, A. A., Wojciechowski, S. E. & Hildeman, D. A. Androgens suppress antigen-specific T cell responses and IFN-γ production during intracranial LCMV infection. J. Neuroimmunol. 226, 8–19 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ashley, D. J. The two “hit” and multiple “hit” theories of carcinogenesis. Br. J. Cancer 23, 313–328 (1969).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cartwright, R. A., Gurney, K. A. & Moorman, A. V. Sex ratios and the risks of haematological malignancies. Br. J. Haematol. 118, 1071–1077 (2002).

    Article  PubMed  Google Scholar 

  41. Fish, E. N. The X-files in immunity: sex-based differences predispose immune responses. Nat. Rev. Immunol. 8, 737–744 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Cook, M. B., Chow, W. H. & Devesa, S. S. Oesophageal cancer incidence in the United States by race, sex, and histologic type, 1977–2005. Br. J. Cancer 101, 855–859 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Edgren, G., Liang, L., Adami, H. O. & Chang, E. T. Enigmatic sex disparities in cancer incidence. Eur. J. Epidemiol. 27, 187–196 (2012).

    Article  PubMed  Google Scholar 

  44. Klein, S. L. & Flanagan, K. L. Sex differences in immune responses. Nat. Rev. Immunol. 16, 626–638 (2016).

    Article  CAS  PubMed  Google Scholar 

  45. Montgomery, R. B. et al. Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res. 68, 4447–4454 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Pernigoni, N. et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science 374, 216–224 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  47. Edwards, A., Hammond, H. A., Jin, L., Caskey, C. T. & Chakraborty, R. Genetic variation at five trimeric and tetrameric tandem repeat loci in four human population groups. Genomics 12, 241–253 (1992).

    Article  CAS  PubMed  Google Scholar 

  48. Kazemi-Esfarjani, P., Trifiro, M. A. & Pinsky, L. Evidence for a repressive function of the long polyglutamine tract in the human androgen receptor: possible pathogenetic relevance for the (CAG)n-expanded neuronopathies. Hum. Mol. Genet. 4, 523–527 (1995).

    Article  CAS  PubMed  Google Scholar 

  49. Rayford, W. et al. Comparative analysis of 1152 African-American and European-American men with prostate cancer identifies distinct genomic and immunological differences. Commun. Biol. 4, 670 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Higano, C. S. et al. Real-world outcomes of sipuleucel-T treatment in PROCEED, a prospective registry of men with metastatic castration-resistant prostate. cancer. Cancer 125, 4172–4180 (2019).

    Article  CAS  PubMed  Google Scholar 

  51. He, M. X. et al. Transcriptional mediators of treatment resistance in lethal prostate cancer. Nat. Med. 27, 426–433 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Bishop, J. L. et al. PD-L1 is highly expressed in Enzalutamide resistant prostate cancer. Oncotarget 6, 234–242 (2015).

    Article  PubMed  Google Scholar 

  53. Aggarwal, R. et al. Clinical and genomic characterization of treatment-emergent small-cell neuroendocrine prostate cancer: a multi-institutional prospective study. J. Clin. Oncol. 36, 2492–2503 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Moran, A. E., Polesso, F. & Weinberg, A. D. Immunotherapy expands and maintains the function of high-affinity tumor-infiltrating CD8 T cells in situ. J. Immunol. 197, 2509–2521 (2016).

    Article  CAS  PubMed  Google Scholar 

  55. Polesso, F., Sarker, M., Weinberg, A. D., Murray, S. E. & Moran, A. E. OX40 agonist tumor immunotherapy does not impact regulatory T cell suppressive function. J. Immunol. 203, 2011–2019 (2019).

    Article  CAS  PubMed  Google Scholar 

  56. Polesso, F., Weinberg, A. D. & Moran, A. E. Late-stage tumor regression after PD-L1 blockade plus a concurrent OX40 agonist. Cancer Immunol. Res. 7, 269–281 (2019).

    Article  CAS  PubMed  Google Scholar 

  57. Schofield, D. J. et al. Activity of murine surrogate antibodies for durvalumab and tremelimumab lacking effector function and the ability to deplete regulatory T cells in mouse models of cancer. mAbs 13, 1857100 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Polesso, F. et al. PD-1-specific “blocking” antibodies that deplete PD-1+ T cells present an inconvenient variable in preclinical immunotherapy experiments. Eur. J. Immunol. 51, 1473–1481 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Valkenburg, K. C., Amend, S. R. & Pienta, K. J. Murine prostate micro-dissection and surgical castration. J. Vis. Exp. https://doi.org/10.3791/53984 (2016).

  60. Pavese, J., Ogden, I. M. & Bergan, R. C. An orthotopic murine model of human prostate cancer metastasis. J. Vis. Exp. https://doi.org/10.3791/50873 (2013).

  61. Moran, A. E. et al. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J. Exp. Med. 208, 1279–1289 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Murray, S. E. et al. Fibroblast-adapted human CMV vaccines elicit predominantly conventional CD8 T cell responses in humans. J. Exp. Med. 214, 1889–1899 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gruner, B. M. et al. An in vivo multiplexed small-molecule screening platform. Nat. Methods 13, 883–889 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Wu, D. & Smyth, G. K. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res. 40, e133 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Robertson, A. G. et al. Integrative analysis identifies four molecular and clinical subsets in uveal melanoma. Cancer Cell 32, 204–220.e215 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  68. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinf. 12, 323 (2011).

    Article  CAS  Google Scholar 

  69. Ellrott, K. et al. Scalable open science approach for mutation calling of tumor exomes using multiple genomic pipelines. Cell Syst. 6, 271–281.e277 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Adey and R. Searle for sharing expertise as we developed protocols for single-cell RNA-seq. We are grateful to the OHSU Department of Comparative Medicine for outstanding animal husbandry, the Massively Parallel Sequencing Shared Resource (MPSSR) for their support, and the Knight Cancer Institute Prostate Programs outstanding clinical research team. This work is funded in part by the Collins Medical Trust, OHSU Foundation, Prostate Cancer Foundation, Pacific Northwest Prostate Cancer SPORE NCI 5P50CA097186, NIH 1R37 CA263592-01 (to A.E.M.), M.J. Murdock Charitable Trust NS-201812034 (to S.E.M.), and a sponsored research agreement with MedImmune (to A.E.M.). This work is also supported by Medical Research Foundation at Oregon, NIH 5K01LM012877 and NIH 1R21HL145426 (to Z.X.). The resources of the Exacloud high performance computing environment developed jointly by OHSU and Intel and the technical support of the OHSU Advanced Computing Center are gratefully acknowledged. BioRender.com software was used for the creation of some figures.

Author information

Authors and Affiliations

Authors

Contributions

A.E.M. conceived the study, designed and performed experiments, interpreted data and wrote the manuscript. Z.X., X.G., and C.W. designed, performed and interpreted computational analysis. X.G., F.P. and A.E.M. wrote the manuscript. F.P. and C.H. designed, performed and interpreted mouse experiments. C.W. prepared samples for sequencing. A.S. performed ChIP experiments. S.A.H. interpreted data and contributed to manuscript writing. J.N.G. conducted the clinical trial. R.F.T. and M.A.W. performed WES analysis, variant calling and tumour mutational analysis. R.M.H., S.E.M. and B.C. performed experiments for resubmission, interpretated data and contributed to writing and/or editing the manuscript. G.V.T. reviewed pathology.

Corresponding author

Correspondence to Amy E. Moran.

Ethics declarations

Competing interests

R.F.T. and J.N.G. are employees of the US Government. The contents do not represent the views of the US Department of Veterans Affairs or the United States Government. S.A.H. is an employee of AstraZeneca. A.E.M. received research funding from AstraZeneca.

Peer review

Peer review information

Nature thanks Gerhardt Attard, Joushua Rubin and the other, anonymous, reviewers for their contribution to the peer review of this work. Peer review reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Clinical trial scheme of patients enrolled and details on biopsy location and genomics.

a. Clinical trial study scheme. b. Per-patient tumor mutations are shown in a table with each row representing an individual participant on study, and each column representing the unique participant identifier (StudyID), the participant’s response to study treatment (Outcome), the site of biopsied tissue specimen analyzed (Biopsy Site), the relative (%) change in PSA with treatment (PSA change), the number of somatic variants detected in that tumor specimen (Somatic_variant_count), and the coverage-adjusted tumor mutational burden defined as the Somatic_variant_count / #Mbp genome covered by ≥ 6 reads (Coverage_adj_mtl_burden). c. Comparison of the somatic variant counts (left) or coverage-adjusted tumor mutational burdens (right) for study responders (R, n = 3 patients) versus non-responders (NR, n = 5 patients); NS represents no significant difference detected by two-tailed Student’s t-test; mean values are depicted as bold horizontal lines. Error bars represent S.E.M.

Extended Data Fig. 2 CD8 T cell subset associated with response to checkpoint therapy in mCRPC patients.

a, Representative flow cytogram for sorting tumor-associated leukocytes prior to scRNAseq. b, UMAP of all single cells (n = 16,044 cells) in this study colored by patient. c, Stack bar graph showing the % of cells per sample for immune cell clusters across each patient biopsy. d, e, Box plots comparing the % of cells per sample for immune cell clusters between responders (n = 3 patients) and non-responders (n = 5 patients). Percentage was calculated out of all immune cells (d) or all T/NK cells (e). Two-tailed unpaired Student’s t-test. Box center line, median; box, the interquartile range (IQR, the range between the 25th and 75th percentile); whiskers, 1.58 times IQR. f, Heatmap showing the expression of CTLA4, HAVCR2, PDCD1, TIGIT, CD274, LAG3, ICOS, BTLA in various T cell clusters. g, Pathways enriched in dysfunctional CD8 T cells (C4 cluster). h, Percentage of cells co-expressing a combination of PDCD1, LAG3, HAVCR2, CTLA4, TNFRSF4, and TIGIT in dysfunctional CD8 T cells (C4 cluster)

Source data

Extended Data Fig. 3 Expression of various genes associated with CD8 T cytotoxicity and exhaustion.

a, Venn diagram and contingency table showing the significant overlap between CD8_R and CD8_k1 (Top, P< 0.0001) and between CD8_NR and CD8_k2 (Bottom, P < 0.0001). All cells: all the single cells that passed quality control in this study, as shown in Fig. 1a. Two-tailed Fisher’s exact test. b, Percentage of CD8_k1 or CD8_k2 clusters per sample in responders (n = 3 patients) and non-responders (n = 5 patients). Two-tailed unpaired Student’s t-test. Box center line, median; box, the interquartile range (IQR, the range between the 25th and 75th percentile); whiskers, 1.58 times IQR. c, d, Violin plot comparing the gene expression in CD8_k1 and CD8_k2 (c), and CD8_R and CD8_NR (d). R, responder; NR, non-responder

Source data

Extended Data Fig. 4 CD4_k1 is not associated with response.

a, UMAP plot showing the two distinct CD4 T cells states identified using k-means clustering (n = 5,322 cells). b, UMAP plot showing CD4 T cells colored by response and non-response patient groups (n = 5,322 cells). c, Percentage of CD4_k1 or CD4_k2 clusters per sample in responders (n = 3 patients) and non-responders (n = 5 patients). Two-tailed unpaired Student’s t-test. Box center line, median; box, the interquartile range (IQR, the range between the 25th and 75th percentile); whiskers, 1.58 times IQR. R, responder; NR, non-responder.

Source data

Extended Data Fig. 5 Survival data following orthotopic PPSM implantation and enzalutamide + anti-PD-L1 treatment.

a, Ar expression by qPCR in mouse CD8 T cells, as compared with PPSM and 688m AR positive and negative control cell lines, respectively. Data combined from 3 independent experiments. b, Summary table of the experiments described in Fig. 3a. c, Average tumor growth of PPSM tumor bearing animals treated with different treatment combination as described in Fig. 3a. Data combined from 4 independent experiments, 8 to 10 animals per group. d, 12–14 wk old male mice were orchiectomized and PPSM tumor cells were injected orthotopically in the anterior lobe of the prostate. One week later, animals were treated with enzalutamide or enzalutamide + anti-PD-L1 (5 animals per group). 4 weeks post tumor inoculation, tumors were collected and measured. e–f, PPSM tumor bearing animals were treated along the same timeline as Fig. 3a but in the absence of ADT. Average tumor growth (e) and survival curves (f) of tumor bearing animals treated with combination therapy in the presence or absence of ADT (data depict one representative experiment of two experiments, 8 animals per group). g, Survival curves of PPSM tumor bearing animals orchiectomized or not at day 7 (5 animals per group). h, Average tumor growth of PPSM tumor bearing animals treated with combination therapy and α-CD8 depleting antibody (data depict one representative experiment of two experiments, 10 animals per group). Error bars represent S.E.M. Two-way ANOVA was used for c, e and h, and log-rank (Mantel-Cox) was used for f and g.

Source data

Extended Data Fig. 6 Phenotyping data of tumor infiltrating CD8 T cells from orthotopic PPSM tumors, degarelix treated, and enzalutamide + anti-PD-L1 treated.

ad, PPSM tumor bearing animals were treated as in Fig 3a. CD8 T cell number (a), Ki67 expression (b), PD-1 MFI (c) and CD44 MFI (d) in CD8 T cells in the tumor the day after the 3rd treatment with α-PD-L1. Data representative of 3 independent experiments with 3 animals per group. eg, PPSM tumor cells were surgically injected orthotopically in the prostate, and orchiectomy was performed. One week later, animals were treated with enzalutamide only or enzalutamide + α-PD-L1 (5 animals per group). 4 weeks post tumor inoculation, tumors were harvested and processed for flow cytometry. Graphs show percent IFNγ+ (e), TNFα+ (f) and IFNγ+TNFα+ double producing (g) CD8 T cells in the tumor (n = 5 animals). hl, PPSM tumor bearing animals underwent ADT (degarelix, 1 dose, d14 post tumor inoculation), enzalutamide (started at d14) and α-PD-L1 (3 doses, d14, 17, 20). Tumors were harvested on day 21 and processed for flow cytometry. Graphs show percent Ki67+ (h), IFNγ+ (i), TNFα+ (j), IFNγ+TNFα+ (k) and granzyme B+ (l) CD8 T cells in the tumor. Data representative of 2 independent experiments with 3 animals per group. m–o, PPSM tumor bearing animals were treated with the same timeline as in Fig. 3a, but with enzalutamide + α-PD-L1 or ADT + α-PD-L1. Tumors were harvested the day after the 3rd dose of α-PD-L1 and processed for flow cytometry. m, Percent granzyme B+ CD8 T cells in the tumor. n, Representative flow cytogram showing IFNγ and TNFα expression in CD8 T cells in the tumor, and o, Summarized percent IFNγ+TNFα+ CD8 T cells in the tumor. Data representative of 2 independent experiments with 3 animals per group. Error bars represent S.E.M. Two-tailed unpaired Student t-test.

Source data

Extended Data Fig. 7 Enzalutamide treatment leads to increased cytokine production in tumour specific T cells.

a, Experimental design. Male or female Ripm-OVA animals were implanted with MCA-OVA tumours. Male animals were treated with ADT (degarelix) at time of tumour inoculation. At d7 animals were adoptively transferred with OT1;Thy1.1 CD8 T cells, and half of the animals were started on enzalutamide treatment (5 animals per group). 12 days post adoptive transfer, tumors were harvested, and TILs were stimulated with SIINFEKL peptide followed by ICCS. b, c, Representative flow cytograms showing CD44 and IFNγ expression in OTI T cells in the tumor, and summarized % IFNγ+ and PD-1 MFI in OTI in the tumor in males (b) and females (c). Data representative of 2 independent experiments with 5 animals per group ICCS; intra-cellular cytokine staining. Error bars represent S.E.M. Two-tailed unpaired Student t-test.

Source data

Extended Data Fig. 8 T cell deletion of Ar.

a, Open chromatin regions (OCRs) containing predicted androgen receptor elements (AREs) in Ifng and Gzmb loci. b, Experimental design of the generation of Ar-KO CD8 T cells in vitro using CRISPR/Cas9. Purified CD8 T cells were electroporated with Cas9/gRNA complex (NT or AR gRNA), and put in culture in vitro for 3 days in plates coated with α-CD3 and α-CD28. 3 days later, stimulated cells were harvested, and RNA was extracted or cells were restimulated in vitro for 5 h with PMA/Ionomycin, followed by ICCS (made with www.BioRender.com). c, Ar mRNA levels by qPCR in CD8 T cells electroporated with non-targeting (NT) or Ar gRNA/Cas9 after 3 days of in vitro stimulation. Data representative of 4 independent experiments with 3 replicate wells. d, Representative flow cytograms of IFNγ and TNFα expression after restimulation with PMA/Ionomycin. e, Schematic of LCMV experiment (made with www.BioRender.com), 3 animals per group. f, Ar mRNA levels in purified P14 at day 7 post adoptive transfer (from experiment described in Fig. 4e–g). Data representative of 2 independent experiments with 3 replicate wells. g, PD1 MFI and percent IFNγ+ in P14 in the blood at day 7 post adoptive transfer. Error bars represent S.D. for c and f, and S.E.M for g.

Source data

Supplementary information

Supplementary Information

This file contains Supplementary Figs. 1–3.

Reporting Summary

Peer Review File

Supplementary Table 1

Leukocyte subset frequency per patient of total cells captured by scRNA sequencing.

Supplementary Table 2

Marker genes for each T cell cluster.

Supplementary Table 3

Differentially expressed genes of CD8 k1 vs CD8 k2.

Supplementary Table 4

Differentially expressed genes of mouse TILs + enza versus TILs + ctl.

Supplementary Table 5

Open chromatin regions within the IFNG gene screened.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guan, X., Polesso, F., Wang, C. et al. Androgen receptor activity in T cells limits checkpoint blockade efficacy. Nature 606, 791–796 (2022). https://doi.org/10.1038/s41586-022-04522-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04522-6

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing