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.

  • Review Article
  • Published:

The testosterone paradox of advanced prostate cancer: mechanistic insights and clinical implications

Abstract

The discovery of the benefits of castration for prostate cancer treatment in 1941 led to androgen deprivation therapy, which remains a mainstay of the treatment of men with advanced prostate cancer. However, as early as this original publication, the inevitable development of castration-resistant prostate cancer was recognized. Resistance first manifests as a sustained rise in the androgen-responsive gene, PSA, consistent with reactivation of the androgen receptor axis. Evaluation of clinical specimens demonstrates that castration-resistant prostate cancer cells remain addicted to androgen signalling and adapt to chronic low-testosterone states. Paradoxically, results of several studies have suggested that treatment with supraphysiological levels of testosterone can retard prostate cancer growth. Insights from these studies have been used to investigate administration of supraphysiological testosterone to patients with prostate cancer for clinical benefits, a strategy that is termed bipolar androgen therapy (BAT). BAT involves rapid cycling from supraphysiological back to near-castration testosterone levels over a 4-week cycle. Understanding how BAT works at the molecular and cellular levels might help to rationalize combining BAT with other agents to achieve increased efficacy and tumour responses.

Key points

  • Androgens can drive prostate cancer growth providing the rationale for using deprivation of androgens as a first line of treatment for prostate cancer. Unfortunately, prostate cancer cells adapt to low androgen levels and eventually progress to a castration-resistant state.

  • Results of several studies have indicated a paradoxical decrease in tumour growth in prostate cancer models upon treatment with supraphysiological levels of testosterone. Evidence indicates several complementary mechanisms, including cell death and cytostasis, which might be responsible for paradoxical growth inhibition by supraphysiological testosterone.

  • Adaptive reliance on androgen signalling by castration-resistant prostate cancer cells becomes a therapeutic liability that can be exploited clinically through the administration of supraphysiological testosterone, an approach termed ‘bipolar androgen therapy’ (BAT). The term bipolar is used to emphasize that, with this strategy, rapid cycling occurs between two extremes: from supraphysiological back to near-castration testosterone levels over a 4-week cycle.

  • Understanding how BAT works at the molecular and cellular levels might help to develop biomarkers for patient stratification and to rationally combine BAT with other agents to achieve increased efficacy.

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

Access options

Fig. 1: AR structure and signalling.
Fig. 2: Androgens in prostate homeostasis and regeneration.
Fig. 3: Mechanisms of action of supraphysiological testosterone.

Similar content being viewed by others

References

  1. Sung, H. et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021).

    Article  PubMed  Google Scholar 

  2. Butenandt, A. Über die chemische Untersuchung des SexualHormons. Angew. Chem. 44, 905–908 (1931).

    Article  CAS  Google Scholar 

  3. Butenandt, A. & Tscherning, K. Androsterone, a crystalline male sex hormone. I. Isolation and purification from male urine. Z. Physiol. Chem. 229, 167 (1934).

    Article  CAS  Google Scholar 

  4. David K, D. E., Freud, J. & Laqueur, E. Über krystallinisches männliches Hormon aus Hoden (Testosteron), wirksamer als aus Harn oder aus Cholesterin bereitetes Androsteron. Hoppe Seylers Z. Physiol. Chem. 233, 281–283 (1935).

    Article  Google Scholar 

  5. Pearlman, W. H. & Crepy, O. Steroid-protein interaction with particular reference to testosterone binding by human serum. J. Biol. Chem. 242, 182–189 (1967).

    Article  CAS  PubMed  Google Scholar 

  6. Rosner, W. & Deakins, S. M. Testosterone-binding globulins in human plasma: studies on sex distribution and specificity. J. Clin. Invest. 47, 2109–2116 (1968).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pearlman, W. H. & Pearlman, M. R. The metabolism in vivo of Δ4-androstene-3, 17-dione-7-H3; its localization in the ventral prostate and other tissues of the rat. J. Biol. Chem. 236, 1321–1327 (1961).

    Article  CAS  PubMed  Google Scholar 

  8. Fang, S., Anderson, K. M. & Liao, S. Receptor proteins for androgens. On the role of specific proteins in selective retention of 17-β-hydroxy-5-α-androstan-3-one by rat ventral prostate in vivo and in vitro. J. Biol. Chem. 244, 6584–6595 (1969).

    Article  CAS  PubMed  Google Scholar 

  9. Bruchovsky, N. & Wilson, J. D. The conversion of testosterone to 5-α-androstan-17-β-ol-3-one by rat prostate in vivo and in vitro. J. Biol. Chem. 243, 2012–2021 (1968).

    Article  CAS  PubMed  Google Scholar 

  10. Imperato-McGinley, J., Guerrero, L., Gautier, T. & Peterson, R. E. Steroid 5α-reductase deficiency in man: an inherited form of male pseudohermaphroditism. Science 186, 1213–1215 (1974).

    Article  CAS  PubMed  Google Scholar 

  11. Siiteri, P. K. & Wilson, J. D. Testosterone formation and metabolism during male sexual differentiation in the human embryo. J. Clin. Endocrinol. Metab. 38, 113–125 (1974).

    Article  CAS  PubMed  Google Scholar 

  12. Anderson, K. M. & Liao, S. Selective retention of dihydrotestosterone by prostatic nuclei. Nature 219, 277–279 (1968).

    Article  CAS  PubMed  Google Scholar 

  13. Lubahn, D. B. et al. Cloning of human androgen receptor complementary DNA and localization to the X chromosome. Science 240, 327–330 (1988).

    Article  CAS  PubMed  Google Scholar 

  14. Chang, C. S., Kokontis, J. & Liao, S. T. Molecular cloning of human and rat complementary DNA encoding androgen receptors. Science 240, 324–326 (1988).

    Article  CAS  PubMed  Google Scholar 

  15. Mangelsdorf, D. J. et al. The nuclear receptor superfamily: the second decade. Cell 83, 835–839 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Velasco, A. M. et al. Identification and validation of novel androgen-regulated genes in prostate cancer. Endocrinology 145, 3913–3924 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Sahu, B. et al. FoxA1 specifies unique androgen and glucocorticoid receptor binding events in prostate cancer cells. Cancer Res. 73, 1570–1580 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Gao, S. et al. Chromatin binding of FOXA1 is promoted by LSD1-mediated demethylation in prostate cancer. Nat. Genet. 52, 1011–1017 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sahu, B. et al. Dual role of FoxA1 in androgen receptor binding to chromatin, androgen signalling and prostate cancer. EMBO J. 30, 3962–3976 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jia, L. et al. Genomic androgen receptor-occupied regions with different functions, defined by histone acetylation, coregulators and transcriptional capacity. PLoS One 3, e3645 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Visakorpi, T. et al. In vivo amplification of the androgen receptor gene and progression of human prostate cancer. Nat. Genet. 9, 401–406 (1995).

    Article  CAS  PubMed  Google Scholar 

  22. Li, Y. et al. Androgen receptor splice variants mediate enzalutamide resistance in castration-resistant prostate cancer cell lines. Cancer Res. 73, 483–489 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Huggins, C. & Hodges, C. V. Studies on prostatic cancer. I. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. Cancer Res. 1, 293–297 (1941).

    CAS  Google Scholar 

  24. Fu, A. Z. et al. Mortality and androgen deprivation therapy as salvage treatment for biochemical recurrence after primary therapy for clinically localized prostate cancer. J. Urol. 197, 1448–1454 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Sharifi, N., Gulley, J. L. & Dahut, W. L. Androgen deprivation therapy for prostate cancer. JAMA 294, 238–244 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Tangen, C. M. et al. Ten-year survival in patients with metastatic prostate cancer. Clin. Prostate Cancer 2, 41–45 (2003).

    Article  PubMed  Google Scholar 

  27. Pienta, K. J. & Bradley, D. Mechanisms underlying the development of androgen-independent prostate cancer. Clin. Cancer Res. 12, 1665–1671 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Denmeade, S. R. & Isaacs, J. T. Bipolar androgen therapy: the rationale for rapid cycling of supraphysiologic androgen/ablation in men with castration resistant prostate cancer. Prostate 70, 1600–1607 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. McNeal, J. E. Regional morphology and pathology of the prostate. Am. J. Clin. Pathol. 49, 347–357 (1968).

    Article  CAS  PubMed  Google Scholar 

  30. McNeal, J. E. Normal histology of the prostate. Am. J. Surg. Pathol. 12, 619–633 (1988).

    Article  CAS  PubMed  Google Scholar 

  31. Cunha, G. R. & Chung, L. W. Stromal-epithelial interactions — I. Induction of prostatic phenotype in urothelium of testicular feminized (Tfm/y) mice. J. Steroid Biochem. 14, 1317–1324 (1981).

    Article  CAS  PubMed  Google Scholar 

  32. Cunha, G. R. et al. Normal and abnormal development of the male urogenital tract. Role of androgens, mesenchymal-epithelial interactions, and growth factors. J. Androl. 13, 465–475 (1992).

    CAS  PubMed  Google Scholar 

  33. Isaacs, J. T. & Coffey, D. S. Etiology and disease process of benign prostatic hyperplasia. Prostate Suppl. 2, 33–50 (1989).

    Article  CAS  PubMed  Google Scholar 

  34. English, H. F., Santen, R. J. & Isaacs, J. T. Response of glandular versus basal rat ventral prostatic epithelial cells to androgen withdrawal and replacement. Prostate 11, 229–242 (1987).

    Article  CAS  PubMed  Google Scholar 

  35. Collins, A. T., Habib, F. K., Maitland, N. J. & Neal, D. E. Identification and isolation of human prostate epithelial stem cells based on α2β1-integrin expression. J. Cell Sci. 114, 3865–3872 (2001).

    Article  CAS  PubMed  Google Scholar 

  36. Bonkhoff, H. & Remberger, K. Widespread distribution of nuclear androgen receptors in the basal cell layer of the normal and hyperplastic human prostate. Virchows Arch. A Pathol. Anat. Histopathol. 422, 35–38 (1993).

    Article  CAS  PubMed  Google Scholar 

  37. Bonkhoff, H., Stein, U. & Remberger, K. The proliferative function of basal cells in the normal and hyperplastic human prostate. Prostate 24, 114–118 (1994).

    Article  CAS  PubMed  Google Scholar 

  38. Germann, M. et al. Stem-like cells with luminal progenitor phenotype survive castration in human prostate cancer. Stem Cell 30, 1076–1086 (2012).

    Article  CAS  Google Scholar 

  39. Wang, X. et al. A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature 461, 495–500 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Choi, N., Zhang, B., Zhang, L., Ittmann, M. & Xin, L. Adult murine prostate basal and luminal cells are self-sustained lineages that can both serve as targets for prostate cancer initiation. Cancer Cell 21, 253–265 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ousset, M. et al. Multipotent and unipotent progenitors contribute to prostate postnatal development. Nat. Cell Biol. 14, 1131–1138 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Wang, Z. A. et al. Lineage analysis of basal epithelial cells reveals their unexpected plasticity and supports a cell-of-origin model for prostate cancer heterogeneity. Nat. Cell Biol. 15, 274–283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wu, X. et al. Generation of a prostate epithelial cell-specific Cre transgenic mouse model for tissue-specific gene ablation. Mech. Dev. 101, 61–69 (2001).

    Article  CAS  PubMed  Google Scholar 

  44. Xie, Q. et al. Dissecting cell-type-specific roles of androgen receptor in prostate homeostasis and regeneration through lineage tracing. Nat. Commun. 8, 14284 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Karthaus, W. R. et al. Regenerative potential of prostate luminal cells revealed by single-cell analysis. Science 368, 497–505 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dai, C., Heemers, H. & Sharifi, N. Androgen signaling in prostate cancer. Cold Spring Harb. Perspect. Med. https://doi.org/10.1101/cshperspect.a030452 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Kumar, A. et al. Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer. Nat. Med. 22, 369–378 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ware, K. E., Garcia-Blanco, M. A., Armstrong, A. J. & Dehm, S. M. Biologic and clinical significance of androgen receptor variants in castration resistant prostate cancer. Endocr. Relat. Cancer 21, T87–T103 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell 163, 1011–1025 (2015).

    Article  Google Scholar 

  50. Chen, E. J. et al. Abiraterone treatment in castration-resistant prostate cancer selects for progesterone responsive mutant androgen receptors. Clin. Cancer Res. 21, 1273–1280 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Korpal, M. et al. An F876L mutation in androgen receptor confers genetic and phenotypic resistance to MDV3100 (enzalutamide). Cancer Discov. 3, 1030–1043 (2013).

    Article  CAS  PubMed  Google Scholar 

  52. Gottlieb, B., Beitel, L. K., Wu, J. H. & Trifiro, M. The androgen receptor gene mutations database (ARDB): 2004 update. Hum. Mutat. 23, 527–533 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Robinson, J. L. et al. Elevated levels of FOXA1 facilitate androgen receptor chromatin binding resulting in a CRPC-like phenotype. Oncogene 33, 5666–5674 (2014).

    Article  CAS  PubMed  Google Scholar 

  54. Wang, Q. et al. A hierarchical network of transcription factors governs androgen receptor-dependent prostate cancer growth. Mol. Cell 27, 380–392 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Pomerantz, M. M. et al. The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis. Nat. Genet. 47, 1346–1351 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Stelloo, S., Bergman, A. M. & Zwart, W. Androgen receptor enhancer usage and the chromatin regulatory landscape in human prostate cancers. Endocr. Relat. Cancer 26, R267–R285 (2019).

    Article  CAS  PubMed  Google Scholar 

  57. Westaby, D. et al. A new old target: androgen receptor signaling and advanced prostate cancer. Annu. Rev. Pharmacol. Toxicol. 62, 131–153 (2022).

    Article  PubMed  Google Scholar 

  58. Uo, T., Sprenger, C. C. & Plymate, S. R. Androgen receptor signaling and metabolic and cellular plasticity during progression to castration resistant prostate cancer. Front. Oncol. 10, 580617 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Culig, Z. & Santer, F. R. Androgen receptor signaling in prostate cancer. Cancer Metastasis Rev. 33, 413–427 (2014).

    Article  CAS  PubMed  Google Scholar 

  60. Deng, Q. et al. Non-genomic action of androgens is mediated by rapid phosphorylation and regulation of androgen receptor trafficking. Cell. Physiol. Biochem. 43, 223–236 (2017).

    Article  CAS  PubMed  Google Scholar 

  61. Leung, J. K. & Sadar, M. D. Non-genomic actions of the androgen receptor in prostate cancer. Front. Endocrinol. 8, 2 (2017).

    Article  Google Scholar 

  62. Zarif, J. C. & Miranti, C. K. The importance of non-nuclear AR signaling in prostate cancer progression and therapeutic resistance. Cell Signal. 28, 348–356 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Harris, W. P., Mostaghel, E. A., Nelson, P. S. & Montgomery, B. Androgen deprivation therapy: progress in understanding mechanisms of resistance and optimizing androgen depletion. Nat. Clin. Pract. Urol. 6, 76–85 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Huggins, C. & Scott, W. W. Bilateral adrenalectomy in prostatic cancer: clinical features and urinary excretion of 17-ketosteroids and estrogen. Ann. Surg. 122, 1031–1041 (1945).

    Article  PubMed  PubMed Central  Google Scholar 

  65. de Bono, J. S. et al. Abiraterone and increased survival in metastatic prostate cancer. N. Engl. J. Med. 364, 1995–2005 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Ryan, C. J., Smith, M. R. & Bono, J. S. Abiraterone in metastatic prostate cancer without previous chemotherapy. N. Engl. J. Med. 368, 138–148 (2013).

    Article  CAS  PubMed  Google Scholar 

  67. Fizazi, K., Tran, N. & Fein, L. Abiraterone plus prednisone in metastatic, castration-sensitive prostate cancer. N. Engl. J. Med. 377, 352–360 (2017).

    Article  CAS  PubMed  Google Scholar 

  68. Liao, S., Howell, D. K. & Chang, T. M. Action of a nonsteroidal antiandrogen, flutamide, on the receptor binding and nuclear retention of 5 α-dihydrotestosterone in rat ventral prostate. Endocrinology 94, 1205–1209 (1974).

    Article  CAS  PubMed  Google Scholar 

  69. Tran, C. et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 324, 787–790 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Beer, T. M. et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N. Engl. J. Med. 371, 424–433 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Chi, K. N. et al. Apalutamide for metastatic, castration-sensitive prostate cancer. N. Engl. J. Med. 381, 13–24 (2019).

    Article  CAS  PubMed  Google Scholar 

  72. Fizazi, K. et al. Darolutamide in nonmetastatic, castration-resistant prostate cancer. N. Engl. J. Med. 380, 1235–1246 (2019).

    Article  CAS  PubMed  Google Scholar 

  73. Hussain, M. et al. Enzalutamide in men with nonmetastatic, castration-resistant prostate cancer. N. Engl. J. Med. 378, 2465–2474 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Scher, H. I. et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N. Engl. J. Med. 367, 1187–1197 (2012).

    Article  CAS  PubMed  Google Scholar 

  75. Maron, S. B. et al. Pembrolizumab with trastuzumab and chemotherapy (PTC) in HER2-positive metastatic esophagogastric cancer (mEG): plasma and tumor-based biomarker analysis. J. Clin. Oncol. 38 (Suppl. 15), 4559 (2020).

    Article  Google Scholar 

  76. ClinicalTrials.gov. US National Library of Medicine. https://ClinicalTrials.gov/show/NCT03888612 (2021).

  77. Linja, M. J., Savinainen, K. J. & Saramäki, O. R. Amplification and overexpression of androgen receptor gene in hormone-refractory prostate cancer. Cancer Res. 61, 3550–3555 (2001).

    CAS  PubMed  Google Scholar 

  78. Azad, A. A., Volik, S. V. & Wyatt, A. W. Androgen receptor gene aberrations in circulating cell-free DNA: biomarkers of therapeutic resistance in castration-resistant prostate cancer. Clin. Cancer Res. 21, 2315–2324 (2015).

    Article  CAS  PubMed  Google Scholar 

  79. Isaacs, J. T. & Isaacs, W. B. Androgen receptor outwits prostate cancer drugs. Nat. Med 10, 26–27 (2004).

    Article  CAS  PubMed  Google Scholar 

  80. Antonarakis, E. S., Lu, C. & Wang, H. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N. Engl. J. Med. 371, 1028–1038 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Scher, H. I. & Sawyers, C. L. Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. J. Clin. Oncol. 23, 8253–8261 (2005).

    Article  CAS  PubMed  Google Scholar 

  82. Huggins, C. & Yang, N. C. Induction and extinction of mammary cancer. A striking effect of hydrocarbons permits analysis of mechanisms of causes and cure of breast cancer. Science 137, 257–262 (1962).

    Article  CAS  PubMed  Google Scholar 

  83. Horoszewicz, J. S. et al. LNCaP model of human prostatic carcinoma. Cancer Res. 43, 1809–1818 (1983).

    CAS  PubMed  Google Scholar 

  84. Berns, E. M., de Boer, W. & Mulder, E. Androgen-dependent growth regulation of and release of specific protein(s) by the androgen receptor containing human prostate tumor cell line LNCaP. Prostate 9, 247–259 (1986).

    Article  CAS  PubMed  Google Scholar 

  85. Dai, J. L., Maiorino, C. A., Gkonos, P. J. & Burnstein, K. L. Androgenic up-regulation of androgen receptor cDNA expression in androgen-independent prostate cancer cells. Steroids 61, 531–539 (1996).

    Article  CAS  PubMed  Google Scholar 

  86. Kokontis, J., Takakura, K., Hay, N. & Liao, S. Increased androgen receptor activity and altered c-myc expression in prostate cancer cells after long-term androgen deprivation. Cancer Res. 54, 1566–1573 (1994).

    CAS  PubMed  Google Scholar 

  87. Heisler, L. E. et al. Androgen-dependent cell cycle arrest and apoptotic death in PC-3 prostatic cell cultures expressing a full-length human androgen receptor. Mol. Cell. Endocrinol. 126, 59–73 (1997).

    Article  CAS  PubMed  Google Scholar 

  88. Kokontis, J. M. et al. Androgen suppresses the proliferation of androgen receptor-positive castration-resistant prostate cancer cells via inhibition of Cdk2, CyclinA, and Skp2. PLoS One 9, e109170 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Ling, M. T., Chan, K. W. & Choo, C. K. Androgen induces differentiation of a human papillomavirus 16 E6/E7 immortalized prostate epithelial cell line. J. Endocrinol. 170, 287–296 (2001).

    Article  CAS  PubMed  Google Scholar 

  90. Berthon, P. et al. Androgens are not a direct requirement for the proliferation of human prostatic epithelium in vitro. Int. J. Cancer 73, 910–916 (1997).

    Article  CAS  PubMed  Google Scholar 

  91. Antony, L., van der Schoor, F., Dalrymple, S. L. & Isaacs, J. T. Androgen receptor (AR) suppresses normal human prostate epithelial cell proliferation via AR/β-catenin/TCF-4 complex inhibition of c-MYC transcription. Prostate 74, 1118–1131 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. D’Antonio, J. M., Vander Griend, D. J. & Isaacs, J. T. DNA licensing as a novel androgen receptor mediated therapeutic target for prostate cancer. Endocr. Relat. Cancer 16, 325–332 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  93. Vander Griend, D. J., Litvinov, I. V. & Isaacs, J. T. Stabilizing androgen receptor in mitosis inhibits prostate cancer proliferation. Cell Cycle 6, 647–651 (2007).

    Article  Google Scholar 

  94. Litvinov, I. V. et al. Androgen receptor as a licensing factor for DNA replication in androgen-sensitive prostate cancer cells. Proc. Natl Acad. Sci. USA 103, 15085–15090 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Fragkos, M., Ganier, O., Coulombe, P. & Mechali, M. DNA replication origin activation in space and time. Nat. Rev. Mol. Cell Biol. 16, 360–374 (2015).

    Article  CAS  PubMed  Google Scholar 

  96. Nishitani, H., Taraviras, S., Lygerou, Z. & Nishimoto, T. The human licensing factor for DNA replication Cdt1 accumulates in G1 and is destabilized after initiation of S-phase. J. Biol. Chem. 276, 44905–44911 (2001).

    Article  CAS  PubMed  Google Scholar 

  97. Nishitani, H. & Lygerou, Z. Control of DNA replication licensing in a cell cycle. Genes Cell 7, 523–534 (2002).

    Article  CAS  Google Scholar 

  98. Shi, Y. K. et al. MCM7 interacts with androgen receptor. Am. J. Pathol. 173, 1758–1767 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Wolf, D. A., Herzinger, T., Hermeking, H., Blaschke, D. & Horz, W. Transcriptional and posttranscriptional regulation of human androgen receptor expression by androgen. Mol. Endocrinol. 7, 924–936 (1993).

    CAS  PubMed  Google Scholar 

  100. Henttu, P. & Vihko, P. Growth factor regulation of gene expression in the human prostatic carcinoma cell line LNCaP. Cancer Res. 53, 1051–1058 (1993).

    CAS  PubMed  Google Scholar 

  101. Cai, C. et al. Androgen receptor gene expression in prostate cancer is directly suppressed by the androgen receptor through recruitment of lysine-specific demethylase 1. Cancer Cell 20, 457–471 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Rudolph, T., Beuch, S. & Reuter, G. Lysine-specific histone demethylase LSD1 and the dynamic control of chromatin. Biol. Chem. 394, 1019–1028 (2013).

    Article  CAS  PubMed  Google Scholar 

  103. Cerella, C., Grandjenette, C., Dicato, M. & Diederich, M. Roles of apoptosis and cellular senescence in cancer and aging. Curr. Drug. Targets 17, 405–415 (2016).

    Article  CAS  PubMed  Google Scholar 

  104. Wang, X., Deng, H., Basu, I. & Zhu, L. Induction of androgen receptor-dependent apoptosis in prostate cancer cells by the retinoblastoma protein. Cancer Res. 64, 1377–1385 (2004).

    Article  CAS  PubMed  Google Scholar 

  105. Lin, Y. et al. Androgen and its receptor promote Bax-mediated apoptosis. Mol. Cell Biol. 26, 1908–1916 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Joly-Pharaboz, M. O. et al. Inhibition of growth and induction of apoptosis by androgens of a variant of LNCaP cell line. J. Steroid Biochem. Mol. Biol. 73, 237–249 (2000).

    Article  CAS  PubMed  Google Scholar 

  107. Roediger, J. et al. Supraphysiological androgen levels induce cellular senescence in human prostate cancer cells through the Src-Akt pathway. Mol. Cancer 13, 214 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Mirochnik, Y. et al. Androgen receptor drives cellular senescence. PLoS One 7, e31052 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Han, W. et al. Exploiting the tumor-suppressive activity of the androgen receptor by CDK4/6 inhibition in castration-resistant prostate cancer. Mol. Ther. https://doi.org/10.1016/j.ymthe.2022.01.039 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  110. Demidenko, Z. N. et al. Rapamycin decelerates cellular senescence. Cell Cycle 8, 1888–1895 (2009).

    Article  CAS  PubMed  Google Scholar 

  111. Herranz, N. et al. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat. Cell Biol. 17, 1205–1217 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Bui, A. T. et al. Transient exposure to androgens induces a remarkable self-sustained quiescent state in dispersed prostate cancer cells. Cell Cycle 16, 879–893 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Ju, B. G. et al. A topoisomerase IIβ-mediated dsDNA break required for regulated transcription. Science 312, 1798–1802 (2006).

    Article  CAS  PubMed  Google Scholar 

  114. Lin, C. et al. Nuclear receptor-induced chromosomal proximity and DNA breaks underlie specific translocations in cancer. Cell 139, 1069–1083 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Haffner, M. C. et al. Androgen-induced TOP2B-mediated double-strand breaks and prostate cancer gene rearrangements. Nat. Genet. 42, 668–675 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Tomlins, S. A. et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310, 644–648 (2005).

    Article  CAS  PubMed  Google Scholar 

  117. Kim, N. & Jinks-Robertson, S. Transcription as a source of genome instability. Nat. Rev. Genet. 13, 204–214 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Cristini, A., Geraud, M. & Sordet, O. Transcription-associated DNA breaks and cancer: a matter of DNA topology. Int. Rev. Cell Mol. Biol. 364, 195–240 (2021).

    Article  CAS  PubMed  Google Scholar 

  119. Chatterjee, P. et al. Supraphysiological androgens suppress prostate cancer growth through androgen receptor-mediated DNA damage. J. Clin. Invest. 129, 4245–4260 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Lam, H. M. et al. Durable response of enzalutamide-resistant prostate cancer to supraphysiological testosterone is associated with a multifaceted growth suppression and impaired DNA damage response transcriptomic program in patient-derived xenografts. Eur. Urol. 77, 144–155 (2020).

    Article  CAS  PubMed  Google Scholar 

  121. Markowski, M. C. et al. Molecular and clinical characterization of patients with metastatic castration resistant prostate cancer achieving deep responses to bipolar androgen therapy. Clin. Genitourin. Cancer https://doi.org/10.1016/j.clgc.2021.08.001 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  122. Markowski, M. C. et al. Extreme responses to immune checkpoint blockade following bipolar androgen therapy and enzalutamide in patients with metastatic castration resistant prostate cancer. Prostate 80, 407–411 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Teply, B. A., Kachhap, S., Eisenberger, M. A. & Denmeade, S. R. Extreme response to high-dose testosterone in BRCA2- and ATM-mutated prostate cancer. Eur. Urol. 71, 499 (2017).

    Article  CAS  PubMed  Google Scholar 

  124. Kumar, R. et al. Supraphysiologic testosterone induces ferroptosis and activates immune pathways through nucleophagy in prostate cancer. Cancer Res. 81, 5948–5962 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Torres-Estay, V. et al. Androgen receptor in human endothelial cells. J. Endocrinol. 224, R131–R137 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Mantalaris, A. et al. Localization of androgen receptor expression in human bone marrow. J. Pathol. 193, 361–366 (2001).

    Article  CAS  PubMed  Google Scholar 

  127. Blanquart, E., Laffont, S. & Guéry, J.-C. Sex hormone regulation of innate lymphoid cells. Biomed. J. 44, 144–156 (2021).

    Article  CAS  PubMed  Google Scholar 

  128. Guan, X. et al. Androgen receptor activity in T cells limits checkpoint blockade efficacy. Nature https://doi.org/10.1038/s41586-022-04522-6 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  129. Lai, J.-J. et al. Monocyte/macrophage androgen receptor suppresses cutaneous wound healing in mice by enhancing local TNF-α expression. J. Clin. Invest. 119, 3739–3751 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Tagnon, H. J., Schulman, P., Whitmore, W. F. & Leone, L. A. Prostatic fibrinolysin: study of a case illustrating role in hemorrhagic diathesis of cancer of the prostate. Am. J. Med. 15, 875–884 (1953).

    Article  CAS  PubMed  Google Scholar 

  131. Bonner, C. D., Fishman, W. H. & Homburger, F. Serum prostatic acid phosphatase and cancer of the prostate. N. Engl. J. Med. 255, 925–933 (1956).

    Article  CAS  PubMed  Google Scholar 

  132. Fowler Jr, J. E. & Whitmore Jr, W. F. The response of metastatic adenocarcinoma of the prostate to exogenous testosterone. J. Urol. 126, 372–375 (1981).

    Article  CAS  PubMed  Google Scholar 

  133. Manni, A., Bartholomew, M. & Caplan, R. Androgen priming and chemotherapy in advanced prostate cancer: evaluation of determinants of clinical outcome. J. Clin. Oncol. 6, 1456–1466 (1988).

    Article  CAS  PubMed  Google Scholar 

  134. Suarez, A. J., Lamm, D. L. & Radwin, H. M. Androgen priming and cytotoxic chemotherapy in advanced prostatic cancer. Cancer Chemother. Pharmacol. 8, 261–265 (1982).

    Article  CAS  PubMed  Google Scholar 

  135. Donati, R. M., Ellis, H. & Gallagher, N. I. Testosterone potentiated 32P therapy in prostatic carcinoma. Cancer 19, 1088–1090 (1966).

    Article  CAS  PubMed  Google Scholar 

  136. Prout Jr, G. R. & Brewer, W. R. Response of men with advanced prostatic carcinoma to exogenous administration of testosterone. Cancer 20, 1871–1878 (1967).

    Article  CAS  PubMed  Google Scholar 

  137. Khera, M. et al. Testosterone replacement therapy following radical prostatectomy. J. Sex. Med. 6, 1165–1170 (2009).

    Article  CAS  PubMed  Google Scholar 

  138. Pastuszak, A. W. et al. Testosterone replacement therapy in patients with prostate cancer after radical prostatectomy. J. Urol. 190, 639–644 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Pastuszak, A. W. et al. Testosterone replacement therapy in the setting of prostate cancer treated with radiation. Int. J. Impot. Res. 25, 24–28 (2013).

    Article  CAS  PubMed  Google Scholar 

  140. Ahlering, T. E. et al. Testosterone replacement therapy reduces biochemical recurrence after radical prostatectomy. BJU Int. 126, 91–96 (2020).

    Article  CAS  PubMed  Google Scholar 

  141. Morgentaler, A. et al. Testosterone therapy in men with untreated prostate cancer. J. Urol. 185, 1256–1260 (2011).

    Article  CAS  PubMed  Google Scholar 

  142. Cui, Y., Zong, H., Yan, H. & Zhang, Y. The effect of testosterone replacement therapy on prostate cancer: a systematic review and meta-analysis. Prostate Cancer Prostatic Dis. 17, 132–143 (2014).

    Article  CAS  PubMed  Google Scholar 

  143. Morris, M. J., Huang, D. & Kelly, W. K. Phase 1 trial of high-dose exogenous testosterone in patients with castration-resistant metastatic prostate cancer. Eur. Urol. 56, 237–244 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Szmulewitz, R., Mohile, S. & Posadas, E. A randomized phase 1 study of testosterone replacement for patients with low-risk castration-resistant prostate cancer. Eur. Urol. 56, 97–103 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Schweizer, M. T. et al. Effect of bipolar androgen therapy for asymptomatic men with castration-resistant prostate cancer: results from a pilot clinical study. Sci. Transl. Med. 7, 269ra2 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  146. Umekita, Y., Hiipakka, R. A., Kokontis, J. M. & Liao, S. Human prostate tumor growth in athymic mice: inhibition by androgens and stimulation by finasteride. Proc. Natl Acad. Sci. USA 93, 11802–11807 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Schweizer, M. T. et al. Bipolar androgen therapy for men with androgen ablation naive prostate cancer: results from the phase II BATMAN study. Prostate 76, 1218–1226 (2016).

    Article  CAS  PubMed  Google Scholar 

  148. Markowski, M. C. et al. A multicohort open-label phase II trial of bipolar androgen therapy in men with metastatic castration-resistant prostate cancer (RESTORE): a comparison of post-abiraterone versus post-enzalutamide cohorts. Eur. Urol. 79, 692–699 (2021).

    Article  CAS  PubMed  Google Scholar 

  149. Sena, L. A. et al. Bipolar androgen therapy sensitizes castration-resistant prostate cancer to subsequent androgen receptor ablative therapy. Eur. J. Cancer 144, 302–309 (2021).

    Article  CAS  PubMed  Google Scholar 

  150. Teply, B. A. et al. Bipolar androgen therapy in men with metastatic castration-resistant prostate cancer after progression on enzalutamide: an open-label, phase 2, multicohort study. Lancet Oncol. 19, 76–86 (2018).

    Article  CAS  PubMed  Google Scholar 

  151. Denmeade, S. R. et al. TRANSFORMER: a randomized phase II study comparing bipolar androgen therapy versus enzalutamide in asymptomatic men with castration-resistant metastatic prostate cancer. J. Clin. Oncol. 39, 1371–1382 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Markowski, M. C. et al. COMBAT-CRPC: concurrent administration of bipolar androgen therapy (BAT) and nivolumab in men with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. https://doi.org/10.1200/JCO.2021.39.15_suppl.5014 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  153. Schweizer, M. et al. 592P Bipolar androgen therapy (BAT) plus olaparib in men with metastatic castration-resistant prostate cancer (mCRPC). Ann. Oncol. 32, S639–S640 (2021).

    Article  Google Scholar 

  154. Manni, A. et al. Androgen depletion and repletion as a means of potentiating the effect of cytotoxic chemotherapy in advanced prostate cancer. J. Steroid Biochem. 27, 551–556 (1987).

    Article  CAS  PubMed  Google Scholar 

  155. Johnson, D. & Haynie, T. Phosphorus-32 for intractable pain in carcinoma of prostate: analysis of androgen priming, parathormone rebound, and combination therapy. Urology 9, 137–139 (1977).

    Article  CAS  PubMed  Google Scholar 

  156. Schwartz, L. H. et al. RECIST 1.1-Update and clarification: From the RECIST committee. Eur. J. Cancer 62, 132–137 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  157. Scher, H. I. et al. Trial design and objectives for castration-resistant prostate cancer: updated recommendations from the Prostate Cancer Clinical Trials Working Group 3. J. Clin. Oncol. 34, 1402–1418 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  158. Sena, L. A. et al. Prostate cancer androgen receptor activity dictates efficacy of bipolar androgen therapy through MYC. J. Clin. Invest. https://doi.org/10.1172/JCI162396 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  159. ClinicalTrials.gov. US National Library of Medicine. https://ClinicalTrials.gov/show/NCT02090114 (2022).

  160. ClinicalTrials.gov. US National Library of Medicine. https://ClinicalTrials.gov/show/NCT03522064 (2021).

  161. Abida, W., Cyrta, J. & Heller, G. Genomic correlates of clinical outcome in advanced prostate cancer. Proc. Natl Acad. Sci. USA 116, 11428–11436 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. ClinicalTrials.gov. US National Library of Medicine. https://ClinicalTrials.gov/show/NCT04363164 (2022).

  163. Sena, L. A., Denmeade, S. R. & Antonarakis, E. S. Targeting the spectrum of immune checkpoints in prostate cancer. Expert. Rev. Clin. Pharmacol. 14, 1253–1266 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Hussain, M., Mateo, J. & Fizazi, K. Survival with olaparib in metastatic castration-resistant prostate cancer. N. Engl. J. Med. 383, 2345–2357 (2020).

    Article  CAS  PubMed  Google Scholar 

  165. Nyquist, M. D. et al. Selective androgen receptor modulators activate the canonical prostate cancer androgen receptor program and repress cancer growth. J. Clin. Invest. https://doi.org/10.1172/JCI146777 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  166. D’Andrea, A. D. Mechanisms of PARP inhibitor sensitivity and resistance. DNA Repair. 71, 172–176 (2018).

    Article  PubMed  Google Scholar 

  167. Bouman, A., Heineman, M. J. & Faas, M. M. Sex hormones and the immune response in humans. Hum. Reprod. Update 11, 411–423 (2005).

    Article  CAS  PubMed  Google Scholar 

  168. Isaacs, J. T. Resolving the Coffey Paradox: what does the androgen receptor do in normal vs. malignant prostate epithelial cells? Am. J. Clin. Exp. Urol. 6, 55–61 (2018).

    PubMed  PubMed Central  Google Scholar 

  169. Notelovitz, M. Androgen effects on bone and muscle. Fertil. Steril. 77 (Suppl. 4), S34–S41 (2002).

    Article  PubMed  Google Scholar 

  170. Lu, S., Tsai, S. Y. & Tsai, M.-J. Regulation of androgen-dependent prostatic cancer cell growth: androgen regulation of CDK2, CDK4, and CKI p16 genes. Cancer Res. 57, 4511–4516 (1997).

    CAS  PubMed  Google Scholar 

  171. Berger, M. F. et al. The genomic complexity of primary human prostate cancer. Nature 470, 214–220 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Chuang, K.-H. et al. Neutropenia with impaired host defense against microbial infection in mice lacking androgen receptor. J. Exp. Med. 206, 1181–1199 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Tomlins, S. A. et al. Role of the TMPRSS2-ERG gene fusion in prostate cancer. Neoplasia 10, 177–IN179 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Tomlins, S. A. et al. ETS gene fusions in prostate cancer: from discovery to daily clinical practice. Eur. Urol. 56, 275–286 (2009).

    Article  CAS  PubMed  Google Scholar 

  175. Heemers, H. V. & Tindall, D. J. Unraveling the complexities of androgen receptor signaling in prostate cancer cells. Cancer Cell 15, 245–247 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Sharma, N. L. et al. The androgen receptor induces a distinct transcriptional program in castration-resistant prostate cancer in man. Cancer Cell 23, 35–47 (2013).

    Article  CAS  PubMed  Google Scholar 

  177. Wang, Q. et al. Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell 138, 245–256 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Chuu, C. P. et al. Androgen suppresses proliferation of castration‐resistant LNCaP 104‐R2 prostate cancer cells through androgen receptor, Skp2, and c‐Myc. Cancer Sci. 102, 2022–2028 (2011).

    Article  CAS  PubMed  Google Scholar 

  179. Kokontis, J. M., Hay, N. & Liao, S. Progression of LNCaP prostate tumor cells during androgen deprivation: hormone-independent growth, repression of proliferation by androgen, and role for p27Kip1 in androgen-induced cell cycle arrest. Mol. Endocrinol. 12, 941–953 (1998).

    Article  CAS  PubMed  Google Scholar 

  180. Cornforth, A., Davis, J., Khanifar, E., Nastiuk, K. & Krolewski, J. FOXO3a mediates the androgen-dependent regulation of FLIP and contributes to TRAIL-induced apoptosis of LNCaP cells. Oncogene 27, 4422–4433 (2008).

    Article  CAS  PubMed  Google Scholar 

  181. Wang, Y. et al. Regulation of androgen receptor transcriptional activity by rapamycin in prostate cancer cell proliferation and survival. Oncogene 27, 7106–7117 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Liao, X. et al. Androgen stimulates matrix metalloproteinase-2 expression in human prostate cancer. Endocrinology 144, 1656–1663 (2003).

    Article  CAS  PubMed  Google Scholar 

  183. Chuan, Y.-C. et al. Androgen induction of prostate cancer cell invasion is mediated by ezrin. J. Biol. Chem. 281, 29938–29948 (2006).

    Article  CAS  PubMed  Google Scholar 

  184. Hara, T., Miyazaki, H., Lee, A., Tran, C. P. & Reiter, R. E. Androgen receptor and invasion in prostate cancer. Cancer Res. 68, 1128–1135 (2008).

    Article  CAS  PubMed  Google Scholar 

  185. Teh, M.-T. et al. FOXM1 induces a global methylation signature that mimics the cancer epigenome in head and neck squamous cell carcinoma. PLoS One 7, e34329 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Tsouko, E. et al. Regulation of the pentose phosphate pathway by an androgen receptor–mTOR-mediated mechanism and its role in prostate cancer cell growth. Oncogenesis 3, e103–e103 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Choi, S. Y. C. et al. The MCT4 gene: a novel, potential target for therapy of advanced prostate cancer. Clin. Cancer Res. 22, 2721–2733 (2016).

    Article  CAS  PubMed  Google Scholar 

  188. Koundouros, N. & Poulogiannis, G. Reprogramming of fatty acid metabolism in cancer. Br. J. Cancer 122, 4–22 (2020).

    Article  CAS  PubMed  Google Scholar 

  189. Poulose, N., Mills, I. G. & Steele, R. E. The impact of transcription on metabolism in prostate and breast cancers. Endocr. Relat. Cancer 25, R435–R452 (2018).

    Article  CAS  PubMed  Google Scholar 

  190. Ono, M. et al. [14C] fluciclovine (alias anti-[14C] FACBC) uptake and ASCT2 expression in castration-resistant prostate cancer cells. Nucl. Med. Biol. 42, 887–892 (2015).

    Article  CAS  PubMed  Google Scholar 

  191. White, M. A. et al. Glutamine transporters are targets of multiple oncogenic signaling pathways in prostate cancer. Mol. Cancer Res. 15, 1017–1028 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Corbin, J. M. & Ruiz-Echevarría, M. J. One-carbon metabolism in prostate cancer: the role of androgen signaling. Int. J. Mol. Sci. 17, 1208 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  193. Shukla-Dave, A. et al. Ornithine decarboxylase is sufficient for prostate tumorigenesis via androgen receptor signaling. Am. J. Pathol. 186, 3131–3145 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Polkinghorn, W. R. et al. Androgen receptor signaling regulates DNA repair in prostate cancers. Cancer Discov. 3, 1245–1253 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Sandhu, S. et al. Poly (ADP-ribose) polymerase (PARP) inhibitors for the treatment of advanced germline BRCA2 mutant prostate cancer. Ann. Oncol. 24, 1416–1418 (2013).

    Article  CAS  PubMed  Google Scholar 

  196. Goodwin, J. F. et al. A hormone–DNA repair circuit governs the response to genotoxic insult. Cancer Discov. 3, 1254–1271 (2013).

    Article  CAS  PubMed  Google Scholar 

  197. Guo, Z. et al. Regulation of androgen receptor activity by tyrosine phosphorylation. Cancer Cell 10, 309–319 (2006).

    Article  CAS  PubMed  Google Scholar 

  198. Liu, Y. et al. Dasatinib inhibits site-specific tyrosine phosphorylation of androgen receptor by Ack1 and Src kinases. Oncogene 29, 3208–3216 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Mellinghoff, I. K. et al. HER2/neu kinase-dependent modulation of androgen receptor function through effects on DNA binding and stability. Cancer Cell 6, 517–527 (2004).

    Article  CAS  PubMed  Google Scholar 

  200. Seaton, A. et al. Interleukin-8 signaling promotes androgen-independent proliferation of prostate cancer cells via induction of androgen receptor expression and activation. Carcinogenesis 29, 1148–1156 (2008).

    Article  CAS  PubMed  Google Scholar 

  201. Fan, W. et al. Insulin-like growth factor 1/insulin signaling activates androgen signaling through direct interactions of Foxo1 with androgen receptor. J. Biol. Chem. 282, 7329–7338 (2007).

    Article  CAS  PubMed  Google Scholar 

  202. Migliaccio, A. et al. Steroid-induced androgen receptor–oestradiol receptor β–Src complex triggers prostate cancer cell proliferation. EMBO J. 19, 5406–5417 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Oliver, V. L., Poulios, K., Ventura, S. & Haynes, J. M. A novel androgen signalling pathway uses dihydrotestosterone, but not testosterone, to activate the EGF receptor signalling cascade in prostate stromal cells. Br. J. Pharmacol. 170, 592–601 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Sun, Y. H., Gao, X., Tang, Y. J., Xu, C. L. & Wang, L. H. Androgens induce increases in intracellular calcium via a G protein-coupled receptor in LNCaP prostate cancer cells. J. Androl. 27, 671–678 (2006).

    Article  CAS  PubMed  Google Scholar 

  205. ClinicalTrials.gov. US National Library of Medicine. https://ClinicalTrials.gov/show/NCT01084759 (2016).

  206. ClinicalTrials.gov. US National Library of Medicine. https://ClinicalTrials.gov/show/NCT01750398 (2016).

  207. ClinicalTrials.gov. US National Library of Medicine. https://ClinicalTrials.gov/show/NCT02286921 (2020).

  208. ClinicalTrials.gov. US National Library of Medicine. https://ClinicalTrials.gov/show/NCT03554317 (2022).

  209. ClinicalTrials.gov. US National Library of Medicine. https://ClinicalTrials.gov/show/NCT03516812 (2022).

Download references

Acknowledgements

S.K. is partly supported by the W81XWH1910724, 1R01CA243184 and PCF Challenge awards. R.K. is supported by the W81XWH2210118 and PCF Young Investigator Award 21YOUN22. L.A.S. is supported by W81XWH2010079 and Johns Hopkins University Clinician-Scientist Award.

Author information

Authors and Affiliations

Authors

Contributions

R.K., L.A.S. and S.K. researched data for the article. All authors contributed substantially to discussion of the content. R.K., L.A.S. and S.K. wrote the article. All authors reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Sushant Kachhap.

Ethics declarations

Competing interests

All the authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Urology thanks Stephen Plymate, Alessandro Tafuri and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kumar, R., Sena, L.A., Denmeade, S.R. et al. The testosterone paradox of advanced prostate cancer: mechanistic insights and clinical implications. Nat Rev Urol 20, 265–278 (2023). https://doi.org/10.1038/s41585-022-00686-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41585-022-00686-y

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer